1
|
Thylur Puttalingaiah R, Dean MJ, Zheng L, Philbrook P, Wyczechowska D, Kayes T, Del Valle L, Danos D, Sanchez-Pino MD. Excess Potassium Promotes Autophagy to Maintain the Immunosuppressive Capacity of Myeloid-Derived Suppressor Cells Independent of Arginase 1. Cells 2024; 13:1736. [PMID: 39451254 PMCID: PMC11505641 DOI: 10.3390/cells13201736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/10/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024] Open
Abstract
Potassium ions (K+) are critical electrolytes that regulate multiple functions in immune cells. Recent studies have shown that the elevated concentration of extracellular potassium in the tumor interstitial fluid limits T cell effector function and suppresses the anti-tumor capacity of tumor-associated macrophages (TAMs). The effect of excess potassium on the biology of myeloid-derived suppressor cells (MDSCs), another important immune cell component of the tumor microenvironment (TME), is unknown. Here, we present data showing that increased concentrations of potassium chloride (KCl), as the source of K+ ions, facilitate autophagy by increasing the expression of the autophagosome marker LC3β. Simultaneously, excess potassium ions significantly decrease the expression of arginase I (Arg I) and inducible nitric oxide synthase (iNOS) without reducing the ability of MDSCs to suppress T cell proliferation. Further investigation reveals that excess K+ ions decrease the expression of the transcription factor C/EBP-β and alter the expression of phosphorylated kinases. While excess K+ ions downregulated the expression levels of phospho-AMPKα (pAMPKα), it increased the levels of pAKT and pERK. Additionally, potassium increased mitochondrial respiration as measured by the oxygen consumption rate (OCR). Interestingly, all these alterations induced by K+ ions were abolished by the autophagy inhibitor 3-methyladenine (3-MA). Our results suggest that hyperosmotic stress caused by excess K+ ions regulate the mitochondrial respiration and signaling pathways in MDSCs to trigger the process of autophagy to support MDSCs' immunosuppressive function by mechanisms independent of Arg I and iNOS. Overall, our in vitro and ex vivo findings offer valuable insights into the adaptations of MDSCs within the K+ ion-rich TME, which has important implications for MDSCs-targeted therapies.
Collapse
Affiliation(s)
- Ramesh Thylur Puttalingaiah
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (M.J.D.); (L.Z.); (P.P.); (D.W.); (T.K.); (L.D.V.); (D.D.)
| | - Matthew J. Dean
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (M.J.D.); (L.Z.); (P.P.); (D.W.); (T.K.); (L.D.V.); (D.D.)
| | - Liqin Zheng
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (M.J.D.); (L.Z.); (P.P.); (D.W.); (T.K.); (L.D.V.); (D.D.)
| | - Phaethon Philbrook
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (M.J.D.); (L.Z.); (P.P.); (D.W.); (T.K.); (L.D.V.); (D.D.)
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Dorota Wyczechowska
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (M.J.D.); (L.Z.); (P.P.); (D.W.); (T.K.); (L.D.V.); (D.D.)
| | - Timothy Kayes
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (M.J.D.); (L.Z.); (P.P.); (D.W.); (T.K.); (L.D.V.); (D.D.)
| | - Luis Del Valle
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (M.J.D.); (L.Z.); (P.P.); (D.W.); (T.K.); (L.D.V.); (D.D.)
| | - Denise Danos
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (M.J.D.); (L.Z.); (P.P.); (D.W.); (T.K.); (L.D.V.); (D.D.)
- Department of Interdisciplinary Oncology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
- School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Maria Dulfary Sanchez-Pino
- Stanley S. Scott Cancer Center, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (M.J.D.); (L.Z.); (P.P.); (D.W.); (T.K.); (L.D.V.); (D.D.)
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
- Department of Interdisciplinary Oncology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|
2
|
Swift NA, Yang Q, Jester HM, Zhou X, Manuel A, Kemp BE, Steinberg GR, Ma T. Suppression of neuronal AMPKβ2 isoform impairs recognition memory and synaptic plasticity. Neurobiol Dis 2024; 201:106664. [PMID: 39278510 PMCID: PMC11539201 DOI: 10.1016/j.nbd.2024.106664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/03/2024] [Accepted: 09/09/2024] [Indexed: 09/18/2024] Open
Abstract
AMP-activated protein kinase (AMPK) is an αβγ heterotrimer protein kinase that functions as a molecular sensor to maintain energy homeostasis. Accumulating evidence suggests a role of AMPK signaling in the regulation of synaptic plasticity and cognitive function; however, isoform-specific roles of AMPK in the central nervous system (CNS) remain elusive. Regulation of the AMPK activities has focused on the manipulation of the α or γ subunit. Meanwhile, accumulating evidence indicates that the β subunit is critical for sensing nutrients such as fatty acids and glycogen to control AMPK activity. Here, we generated transgenic mice with conditional suppression of either AMPKβ1 or β2 in neurons and characterized potential isoform-specific roles of AMPKβ in cognitive function and underlying mechanisms. We found that AMPKβ2 (but not β1) suppression resulted in impaired recognition memory, reduced hippocampal synaptic plasticity, and altered structure of hippocampal postsynaptic densities and dendritic spines. Our study implicates a role for the AMPKβ2 isoform in the regulation of synaptic and cognitive function.
Collapse
Affiliation(s)
- Nathaniel A Swift
- Department of Internal Medicine, Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA
| | - Qian Yang
- Department of Internal Medicine, Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA
| | - Hannah M Jester
- Department of Internal Medicine, Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA
| | - Xueyan Zhou
- Department of Internal Medicine, Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA
| | - Adam Manuel
- Department of Internal Medicine, Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA
| | - Bruce E Kemp
- St. Vincent's Institute of Medical Research and Department of Medicine, University of Melbourne, Parkville, VIC 3010, Australia; Mary McKillop Institute for Health Research, Australian Catholic University, Melbourne 3000, VIC, Australia
| | - Gregory R Steinberg
- Centre for Metabolism, Obesity and Diabetes Research, Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Tao Ma
- Department of Internal Medicine, Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA; Department of Translational Neuroscience, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA.
| |
Collapse
|
3
|
Smiles WJ, Ovens AJ, Oakhill JS, Kofler B. The metabolic sensor AMPK: Twelve enzymes in one. Mol Metab 2024; 90:102042. [PMID: 39362600 DOI: 10.1016/j.molmet.2024.102042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 09/12/2024] [Accepted: 09/27/2024] [Indexed: 10/05/2024] Open
Abstract
BACKGROUND AMP-activated protein kinase (AMPK) is an evolutionarily conserved regulator of energy metabolism. AMPK is sensitive to acute perturbations to cellular energy status and leverages fundamental bioenergetic pathways to maintain cellular homeostasis. AMPK is a heterotrimer comprised of αβγ-subunits that in humans are encoded by seven individual genes (isoforms α1, α2, β1, β2, γ1, γ2 and γ3), permitting formation of at least 12 different complexes with personalised biochemical fingerprints and tissue expression patterns. While the canonical activation mechanisms of AMPK are well-defined, delineation of subtle, as well as substantial, differences in the regulation of heterogenous AMPK complexes remain poorly defined. SCOPE OF REVIEW Here, taking advantage of multidisciplinary findings, we dissect the many aspects of isoform-specific AMPK function and links to health and disease. These include, but are not limited to, allosteric activation by adenine nucleotides and small molecules, co-translational myristoylation and post-translational modifications (particularly phosphorylation), governance of subcellular localisation, and control of transcriptional networks. Finally, we delve into current debate over whether AMPK can form novel protein complexes (e.g., dimers lacking the α-subunit), altogether highlighting opportunities for future and impactful research. MAJOR CONCLUSIONS Baseline activity of α1-AMPK is higher than its α2 counterpart and is more sensitive to synergistic allosteric activation by metabolites and small molecules. α2 complexes however, show a greater response to energy stress (i.e., AMP production) and appear to be better substrates for LKB1 and mTORC1 upstream. These differences may explain to some extent why in certain cancers α1 is a tumour promoter and α2 a suppressor. β1-AMPK activity is toggled by a 'myristoyl-switch' mechanism that likely precedes a series of signalling events culminating in phosphorylation by ULK1 and sensitisation to small molecules or endogenous ligands like fatty acids. β2-AMPK, not entirely beholden to this myristoyl-switch, has a greater propensity to infiltrate the nucleus, which we suspect contributes to its oncogenicity in some cancers. Last, the unique N-terminal extensions of the γ2 and γ3 isoforms are major regulatory domains of AMPK. mTORC1 may directly phosphorylate this region in γ2, although whether this is inhibitory, especially in disease states, is unclear. Conversely, γ3 complexes might be preferentially regulated by mTORC1 in response to physical exercise.
Collapse
Affiliation(s)
- William J Smiles
- Research Program for Receptor Biochemistry and Tumour Metabolism, Department of Paediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria; Metabolic Signalling Laboratory, St. Vincent's Institute of Medical Research, Fitzroy, Melbourne, Australia.
| | - Ashley J Ovens
- Protein Engineering in Immunity & Metabolism, St. Vincent's Institute of Medical Research, Fitzroy, Melbourne, Australia
| | - Jonathan S Oakhill
- Metabolic Signalling Laboratory, St. Vincent's Institute of Medical Research, Fitzroy, Melbourne, Australia; Department of Medicine, University of Melbourne, Parkville, Australia
| | - Barbara Kofler
- Research Program for Receptor Biochemistry and Tumour Metabolism, Department of Paediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
4
|
Wang X, Yang Q, Zhou X, Keene CD, Ryazanov AG, Ma T. Suppression of eEF2 phosphorylation alleviates synaptic failure and cognitive deficits in mouse models of Down syndrome. Alzheimers Dement 2024; 20:5357-5374. [PMID: 38934363 PMCID: PMC11350057 DOI: 10.1002/alz.13916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/11/2024] [Accepted: 05/01/2024] [Indexed: 06/28/2024]
Abstract
INTRODUCTION Cognitive impairment is a core feature of Down syndrome (DS), and the underlying neurobiological mechanisms remain unclear. Translation dysregulation is linked to multiple neurological disorders characterized by cognitive impairments. Phosphorylation of the translational factor eukaryotic elongation factor 2 (eEF2) by its kinase eEF2K results in inhibition of general protein synthesis. METHODS We used genetic and pharmacological methods to suppress eEF2K in two lines of DS mouse models. We further applied multiple approaches to evaluate the effects of eEF2K inhibition on DS pathophysiology. RESULTS We found that eEF2K signaling was overactive in the brain of patients with DS and DS mouse models. Inhibition of eEF2 phosphorylation through suppression of eEF2K in DS model mice improved multiple aspects of DS-associated pathophysiology including de novo protein synthesis deficiency, synaptic morphological defects, long-term synaptic plasticity failure, and cognitive impairments. DISCUSSION Our data suggested that eEF2K signaling dysregulation mediates DS-associated synaptic and cognitive impairments. HIGHLIGHTS Phosphorylation of the translational factor eukaryotic elongation factor 2 (eEF2) is increased in the Down syndrome (DS) brain. Suppression of the eEF2 kinase (eEF2K) alleviates cognitive deficits in DS models. Suppression of eEF2K improves synaptic dysregulation in DS models. Cognitive and synaptic impairments in DS models are rescued by eEF2K inhibitors.
Collapse
Affiliation(s)
- Xin Wang
- Department of Internal MedicineGerontology and Geriatric MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Qian Yang
- Department of Internal MedicineGerontology and Geriatric MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Xueyan Zhou
- Department of Internal MedicineGerontology and Geriatric MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - C. Dirk Keene
- Department of PathologyUniversity of Washington School of MedicineSeattleWashingtonUSA
| | - Alexey G. Ryazanov
- Department of PharmacologyRutgers Robert Wood Johnson Medical SchoolPiscatawayNew JerseyUSA
| | - Tao Ma
- Department of Internal MedicineGerontology and Geriatric MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
- Department of Translational NeuroscienceWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| |
Collapse
|
5
|
Langer HT, Rohm M, Goncalves MD, Sylow L. AMPK as a mediator of tissue preservation: time for a shift in dogma? Nat Rev Endocrinol 2024:10.1038/s41574-024-00992-y. [PMID: 38760482 DOI: 10.1038/s41574-024-00992-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/19/2024] [Indexed: 05/19/2024]
Abstract
Ground-breaking discoveries have established 5'-AMP-activated protein kinase (AMPK) as a central sensor of metabolic stress in cells and tissues. AMPK is activated through cellular starvation, exercise and drugs by either directly or indirectly affecting the intracellular AMP (or ADP) to ATP ratio. In turn, AMPK regulates multiple processes of cell metabolism, such as the maintenance of cellular ATP levels, via the regulation of fatty acid oxidation, glucose uptake, glycolysis, autophagy, mitochondrial biogenesis and degradation, and insulin sensitivity. Moreover, AMPK inhibits anabolic processes, such as lipogenesis and protein synthesis. These findings support the notion that AMPK is a crucial regulator of cell catabolism. However, studies have revealed that AMPK's role in cell homeostasis might not be as unidirectional as originally thought. This Review explores emerging evidence for AMPK as a promoter of cell survival and an enhancer of anabolic capacity in skeletal muscle and adipose tissue during catabolic crises. We discuss AMPK-activating interventions for tissue preservation during tissue wasting in cancer-associated cachexia and explore the clinical potential of AMPK activation in wasting conditions. Overall, we provide arguments that call for a shift in the current dogma of AMPK as a mere regulator of cell catabolism, concluding that AMPK has an unexpected role in tissue preservation.
Collapse
Affiliation(s)
- Henning Tim Langer
- Division of Endocrinology, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riβ, Germany.
| | - Maria Rohm
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Marcus DaSilva Goncalves
- Division of Endocrinology, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Lykke Sylow
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
6
|
Lv T, Lou Y, Yan Q, Nie L, Cheng Z, Zhou X. Phosphorylation: new star of pathogenesis and treatment in steatotic liver disease. Lipids Health Dis 2024; 23:50. [PMID: 38368351 PMCID: PMC10873984 DOI: 10.1186/s12944-024-02037-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 01/31/2024] [Indexed: 02/19/2024] Open
Abstract
Steatotic liver disease poses a serious threat to human health and has emerged as one of the most significant burdens of chronic liver disease worldwide. Currently, the research mechanism is not clear, and there is no specific targeted drug for direct treatment. Phosphorylation is widely regarded as the most common type of protein modification, closely linked to steatotic liver disease in previous studies. However, there is no systematic review to clarify the relationship and investigate from the perspective of phosphorylation. Phosphorylation has been found to mainly regulate molecule stability, affect localization, transform molecular function, and cooperate with other protein modifications. Among them, adenosine 5'-monophosphate-activated protein kinase (AMPK), serine/threonine kinase (AKT), and nuclear factor kappa-B (NF-kB) are considered the core mechanisms in steatotic liver disease. As to treatment, lifestyle changes, prescription drugs, and herbal ingredients can alleviate symptoms by influencing phosphorylation. It demonstrates the significant role of phosphorylation as a mechanism occurrence and a therapeutic target in steatotic liver disease, which could be a new star for future exploration.
Collapse
Affiliation(s)
- Tiansu Lv
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yan Lou
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Qianhua Yan
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lijuan Nie
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhe Cheng
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiqiao Zhou
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
7
|
Wang S, Chen Q, Wang F. Differences of Pine Wood Nematode ( Bursaphelenchus xylophilus) Developmental Stages under High-Osmotic-Pressure Stress. BIOLOGY 2024; 13:123. [PMID: 38392341 PMCID: PMC10886877 DOI: 10.3390/biology13020123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/06/2024] [Accepted: 02/11/2024] [Indexed: 02/24/2024]
Abstract
Under ion imbalance, water deficiency, and salt stress, the osmotic pressure of the tree sap increases, and pine wood nematodes (Bursaphelenchus xylophilus, PWN) parasitizing in the trees may be subjected to high-osmotic-pressure stress. KCl, L-malic acid, sucrose, and glycerol solutions were used as osmolytes to explore the highest osmotic concentration that PWN can tolerate. Survival analysis showed that when the treatment concentration exceeded 90%, only a few nematodes in the glycerol group survived under 6 h treatment, and most of the survivors were third-stage dispersal juveniles (DJ3). Further examination revealed that under different concentrations of glycerol-induced high osmotic pressure, the survival rate and body length change rate were the highest in the DJ3 and the lowest in the second-stage propagative juveniles. In order to explore the molecular mechanism of resistance of DJ3 to high osmotic stress, transcriptome sequencing was performed at each developmental stage of PWN and differentially expressed genes that were up-regulated or down-regulated only in DJ3 were screened. The expression of genes related to CoA in DJ3, a key enzyme in metabolism, was significantly higher than the other developmental stages. In addition, the expression of the anti-reversal signal pathway-related gene AKT-1 in DJ3 was significantly lower than in the other development stages. Therefore, the specific expression of genes in DJ3 under high osmotic pressure may help them rapidly produce and accumulate energy-related compounds and activate the adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) pathway to respond to damage caused by high-osmotic-pressure stress in time, thus promoting survival.
Collapse
Affiliation(s)
- Shuting Wang
- Key Laboratory of Alien Forest Pests Monitoring and Control-Heilongjiang Province, School of Forestry, Northeast Forestry University, Harbin 150040, China
| | - Qiaoli Chen
- Key Laboratory of Alien Forest Pests Monitoring and Control-Heilongjiang Province, School of Forestry, Northeast Forestry University, Harbin 150040, China
- Key Laboratory of Sustainable Forest Ecosystem Management-Ministry of Education, Northeast Forestry University, Harbin 150040, China
| | - Feng Wang
- Key Laboratory of Alien Forest Pests Monitoring and Control-Heilongjiang Province, School of Forestry, Northeast Forestry University, Harbin 150040, China
- Key Laboratory of Sustainable Forest Ecosystem Management-Ministry of Education, Northeast Forestry University, Harbin 150040, China
- State Key Laboratory of Tree Genetics and Breeding, Northeast Forestry University, Harbin 150040, China
| |
Collapse
|
8
|
Maestri A, Garagnani P, Pedrelli M, Hagberg CE, Parini P, Ehrenborg E. Lipid droplets, autophagy, and ageing: A cell-specific tale. Ageing Res Rev 2024; 94:102194. [PMID: 38218464 DOI: 10.1016/j.arr.2024.102194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 12/22/2023] [Accepted: 01/08/2024] [Indexed: 01/15/2024]
Abstract
Lipid droplets are the essential organelle for storing lipids in a cell. Within the variety of the human body, different cells store, utilize and release lipids in different ways, depending on their intrinsic function. However, these differences are not well characterized and, especially in the context of ageing, represent a key factor for cardiometabolic diseases. Whole body lipid homeostasis is a central interest in the field of cardiometabolic diseases. In this review we characterize lipid droplets and their utilization via autophagy and describe their diverse fate in three cells types central in cardiometabolic dysfunctions: adipocytes, hepatocytes, and macrophages.
Collapse
Affiliation(s)
- Alice Maestri
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Paolo Garagnani
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy; IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Matteo Pedrelli
- Cardio Metabolic Unit, Department of Laboratory Medicine, and Department of Medicine (Huddinge), Karolinska Institutet, Stockholm, Sweden; Medicine Unit of Endocrinology, Theme Inflammation and Ageing, Karolinska University Hospital, Stockholm, Sweden
| | - Carolina E Hagberg
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Paolo Parini
- Cardio Metabolic Unit, Department of Laboratory Medicine, and Department of Medicine (Huddinge), Karolinska Institutet, Stockholm, Sweden; Medicine Unit of Endocrinology, Theme Inflammation and Ageing, Karolinska University Hospital, Stockholm, Sweden
| | - Ewa Ehrenborg
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
9
|
Pearah A, Ramatchandirin B, Liu T, Wolf RM, Ikeda A, Radovick S, Sesaki H, Wondisford FE, O'Rourke B, He L. Blocking AMPKαS496 phosphorylation improves mitochondrial dynamics and hyperglycemia in aging and obesity. Cell Chem Biol 2023; 30:1585-1600.e6. [PMID: 37890479 PMCID: PMC10841824 DOI: 10.1016/j.chembiol.2023.09.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 08/23/2023] [Accepted: 09/28/2023] [Indexed: 10/29/2023]
Abstract
Impaired mitochondrial dynamics causes aging-related or metabolic diseases. Yet, the molecular mechanism responsible for the impairment of mitochondrial dynamics is still not well understood. Here, we report that elevated blood insulin and/or glucagon levels downregulate mitochondrial fission through directly phosphorylating AMPKα at S496 by AKT or PKA, resulting in the impairment of AMPK-MFF-DRP1 signaling and mitochondrial dynamics and activity. Since there are significantly increased AMPKα1 phosphorylation at S496 in the liver of elderly mice, obese mice, and obese patients, we, therefore, designed AMPK-specific targeting peptides (Pa496m and Pa496h) to block AMPKα1S496 phosphorylation and found that these targeting peptides can increase AMPK kinase activity, augment mitochondrial fission and oxidation, and reduce ROS, leading to the rejuvenation of mitochondria. Furthermore, these AMPK targeting peptides robustly suppress liver glucose production in obese mice. Our data suggest these targeting peptides are promising therapeutic agents for improving mitochondrial dynamics and activity and alleviating hyperglycemia in elderly and obese patients.
Collapse
Affiliation(s)
- Alexia Pearah
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | | | - Ting Liu
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Risa M Wolf
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Arisa Ikeda
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Sally Radovick
- Departments of Pediatrics and Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Hiromi Sesaki
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Fredric E Wondisford
- Departments of Pediatrics and Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Brian O'Rourke
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ling He
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Departments of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
10
|
Ross FA, Hawley SA, Russell FM, Goodman N, Hardie DG. Frequent loss-of-function mutations in the AMPK-α2 catalytic subunit suggest a tumour suppressor role in human skin cancers. Biochem J 2023; 480:1951-1968. [PMID: 37962491 PMCID: PMC10754287 DOI: 10.1042/bcj20230380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/07/2023] [Accepted: 11/13/2023] [Indexed: 11/15/2023]
Abstract
The AMP-activated protein kinase (AMPK) is a sensor of cellular energy status activated by increases in AMP or ADP relative to ATP. Once activated, it phosphorylates targets that promote ATP-generating catabolic pathways or inhibit ATP-consuming anabolic pathways, helping to restore cellular energy balance. Analysis of human cancer genome studies reveals that the PRKAA2 gene (encoding the α2 isoform of the catalytic subunit) is often subject to mis-sense mutations in cancer, particularly in melanoma and non-melanoma skin cancers, where up to 70 mis-sense mutations have been documented, often accompanied by loss of the tumour suppressor NF1. Recently it has been reported that knockout of PRKAA2 in NF1-deficient melanoma cells promoted anchorage-independent growth in vitro, as well as growth as xenografts in immunodeficient mice in vivo, suggesting that AMPK-α2 can act as a tumour suppressor in that context. However, very few of the mis-sense mutations in PRKAA2 that occur in human skin cancer and melanoma have been tested to see whether they cause loss-of-function. We have addressed this by making most of the reported mutations and testing their activity when expressed in AMPK knockout cells. Of 55 different mis-sense mutations (representing 75 cases), 9 (12%) appeared to cause a total loss of activity, 18 (24%) a partial loss, 11 (15%) an increase in phenformin-stimulated kinase activity, while just 37 (49%) had no clear effect on kinase activity. This supports the idea that AMPK-α2 acts as a tumour suppressor in the context of human skin cancer.
Collapse
Affiliation(s)
- Fiona A. Ross
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Scotland, U.K
| | - Simon A. Hawley
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Scotland, U.K
| | - Fiona M. Russell
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Scotland, U.K
| | - Nicola Goodman
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Scotland, U.K
| | - D. Grahame Hardie
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Scotland, U.K
| |
Collapse
|
11
|
Song Y, Yang H, Kim J, Lee Y, Kim SH, Do IG, Park CY. Gemigliptin, a DPP4 inhibitor, ameliorates nonalcoholic steatohepatitis through AMP-activated protein kinase-independent and ULK1-mediated autophagy. Mol Metab 2023; 78:101806. [PMID: 37739179 PMCID: PMC10542016 DOI: 10.1016/j.molmet.2023.101806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/16/2023] [Accepted: 09/11/2023] [Indexed: 09/24/2023] Open
Abstract
OBJECTIVE Abnormal autophagic function and activated inflammasomes are typical features in the liver of patients with non-alcoholic steatohepatitis (NASH). Here, we explored whether gemigliptin, a dipeptidyl peptidase 4 (DPP4) inhibitor for treatment of type 2 diabetes, can induce autophagy and regulate inflammasome activation as a potential NASH treatment independent of its anti-diabetic effect. METHODS Expression analysis was performed using human liver samples obtained from 18 subjects who underwent hepatectomy. We explored the function and mechanism of gemigliptin using a methionine- and choline-deficient diet (MCD)-induced NASH mouse model and HepG2 cells cultured in MCD-mimicking medium. RESULTS Autophagy was suppressed by marked decreases in the expression of ULK1 and LC3II/LC3I ratio in human NAFLD/NASH patients, a NASH mouse model, and HepG2 cells cultured with MCD-mimicking media. Surprisingly, we found that the expression of p-AMPK decreased in liver tissues from patients with steatosis but was restored in NASH patients. The expression of p-AMPK in the NASH mouse model was similar to that of the control group. Hence, these results indicate that autophagy was reduced in NASH via an AMPK-independent pathway. However, gemigliptin treatment attenuated lipid accumulation, inflammation, and fibrosis in the liver of MCD diet-fed mice with restoration of ULK1 expression and autophagy induction. In vitro, gemigliptin alleviated inflammasome activation through induction of ULK1-dependent autophagy. Furthermore, gemigliptin treatment upregulated ULK1 expression and activated AMPK even after siRNA-mediated knockdown of AMPKα1/2 and ULK1, respectively. CONCLUSIONS Collectively, these results suggest that gemigliptin ameliorated NASH via AMPK-independent, ULK1-mediated effects on autophagy.
Collapse
Affiliation(s)
- Youngmi Song
- Medical Research Institute, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hyekyung Yang
- Medical Research Institute, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Juhee Kim
- Medical Research Institute, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Yoonjin Lee
- Medical Research Institute, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Sung-Ho Kim
- LG Chem Life Sciences, Gangseo-gu, Seoul, South Korea
| | - In-Gu Do
- Department of Pathology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Cheol-Young Park
- Medical Research Institute, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, South Korea; Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| |
Collapse
|
12
|
Göransson O, Kopietz F, Rider MH. Metabolic control by AMPK in white adipose tissue. Trends Endocrinol Metab 2023; 34:704-717. [PMID: 37673765 DOI: 10.1016/j.tem.2023.08.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/10/2023] [Accepted: 08/11/2023] [Indexed: 09/08/2023]
Abstract
White adipose tissue (WAT) plays an important role in the integration of whole-body metabolism by storing fat and mobilizing triacylglycerol when needed. The released free fatty acids can then be oxidized by other tissues to provide ATP. AMP-activated protein kinase (AMPK) is a key regulator of metabolic pathways, and can be targeted by a new generation of direct, small-molecule activators. AMPK activation in WAT inhibits insulin-stimulated lipogenesis and in some situations also inhibits insulin-stimulated glucose uptake, but AMPK-induced inhibition of β-adrenergic agonist-stimulated lipolysis might need to be re-evaluated in vivo. The lack of dramatic effects of AMPK activation on basal metabolism in WAT could be advantageous when treating type 2 diabetes with pharmacological pan-AMPK activators.
Collapse
Affiliation(s)
- Olga Göransson
- Lund University, Department of Experimental Medical Science, BMC, 221 84 Lund, Sweden.
| | - Franziska Kopietz
- Lund University, Department of Experimental Medical Science, BMC, 221 84 Lund, Sweden
| | - Mark H Rider
- Université catholique de Louvain (UCLouvain) and de Duve Institute, Avenue Hippocrate 75, 1200 Brussels, Belgium
| |
Collapse
|
13
|
Wu Q, Meng W, Zhu B, Chen X, Fu J, Zhao C, Liu G, Luo X, Lv Y, Zhao W, Wang F, Hu S, Zhang S. VEGFC ameliorates salt-sensitive hypertension and hypertensive nephropathy by inhibiting NLRP3 inflammasome via activating VEGFR3-AMPK dependent autophagy pathway. Cell Mol Life Sci 2023; 80:327. [PMID: 37837447 PMCID: PMC11072217 DOI: 10.1007/s00018-023-04978-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/29/2023] [Accepted: 09/23/2023] [Indexed: 10/16/2023]
Abstract
Salt-sensitivity hypertension (SSHTN) is an independent predictor for cardiovascular mortality. VEGFC has been reported to be a protective role in SSHTN and hypertensive kidney injury. However, the underlying mechanisms remain largely unclear. The current study aimed to explore the protective effects and mechanisms of VEGFC against SSHTN and hypertensive nephropathy. Here, we reported that VEGFC attenuated high blood pressure as well as protected against renal inflammation and fibrosis in SSHTN mice. Moreover, VEGFC suppressed the activation of renal NLRP3 inflammasome in SSHTN mice. In vitro, we found VEGFC inhibited NLRP3 inflammasome activation, meanwhile, upregulated autophagy in high-salt-induced macrophages, while these effects were reversed by an autophagy inhibitor 3MA. Furthermore, in vivo, 3MA pretreatment weakened the protective effects of VEGFC on SSHTN and hypertensive nephropathy. Mechanistically, VEGF receptor 3 (VEGFR3) kinase domain activated AMPK by promoting the phosphorylation at Thr183 via binding to AMPK, thus enhancing autophagy activity in the context of high-salt-induced macrophages. These findings indicated that VEGFC inhibited NLRP3 inflammasome activation by promoting VEGFR3-AMPK-dependent autophagy pathway in high-salt-induced macrophages, which provided a mechanistic basis for the therapeutic target in SSHTN and hypertensive kidney injury.
Collapse
Affiliation(s)
- Qiuwen Wu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin Medical University, Harbin, 150001, China
| | - Wei Meng
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin Medical University, Harbin, 150001, China
| | - Bin Zhu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin Medical University, Harbin, 150001, China
| | - Xi Chen
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
| | - Jiaxin Fu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
| | - Chunyu Zhao
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
| | - Gang Liu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Xing Luo
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
| | - Ying Lv
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
| | - Wenqi Zhao
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | - Fan Wang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China
| | - Sining Hu
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China.
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin Medical University, Harbin, 150001, China.
| | - Shuo Zhang
- Department of Cardiology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, 150001, China.
- The Key Laboratory of Myocardial Ischemia, Chinese Ministry of Education, Harbin, 150001, China.
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
14
|
Townsend LK, Steinberg GR. AMPK and the Endocrine Control of Metabolism. Endocr Rev 2023; 44:910-933. [PMID: 37115289 DOI: 10.1210/endrev/bnad012] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/10/2023] [Accepted: 04/24/2023] [Indexed: 04/29/2023]
Abstract
Complex multicellular organisms require a coordinated response from multiple tissues to maintain whole-body homeostasis in the face of energetic stressors such as fasting, cold, and exercise. It is also essential that energy is stored efficiently with feeding and the chronic nutrient surplus that occurs with obesity. Mammals have adapted several endocrine signals that regulate metabolism in response to changes in nutrient availability and energy demand. These include hormones altered by fasting and refeeding including insulin, glucagon, glucagon-like peptide-1, catecholamines, ghrelin, and fibroblast growth factor 21; adipokines such as leptin and adiponectin; cell stress-induced cytokines like tumor necrosis factor alpha and growth differentiating factor 15, and lastly exerkines such as interleukin-6 and irisin. Over the last 2 decades, it has become apparent that many of these endocrine factors control metabolism by regulating the activity of the AMPK (adenosine monophosphate-activated protein kinase). AMPK is a master regulator of nutrient homeostasis, phosphorylating over 100 distinct substrates that are critical for controlling autophagy, carbohydrate, fatty acid, cholesterol, and protein metabolism. In this review, we discuss how AMPK integrates endocrine signals to maintain energy balance in response to diverse homeostatic challenges. We also present some considerations with respect to experimental design which should enhance reproducibility and the fidelity of the conclusions.
Collapse
Affiliation(s)
- Logan K Townsend
- Centre for Metabolism Obesity and Diabetes Research, Hamilton, ON L8S 4L8, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Gregory R Steinberg
- Centre for Metabolism Obesity and Diabetes Research, Hamilton, ON L8S 4L8, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
15
|
Marchal MA, Moose DL, Varzavand A, Jordan NE, Taylor D, Tanas MR, Brown JA, Henry MD, Stipp CS. Abl kinases can function as suppressors of tumor progression and metastasis. Front Oncol 2023; 13:1241056. [PMID: 37746268 PMCID: PMC10514900 DOI: 10.3389/fonc.2023.1241056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/23/2023] [Indexed: 09/26/2023] Open
Abstract
Introduction Abl family kinases function as proto-oncogenes in various leukemias, and pro-tumor functions have been discovered for Abl kinases in many solid tumors as well. However, a growing body of evidence indicates that Abl kinases can function to suppress tumor cell proliferation and motility and tumor growth in vivo in some settings. Methods To investigate the role of Abl kinases in tumor progression, we used RNAi to generate Abl-deficient cells in a model of androgen receptor-indifferent, metastatic prostate cancer. The effect of Abl kinase depletion on tumor progression and metastasis was studied in an in vivo orthotopic model, and tumor cell motility, 3D growth, and signaling was studied in vitro. Results Reduced Abl family kinase expression resulted in a highly aggressive, metastatic phenotype in vivo that was associated with AKT pathway activation, increased growth on 3D collagen matrix, and enhanced cell motility in vitro. Inhibiting AKT pathway signaling abolished the increased 3D growth of Abl-deficient cells, while treatment with the Abl kinase inhibitor, imatinib, promoted 3D growth of multiple additional tumor cell types. Moreover, Abl kinase inhibition also promoted soft-agar colony formation by pre-malignant fibroblasts. Conclusions Collectively, our data reveal that Abl family kinases can function to suppress malignant cell phenotypes in vitro, and tumor progression and metastasis in vivo.
Collapse
Affiliation(s)
- Melissa A Marchal
- Department of Biology, College of Liberal Arts and Sciences, University of Iowa, Iowa City, IA, United States
| | - Devon L Moose
- Department of Molecular Physiology & Biophysics, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Afshin Varzavand
- Department of Biology, College of Liberal Arts and Sciences, University of Iowa, Iowa City, IA, United States
| | - Nicole E Jordan
- Department of Biology, College of Liberal Arts and Sciences, University of Iowa, Iowa City, IA, United States
| | - Destiney Taylor
- Department of Biology, College of Liberal Arts and Sciences, University of Iowa, Iowa City, IA, United States
| | - Munir R Tanas
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - James A Brown
- Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Department of Urology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Michael D Henry
- Department of Molecular Physiology & Biophysics, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Christopher S Stipp
- Department of Biology, College of Liberal Arts and Sciences, University of Iowa, Iowa City, IA, United States
- Holden Comprehensive Cancer Center, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
16
|
Chen F, Chai YH, Zhang F, Liu YQ, Zhang Y, Shi YJ, Zhang JM, Leng YF. Network pharmacology analysis combined with experimental validation to explore the therapeutic mechanism of salidroside on intestine ischemia reperfusion. Biosci Rep 2023; 43:BSR20230539. [PMID: 37530723 PMCID: PMC10462912 DOI: 10.1042/bsr20230539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 06/23/2023] [Accepted: 07/05/2023] [Indexed: 08/03/2023] Open
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Salidroside (SAL), a phenolic natural product present in Rhodiola rosea, are commonly used in the treatment of various ischemic-hypoxic diseases, including intestinal ischemia-reperfusion (IR) injury. However, their efficacy and potential mechanisms in the treatment of intestinal IR injury have not been investigated. OBJECTIVE The objective of the present study is to investigate the pharmacological mechanism of action of SAL on intestinal IR injury using a network pharmacology approach combined with experimental validation. METHODS In the present study, we used the Traditional Chinese Medicine Systematic Pharmacology (TCMSP) database and analysis platform and Comparative Toxicogenomics Database (CTD) to predict possible target genes of SAL, collected relevant target genes of intestinal IR injury from GeneCards and DisGenet websites, and collected summary data to screen common target genes. Then, the protein-protein interaction (PPI) target network was constructed and analyzed by STRING database and Cytoscape 3.8.2 with the above intersecting genes. Then, gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses were performed and the component-target-pathway network was constructed, followed by the use of molecular docking and molecular dynamic simulation to verify the possible binding conformation between SAL and candidate targets to further explore the potential targets of SAL in the treatment of intestinal IR injury. Finally, an in vivo model of mouse superior mesenteric artery ligation was established to assess the anti-intestinal IR injury effect of SAL by assessing histopathological changes in mouse small intestine by HE staining, detecting inflammatory factor expression by ELISA kit, and detecting the expression of key protein targets by Western blotting. RESULTS A total of 166 SAL target genes and 1740 disease-related targets were retrieved, and 88 overlapping proteins were obtained as potential therapeutic targets. The pathway enrichment analysis revealed that the pharmacological effects of SAL on intestinal IR injury were anti-hypoxic, anti-inflammatory and metabolic pathway related, and the molecular docking and molecular dynamic simulation results showed that the core bioactive components had good binding affinity for TXNIP and AMPK, and the immunoblotting results indicated that the expression levels of TXNIP and AMPK in the small intestinal tissues of mice in the drug-treated group compared with the model group were significantly changed. CONCLUSION SAL may target AMPK and TXNIP domains to act as a therapeutic agent for intestinal IR. These findings comprehensively reveal the potential therapeutic targets for SAL against intestinal IR and provide theoretical basis for the clinical application of SAL in the treatment of intestinal IR.
Collapse
Affiliation(s)
- Feng Chen
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, GanSu Province, China
- Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, 730000, GanSu Province, China
| | - Yi-hong Chai
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, GanSu Province, China
| | - Fa Zhang
- Department of Urology, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Yong-qiang Liu
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, GanSu Province, China
- Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, 730000, GanSu Province, China
| | - Yan Zhang
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, GanSu Province, China
- Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, 730000, GanSu Province, China
| | - Ya-jing Shi
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, GanSu Province, China
| | - Jian-ming Zhang
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, GanSu Province, China
| | - Yu-fang Leng
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, GanSu Province, China
- Department of Anesthesiology, First Hospital of Lanzhou University, Lanzhou, 730000, GanSu Province, China
| |
Collapse
|
17
|
Li MY, Liu LZ, Xin Q, Zhou J, Zhang X, Zhang R, Wu Z, Yi J, Dong M. Downregulation of mTORC1 and Mcl-1 by lipid-oversupply contributes to islet β-cell apoptosis and dysfunction. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159332. [PMID: 37196823 DOI: 10.1016/j.bbalip.2023.159332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 04/22/2023] [Accepted: 04/30/2023] [Indexed: 05/19/2023]
Abstract
Pancreatic β-cell apoptosis is a key feature of diabetes and can be induced by chronic exposure to saturated fatty acids (FAs). However, the underlying mechanisms remain poorly understood. We presently evaluated the role of Mcl-1 and mTOR in mice fed with high-fat-diet (HFD) and β-cells exposed to the overloaded palmitic acid (PA). Compared with normal-chow-diet (NCD)-fed mice, HFD group showed impaired glucose tolerance after two months. Along with the diabetes progression, pancreatic islets first became hypertrophic and then atrophic, the ratio of β-cell:α-cell increased in the islets of four months HFD-fed mice while decreased after six months. This process was accompanied by significantly increased β-cell apoptosis and AMPK activity, and decreased Mcl-1 expression and mTOR activity. Consistently, glucose-induced insulin secretion dropped. In terms of mechanism, PA with lipotoxic dose could activate AMPK, which in turn inhibited ERK-stimulated Mcl-1Thr163 phosphorylation. Meanwhile, AMPK blocked Akt activity to release Akt inhibition on GSK3β, followed by GSK3β-initiated Mcl-1Ser159 phosphorylation. The context of Mcl-1 phosphorylation finally led to its degradation by ubiquitination. Also, AMPK inhibited the activity of mTORC1, resulting in a lower level of Mcl-1. Suppression of mTORC1 activity and Mcl-1 expression positively related to β-cell failure. Alteration of Mcl-1 or mTOR expression rendered different tolerance of β-cell to different dose of PA. In conclusion, lipid oversupply-induced dual modulation of mTORC1 and Mcl-1 finally led to β-cell apoptosis and impaired insulin secretion. The study may help further understand the pathogenesis of β-cell dysfunction in case of dyslipidemia, and provide promising therapeutic targets for diabetes.
Collapse
Affiliation(s)
- Ming-Yue Li
- Department of Physiology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, Guangdong, China; GuangZhou Laboratory, No.9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou 510005, Guangdong, China
| | - Li-Zhong Liu
- Department of Physiology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Qihang Xin
- Department of Physiology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Jiaying Zhou
- Department of Physiology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Xiaoyang Zhang
- Department of Physiology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Rui Zhang
- GuangZhou Laboratory, No.9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou 510005, Guangdong, China
| | - Zangshu Wu
- GuangZhou Laboratory, No.9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou 510005, Guangdong, China
| | - Junbo Yi
- Department of Physiology, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, Guangdong, China
| | - Ming Dong
- GuangZhou Laboratory, No.9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou 510005, Guangdong, China.
| |
Collapse
|
18
|
Zhang T, Xu D, Trefts E, Lv M, Inuzuka H, Song G, Liu M, Lu J, Liu J, Chu C, Wang M, Wang H, Meng H, Liu H, Zhuang Y, Xie X, Dang F, Guan D, Men Y, Jiang S, Jiang C, Dai X, Liu J, Wang Z, Yan P, Wang J, Tu Z, Babuta M, Erickson E, Hillis AL, Dibble CC, Asara JM, Szabo G, Sicinski P, Miao J, Lee YR, Pan L, Shaw RJ, Yuan J, Wei W. Metabolic orchestration of cell death by AMPK-mediated phosphorylation of RIPK1. Science 2023; 380:1372-1380. [PMID: 37384704 PMCID: PMC10617018 DOI: 10.1126/science.abn1725] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 05/04/2023] [Indexed: 07/01/2023]
Abstract
Adenosine monophosphate-activated protein kinase (AMPK) activity is stimulated to promote metabolic adaptation upon energy stress. However, sustained metabolic stress may cause cell death. The mechanisms by which AMPK dictates cell death are not fully understood. We report that metabolic stress promoted receptor-interacting protein kinase 1 (RIPK1) activation mediated by TRAIL receptors, whereas AMPK inhibited RIPK1 by phosphorylation at Ser415 to suppress energy stress-induced cell death. Inhibiting pS415-RIPK1 by Ampk deficiency or RIPK1 S415A mutation promoted RIPK1 activation. Furthermore, genetic inactivation of RIPK1 protected against ischemic injury in myeloid Ampkα1-deficient mice. Our studies reveal that AMPK phosphorylation of RIPK1 represents a crucial metabolic checkpoint, which dictates cell fate response to metabolic stress, and highlight a previously unappreciated role for the AMPK-RIPK1 axis in integrating metabolism, cell death, and inflammation.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Daichao Xu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201203 Shanghai, China
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Elijah Trefts
- The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Mingming Lv
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Hiroyuki Inuzuka
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Guobin Song
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Min Liu
- Transfusion Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jianlin Lu
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Jianping Liu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201203 Shanghai, China
| | - Chen Chu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Min Wang
- Department of Biliary-Pancreatic Surgery, Affiliated Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, Hubei, China
| | - Huibing Wang
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Huyan Meng
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA 02115, USA
| | - Hui Liu
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Yuan Zhuang
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Xingxing Xie
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201203 Shanghai, China
| | - Fabin Dang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Dongxian Guan
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yuqin Men
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Shuwen Jiang
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Department of Metabolic and Bariatric Surgery, The First Affiliated Hospital of Jinan University, 510632 Guangzhou, China
| | - Cong Jiang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Xiaoming Dai
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jing Liu
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Zhen Wang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Peiqiang Yan
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jingchao Wang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Zhenbo Tu
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Mrigya Babuta
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Emily Erickson
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Alissandra L Hillis
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Christian C Dibble
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - John M Asara
- Division of Signal Transduction, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Gyongy Szabo
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Piotr Sicinski
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
- Department of Histology and Embryology, Center for Biostructure Research, Medical University of Warsaw, 02-004 Warsaw, Poland
| | - Ji Miao
- Division of Endocrinology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yu-Ru Lee
- Institute of Biomedical Sciences, Academia Sinica, Taipei 115201, Taiwan
| | - Lifeng Pan
- State Key Laboratory of Bioorganic and Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 200032 Shanghai, China
| | - Reuben J Shaw
- The Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Junying Yuan
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, 201203 Shanghai, China
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
19
|
Azimzadeh O, Merl-Pham J, Subramanian V, Oleksenko K, Krumm F, Mancuso M, Pasquali E, Tanaka IB, Tanaka S, Atkinson MJ, Tapio S, Moertl S. Late Effects of Chronic Low Dose Rate Total Body Irradiation on the Heart Proteome of ApoE -/- Mice Resemble Premature Cardiac Ageing. Cancers (Basel) 2023; 15:3417. [PMID: 37444528 DOI: 10.3390/cancers15133417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/20/2023] [Accepted: 06/21/2023] [Indexed: 07/15/2023] Open
Abstract
Recent epidemiologic studies support an association between chronic low-dose radiation exposure and the development of cardiovascular disease (CVD). The molecular mechanisms underlying the adverse effect of chronic low dose exposure are not fully understood. To address this issue, we have investigated changes in the heart proteome of ApoE deficient (ApoE-/-) C57Bl/6 female mice chronically irradiated for 300 days at a very low dose rate (1 mGy/day) or at a low dose rate (20 mGy/day), resulting in cumulative whole-body doses of 0.3 Gy or 6.0 Gy, respectively. The heart proteomes were compared to those of age-matched sham-irradiated ApoE-/- mice using label-free quantitative proteomics. Radiation-induced proteome changes were further validated using immunoblotting, enzyme activity assays, immunohistochemistry or targeted transcriptomics. The analyses showed persistent alterations in the cardiac proteome at both dose rates; however, the effect was more pronounced following higher dose rates. The altered proteins were involved in cardiac energy metabolism, ECM remodelling, oxidative stress, and ageing signalling pathways. The changes in PPARα, SIRT, AMPK, and mTOR signalling pathways were found at both dose rates and in a dose-dependent manner, whereas more changes in glycolysis and ECM remodelling were detected at the lower dose rate. These data provide strong evidence for the possible risk of cardiac injury following chronic low dose irradiation and show that several affected pathways following chronic irradiation overlap with those of ageing-associated heart pathology.
Collapse
Affiliation(s)
- Omid Azimzadeh
- Section of Radiation Biology, Federal Office of Radiation Protection (BfS), 85764 Nauenberg, Germany
| | - Juliane Merl-Pham
- Metabolomics and Proteomics Core, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Munich, Germany
| | - Vikram Subramanian
- Abboud Cardiovascular Research Center, Division of Cardiovascular Medicine, Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Kateryna Oleksenko
- Institute of Radiation Biology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany
| | - Franziska Krumm
- Section of Radiation Biology, Federal Office of Radiation Protection (BfS), 85764 Nauenberg, Germany
| | - Mariateresa Mancuso
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), 00196 Rome, Italy
| | - Emanuela Pasquali
- Laboratory of Biomedical Technologies, Agenzia Nazionale per le Nuove Tecnologie, l'Energia e lo Sviluppo Economico Sostenibile (ENEA), 00196 Rome, Italy
| | - Ignacia B Tanaka
- Institute for Environmental Sciences (IES), Rokkasho, Aomori 039-3212, Japan
| | - Satoshi Tanaka
- Institute for Environmental Sciences (IES), Rokkasho, Aomori 039-3212, Japan
| | - Michael J Atkinson
- Institute of Radiation Biology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany
- Radiation Oncology, Klinikum rechts der Isar, Technical University, 80333 Munich, Germany
| | - Soile Tapio
- Institute of Radiation Biology, Helmholtz Zentrum München, German Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany
| | - Simone Moertl
- Section of Radiation Biology, Federal Office of Radiation Protection (BfS), 85764 Nauenberg, Germany
| |
Collapse
|
20
|
Sharma A, Anand SK, Singh N, Dwivedi UN, Kakkar P. AMP-activated protein kinase: An energy sensor and survival mechanism in the reinstatement of metabolic homeostasis. Exp Cell Res 2023; 428:113614. [PMID: 37127064 DOI: 10.1016/j.yexcr.2023.113614] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 04/18/2023] [Accepted: 04/22/2023] [Indexed: 05/03/2023]
Abstract
Cells are programmed to favorably respond towards the nutrient availability by adapting their metabolism to meet energy demands. AMP-activated protein kinase (AMPK) is a highly conserved serine/threonine energy-sensing kinase. It gets activated upon a decrease in the cellular energy status as reflected by an increased AMP/ATP ratio, ADP, and also during the conditions of glucose starvation without change in the adenine nucelotide ratio. AMPK functions as a centralized regulator of metabolism, acting at cellular and physiological levels to circumvent the metabolic stress by restoring energy balance. This review intricately highlights the integrated signaling pathways by which AMPK gets activated allosterically or by multiple non-canonical upstream kinases. AMPK activates the ATP generating processes (e.g., fatty acid oxidation) and inhibits the ATP consuming processes that are non-critical for survival (e.g., cell proliferation, protein and triglyceride synthesis). An integrated signaling network with AMPK as the central effector regulates all the aspects of enhanced stress resistance, qualified cellular housekeeping, and energy metabolic homeostasis. Importantly, the AMPK mediated amelioration of cellular stress and inflammatory responses are mediated by stimulation of transcription factors such as Nrf2, SIRT1, FoxO and inhibition of NF-κB serving as main downstream effectors. Moreover, many lines of evidence have demonstrated that AMPK controls autophagy through mTOR and ULK1 signaling to fine-tune the metabolic pathways in response to different cellular signals. This review also highlights the critical involvement of AMPK in promoting mitochondrial health, and homeostasis, including mitophagy. Loss of AMPK or ULK1 activity leads to aberrant accumulation of autophagy-related proteins and defective mitophagy thus, connecting cellular energy sensing to autophagy and mitophagy.
Collapse
Affiliation(s)
- Ankita Sharma
- Herbal Research Laboratory, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, India; Department of Biochemistry, University of Lucknow, Lucknow, 226007, India; Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Bijnor-Sisendi Road, Post Office Mati, Lucknow, 226002, India.
| | - Sumit Kr Anand
- Herbal Research Laboratory, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Department of Pathology, LSU Health, 1501 Kings Hwy, Shreveport, LA, 71103, USA.
| | - Neha Singh
- Herbal Research Laboratory, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| | | | - Poonam Kakkar
- Herbal Research Laboratory, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, 226001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
21
|
Steinberg GR, Hardie DG. New insights into activation and function of the AMPK. Nat Rev Mol Cell Biol 2023; 24:255-272. [PMID: 36316383 DOI: 10.1038/s41580-022-00547-x] [Citation(s) in RCA: 234] [Impact Index Per Article: 234.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2022] [Indexed: 11/06/2022]
Abstract
The classical role of AMP-activated protein kinase (AMPK) is as a cellular energy sensor activated by falling energy status, signalled by increases in AMP to ATP and ADP to ATP ratios. Once activated, AMPK acts to restore energy homeostasis by promoting ATP-producing catabolic pathways while inhibiting energy-consuming processes. In this Review, we provide an update on this canonical (AMP/ADP-dependent) activation mechanism, but focus mainly on recently described non-canonical pathways, including those by which AMPK senses the availability of glucose, glycogen or fatty acids and by which it senses damage to lysosomes and nuclear DNA. We also discuss new findings on the regulation of carbohydrate and lipid metabolism, mitochondrial and lysosomal homeostasis, and DNA repair. Finally, we discuss the role of AMPK in cancer, obesity, diabetes, nonalcoholic steatohepatitis (NASH) and other disorders where therapeutic targeting may exert beneficial effects.
Collapse
Affiliation(s)
- Gregory R Steinberg
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada.
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada.
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada.
| | - D Grahame Hardie
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Dundee, UK.
| |
Collapse
|
22
|
Ling J, Hua L, Qin Y, Gu T, Jiang S, Zhao J. Perfluorooctane sulfonate promotes hepatic lipid accumulation and steatosis in high-fat diet mice through AMP-activated protein kinase/acetyl-CoA carboxylase (AMPK/ACC) pathway. J Appl Toxicol 2023; 43:312-322. [PMID: 35999056 DOI: 10.1002/jat.4383] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/20/2022] [Accepted: 08/20/2022] [Indexed: 01/17/2023]
Abstract
Perfluorooctane sulfonate (PFOS) is a hepatotoxic environmental organic pollutant that can cause aberrant lipid accumulation in the liver. However, the molecular mechanism underlying PFOS-induced hepatic steatosis remains unclear. Our research showed that subchronic PFOS exposure inhibited AMP-activated protein kinase (AMPK) phosphorylation, leading to increased acetyl-CoA carboxylase (ACC) activity, attenuated fatty acid β-oxidation, and consequent liver lipid accumulation. We found that 1 mg/kg/day PFOS exposure significantly aggravated steatosis in high-fat diet (HFD)-fed mice, along with reduced AMPK activity. Oil Red O results showed that PFOS exposure caused fat accumulation in HepG2 cells. As predicted, PFOS treatment reduced the level of phosphorylated AMPK in a concentration-dependent manner, leading to subsequent increase in ACC activity and lipid droplet accumulation in HepG2 cells. Treatment with 200-μM AMPK agonist AICAR alleviated PFOS-induced ACC activation and lipid accumulation. In summary, our data highlight a crucial role of AMPK/ACC pathway in PFOS-mediated liver lipid metabolic disorders.
Collapse
Affiliation(s)
- Junyi Ling
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, China
| | - Lu Hua
- Department of Oncology, Taizhou People's Hospital, Taizhou, China
| | - Yi Qin
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, China.,Haimen District Center for Disease Control and Prevention, Nantong, China
| | - Tianye Gu
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, China
| | - Shengyang Jiang
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong, China.,Jiangsu Nantong Health Higher Vocational Technical School, Nantong, China
| | - Jianya Zhao
- Department of Nutrition and Food Hygiene, School of Public Health, Nantong University, Nantong, China
| |
Collapse
|
23
|
Huynh C, Ryu J, Lee J, Inoki A, Inoki K. Nutrient-sensing mTORC1 and AMPK pathways in chronic kidney diseases. Nat Rev Nephrol 2023; 19:102-122. [PMID: 36434160 DOI: 10.1038/s41581-022-00648-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2022] [Indexed: 11/27/2022]
Abstract
Nutrients such as glucose, amino acids and lipids are fundamental sources for the maintenance of essential cellular processes and homeostasis in all organisms. The nutrient-sensing kinases mechanistic target of rapamycin (mTOR) and AMP-activated protein kinase (AMPK) are expressed in many cell types and have key roles in the control of cell growth, proliferation, differentiation, metabolism and survival, ultimately contributing to the physiological development and functions of various organs, including the kidney. Dysregulation of these kinases leads to many human health problems, including cancer, neurodegenerative diseases, metabolic disorders and kidney diseases. In the kidney, physiological levels of mTOR and AMPK activity are required to support kidney cell growth and differentiation and to maintain kidney cell integrity and normal nephron function, including transport of electrolytes, water and glucose. mTOR forms two functional multi-protein kinase complexes, mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2). Hyperactivation of mTORC1 leads to podocyte and tubular cell dysfunction and vulnerability to injury, thereby contributing to the development of chronic kidney diseases, including diabetic kidney disease, obesity-related kidney disease and polycystic kidney disease. Emerging evidence suggests that targeting mTOR and/or AMPK could be an effective therapeutic approach to controlling or preventing these diseases.
Collapse
Affiliation(s)
- Christopher Huynh
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA.,Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jaewhee Ryu
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Jooho Lee
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | - Ayaka Inoki
- Department of Biology, Johns Hopkins University, Baltimore, MD, USA.,Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Ken Inoki
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA. .,Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, USA. .,Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
24
|
Novel Anti-Cancer Products Targeting AMPK: Natural Herbal Medicine against Breast Cancer. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28020740. [PMID: 36677797 PMCID: PMC9863744 DOI: 10.3390/molecules28020740] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/01/2023] [Accepted: 01/04/2023] [Indexed: 01/15/2023]
Abstract
Breast cancer is a common cancer in women worldwide. The existing clinical treatment strategies have been able to limit the progression of breast cancer and cancer metastasis, but abnormal metabolism, immunosuppression, and multidrug resistance involving multiple regulators remain the major challenges for the treatment of breast cancer. Adenosine 5'-monophosphate (AMP)-Activated Protein Kinase (AMPK) can regulate metabolic reprogramming and reverse the "Warburg effect" via multiple metabolic signaling pathways in breast cancer. Previous studies suggest that the activation of AMPK suppresses the growth and metastasis of breast cancer cells, as well as stimulating the responses of immune cells. However, some other reports claim that the development and poor prognosis of breast cancer are related to the overexpression and aberrant activation of AMPK. Thus, the role of AMPK in the progression of breast cancer is still controversial. In this review, we summarize the current understanding of AMPK, particularly the comprehensive bidirectional functions of AMPK in cancer progression; discuss the pharmacological activators of AMPK and some specific molecules, including the natural products (including berberine, curcumin, (-)-epigallocatechin-3-gallate, ginsenosides, and paclitaxel) that influence the efficacy of these activators in cancer therapy; and elaborate the role of AMPK as a potential therapeutic target for the treatment of breast cancer.
Collapse
|
25
|
Keerthana CK, Rayginia TP, Shifana SC, Anto NP, Kalimuthu K, Isakov N, Anto RJ. The role of AMPK in cancer metabolism and its impact on the immunomodulation of the tumor microenvironment. Front Immunol 2023; 14:1114582. [PMID: 36875093 PMCID: PMC9975160 DOI: 10.3389/fimmu.2023.1114582] [Citation(s) in RCA: 45] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/03/2023] [Indexed: 02/17/2023] Open
Abstract
Adenosine monophosphate-activated protein kinase (AMPK) is a key metabolic sensor that is pivotal for the maintenance of cellular energy homeostasis. AMPK contributes to diverse metabolic and physiological effects besides its fundamental role in glucose and lipid metabolism. Aberrancy in AMPK signaling is one of the determining factors which lead to the development of chronic diseases such as obesity, inflammation, diabetes, and cancer. The activation of AMPK and its downstream signaling cascades orchestrate dynamic changes in the tumor cellular bioenergetics. It is well documented that AMPK possesses a suppressor role in the context of tumor development and progression by modulating the inflammatory and metabolic pathways. In addition, AMPK plays a central role in potentiating the phenotypic and functional reprogramming of various classes of immune cells which reside in the tumor microenvironment (TME). Furthermore, AMPK-mediated inflammatory responses facilitate the recruitment of certain types of immune cells to the TME, which impedes the development, progression, and metastasis of cancer. Thus, AMPK appears to play an important role in the regulation of anti-tumor immune response by regulating the metabolic plasticity of various immune cells. AMPK effectuates the metabolic modulation of anti-tumor immunity via nutrient regulation in the TME and by virtue of its molecular crosstalk with major immune checkpoints. Several studies including that from our lab emphasize on the role of AMPK in regulating the anticancer effects of several phytochemicals, which are potential anticancer drug candidates. The scope of this review encompasses the significance of the AMPK signaling in cancer metabolism and its influence on the key drivers of immune responses within the TME, with a special emphasis on the potential use of phytochemicals to target AMPK and combat cancer by modulating the tumor metabolism.
Collapse
Affiliation(s)
- Chenicheri Kizhakkeveettil Keerthana
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.,Department of Biotechnology, University of Kerala, Thiruvananthapuram, Kerala, India
| | - Tennyson Prakash Rayginia
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.,Department of Biotechnology, University of Kerala, Thiruvananthapuram, Kerala, India
| | | | - Nikhil Ponnoor Anto
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Kalishwaralal Kalimuthu
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Noah Isakov
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Ruby John Anto
- Division of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| |
Collapse
|
26
|
Hardie DG. 100 years of the Warburg effect: a historical perspective. Endocr Relat Cancer 2022; 29:T1-T13. [PMID: 36094878 DOI: 10.1530/erc-22-0173] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/11/2022] [Indexed: 11/08/2022]
Abstract
Otto Warburg published the first paper describing what became known as the Warburg effect in 1923. All that was known about glucose metabolism at that time was that it occurred in two stages: (i) fermentation (glycolysis) in which glucose was converted to lactate, which did not require oxygen, and (ii) oxidative metabolism, in which the carbon atoms derived from glycolysis were fully oxidized to carbon dioxide, which did require oxygen. Warburg discovered that most tumour tissues produced a large amount of lactate that was reduced but not eliminated in the presence of oxygen, while most normal tissues produced a much smaller amount of lactate that was eliminated by the provision of oxygen. These findings were clearly well ahead of their time because it was another 80 years before they were to have any major impact, and even today the mechanisms underlying the Warburg effect are not completely understood.
Collapse
Affiliation(s)
- D Grahame Hardie
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Dundee, Scotland, UK
| |
Collapse
|
27
|
Hardie DG. AMP-activated protein kinase - a journey from 1 to 100 downstream targets. Biochem J 2022; 479:2327-2343. [PMID: 36383046 PMCID: PMC9704532 DOI: 10.1042/bcj20220255] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 10/26/2022] [Accepted: 10/26/2022] [Indexed: 11/17/2022]
Abstract
A casual decision made one evening in 1976, in a bar near the Biochemistry Department at the University of Dundee, led me to start my personal research journey by following up a paper that suggested that acetyl-CoA carboxylase (ACC) (believed to be a key regulatory enzyme of fatty acid synthesis) was inactivated by phosphorylation by what appeared to be a novel, cyclic AMP-independent protein kinase. This led me to define and name the AMP-activated protein kinase (AMPK) signalling pathway, on which I am still working 46 years later. ACC was the first known downstream target for AMPK, but at least 100 others have now been identified. This article contains some personal reminiscences of that research journey, focussing on: (i) the early days when we were defining the kinase and developing the key tools required to study it; (ii) the late 1990s and early 2000s, an exciting time when we and others were identifying the upstream kinases; (iii) recent times when we have been studying the complex role of AMPK in cancer. The article is published in conjunction with the Sir Philip Randle Lecture of the Biochemical Society, which I gave in September 2022 at the European Workshop on AMPK and AMPK-related kinases in Clydebank, Scotland. During the early years of my research career, Sir Philip acted as a role model, due to his pioneering work on insulin signalling and the regulation of pyruvate dehydrogenase.
Collapse
Affiliation(s)
- D. Grahame Hardie
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, Scotland, U.K
| |
Collapse
|
28
|
Soto-Verdugo J, Siva-Parra J, Hernández-Kelly LC, Ortega A. Acute Manganese Exposure Modifies the Translation Machinery via PI3K/Akt Signaling in Glial Cells. ASN Neuro 2022; 14:17590914221131452. [PMID: 36203371 PMCID: PMC9551334 DOI: 10.1177/17590914221131452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
SUMMARY STATEMENT We demonstrate herein that short-term exposure of radial glia cells to Manganese, a neurotoxic metal, induces an effect on protein synthesis, altering the protein repertoire of these cells.
Collapse
Affiliation(s)
| | | | | | - Arturo Ortega
- Arturo Ortega, Departamento de Toxicología,
Centro de Investigación y de Estudios Avanzados del Instituto Politécnico
Nacional, México City, México, 07360.
| |
Collapse
|
29
|
Cai J, Chen X, Liu X, Li Z, Shi A, Tang X, Xia P, Zhang J, Yu P. AMPK: The key to ischemia-reperfusion injury. J Cell Physiol 2022; 237:4079-4096. [PMID: 36134582 DOI: 10.1002/jcp.30875] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 08/08/2022] [Accepted: 08/23/2022] [Indexed: 11/09/2022]
Abstract
Ischemia-reperfusion injury (IRI) refers to a syndrome in which tissue damage is further aggravated and organ function further deteriorates when blood flow is restored after a period of tissue ischemia. Acute myocardial infarction, stress ulcer, pancreatitis, intestinal ischemia, intermittent claudication, acute tubular necrosis, postshock liver failure, and multisystem organ failure are all related to reperfusion injury. AMP-activated protein kinase (AMPK) has been identified in multiple catabolic and anabolic signaling pathways. The functions of AMPK during health and diseases are intriguing but still need further research. Except for its conventional roles as an intracellular energy switch, emerging evidence reveals the critical role of AMPK in IRI as an energy-sensing signal molecule by regulating metabolism, autophagy, oxidative stress, inflammation, and other progressions. At the same time, drugs based on AMPK for the treatment of IRI are constantly being researched and applied in clinics. In this review, we summarize the mechanisms underlying the effects of AMPK in IRI and describe the AMPK-targeting drugs in treatment, hoping to increase the understanding of AMPK in IRI and provide new insights into future clinical treatment.
Collapse
Affiliation(s)
- Jie Cai
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xinyue Chen
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xingyu Liu
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zhangwang Li
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Ao Shi
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA.,Department of Biochemistry and Molecular Biology, Mayo Graduate School of Biomedical Science, Mayo Clinic, Rochester, Minnesota, USA
| | - Xiaoyi Tang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Panpan Xia
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Peng Yu
- Department of Metabolism and Endocrinology, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, China
| |
Collapse
|
30
|
Harnessing conserved signaling and metabolic pathways to enhance the maturation of functional engineered tissues. NPJ Regen Med 2022; 7:44. [PMID: 36057642 PMCID: PMC9440900 DOI: 10.1038/s41536-022-00246-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 08/05/2022] [Indexed: 11/08/2022] Open
Abstract
The development of induced-pluripotent stem cell (iPSC)-derived cell types offers promise for basic science, drug testing, disease modeling, personalized medicine, and translatable cell therapies across many tissue types. However, in practice many iPSC-derived cells have presented as immature in physiological function, and despite efforts to recapitulate adult maturity, most have yet to meet the necessary benchmarks for the intended tissues. Here, we summarize the available state of knowledge surrounding the physiological mechanisms underlying cell maturation in several key tissues. Common signaling consolidators, as well as potential synergies between critical signaling pathways are explored. Finally, current practices in physiologically relevant tissue engineering and experimental design are critically examined, with the goal of integrating greater decision paradigms and frameworks towards achieving efficient maturation strategies, which in turn may produce higher-valued iPSC-derived tissues.
Collapse
|
31
|
Stocks B, Zierath JR. Post-translational Modifications: The Signals at the Intersection of Exercise, Glucose Uptake, and Insulin Sensitivity. Endocr Rev 2022; 43:654-677. [PMID: 34730177 PMCID: PMC9277643 DOI: 10.1210/endrev/bnab038] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Indexed: 11/19/2022]
Abstract
Diabetes is a global epidemic, of which type 2 diabetes makes up the majority of cases. Nonetheless, for some individuals, type 2 diabetes is eminently preventable and treatable via lifestyle interventions. Glucose uptake into skeletal muscle increases during and in recovery from exercise, with exercise effective at controlling glucose homeostasis in individuals with type 2 diabetes. Furthermore, acute and chronic exercise sensitizes skeletal muscle to insulin. A complex network of signals converge and interact to regulate glucose metabolism and insulin sensitivity in response to exercise. Numerous forms of post-translational modifications (eg, phosphorylation, ubiquitination, acetylation, ribosylation, and more) are regulated by exercise. Here we review the current state of the art of the role of post-translational modifications in transducing exercise-induced signals to modulate glucose uptake and insulin sensitivity within skeletal muscle. Furthermore, we consider emerging evidence for noncanonical signaling in the control of glucose homeostasis and the potential for regulation by exercise. While exercise is clearly an effective intervention to reduce glycemia and improve insulin sensitivity, the insulin- and exercise-sensitive signaling networks orchestrating this biology are not fully clarified. Elucidation of the complex proteome-wide interactions between post-translational modifications and the associated functional implications will identify mechanisms by which exercise regulates glucose homeostasis and insulin sensitivity. In doing so, this knowledge should illuminate novel therapeutic targets to enhance insulin sensitivity for the clinical management of type 2 diabetes.
Collapse
Affiliation(s)
- Ben Stocks
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Juleen R Zierath
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.,Departments of Molecular Medicine and Surgery and Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
32
|
Flores K, Siques P, Brito J, Arribas SM. AMPK and the Challenge of Treating Hypoxic Pulmonary Hypertension. Int J Mol Sci 2022; 23:ijms23116205. [PMID: 35682884 PMCID: PMC9181235 DOI: 10.3390/ijms23116205] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/29/2022] [Accepted: 04/30/2022] [Indexed: 02/01/2023] Open
Abstract
Hypoxic pulmonary hypertension (HPH) is characterized by sustained elevation of pulmonary artery pressure produced by vasoconstriction and hyperproliferative remodeling of the pulmonary artery and subsequent right ventricular hypertrophy (RVH). The search for therapeutic targets for cardiovascular pathophysiology has extended in many directions. However, studies focused on mitigating high-altitude pulmonary hypertension (HAPH) have been rare. Because AMP-activated protein kinase (AMPK) is involved in cardiovascular and metabolic pathology, AMPK is often studied as a potential therapeutic target. AMPK is best characterized as a sensor of cellular energy that can also restore cellular metabolic homeostasis. However, AMPK has been implicated in other pathways with vasculoprotective effects. Notably, cellular metabolic stress increases the intracellular ADP/ATP or AMP/ATP ratio, and AMPK activation restores ATP levels by activating energy-producing catabolic pathways and inhibiting energy-consuming anabolic pathways, such as cell growth and proliferation pathways, promoting cardiovascular protection. Thus, AMPK activation plays an important role in antiproliferative, antihypertrophic and antioxidant pathways in the pulmonary artery in HPH. However, AMPK plays contradictory roles in promoting HPH development. This review describes the main findings related to AMPK participation in HPH and its potential as a therapeutic target. It also extrapolates known AMPK functions to discuss the less-studied HAPH context.
Collapse
Affiliation(s)
- Karen Flores
- Institute of Health Studies, University Arturo Prat, Av. Arturo Prat 2120, Iquique 1110939, Chile; (P.S.); (J.B.)
- Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae, 20251 Hamburg, Germany and Iquique 1100000, Chile
- Correspondence: ; Tel.: +56-572526392
| | - Patricia Siques
- Institute of Health Studies, University Arturo Prat, Av. Arturo Prat 2120, Iquique 1110939, Chile; (P.S.); (J.B.)
- Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae, 20251 Hamburg, Germany and Iquique 1100000, Chile
| | - Julio Brito
- Institute of Health Studies, University Arturo Prat, Av. Arturo Prat 2120, Iquique 1110939, Chile; (P.S.); (J.B.)
- Institute DECIPHER, German-Chilean Institute for Research on Pulmonary Hypoxia and Its Health Sequelae, 20251 Hamburg, Germany and Iquique 1100000, Chile
| | - Silvia M. Arribas
- Department of Physiology, University Autonoma of Madrid, 28049 Madrid, Spain;
| |
Collapse
|
33
|
Le N, Sayers S, Mata-Pacheco V, Wagner EJ. The PACAP Paradox: Dynamic and Surprisingly Pleiotropic Actions in the Central Regulation of Energy Homeostasis. Front Endocrinol (Lausanne) 2022; 13:877647. [PMID: 35721722 PMCID: PMC9198406 DOI: 10.3389/fendo.2022.877647] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 03/30/2022] [Indexed: 12/11/2022] Open
Abstract
Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP), a pleiotropic neuropeptide, is widely distributed throughout the body. The abundance of PACAP expression in the central and peripheral nervous systems, and years of accompanying experimental evidence, indicates that PACAP plays crucial roles in diverse biological processes ranging from autonomic regulation to neuroprotection. In addition, PACAP is also abundantly expressed in the hypothalamic areas like the ventromedial and arcuate nuclei (VMN and ARC, respectively), as well as other brain regions such as the nucleus accumbens (NAc), bed nucleus of stria terminalis (BNST), and ventral tegmental area (VTA) - suggesting that PACAP is capable of regulating energy homeostasis via both the homeostatic and hedonic energy balance circuitries. The evidence gathered over the years has increased our appreciation for its function in controlling energy balance. Therefore, this review aims to further probe how the pleiotropic actions of PACAP in regulating energy homeostasis is influenced by sex and dynamic changes in energy status. We start with a general overview of energy homeostasis, and then introduce the integral components of the homeostatic and hedonic energy balance circuitries. Next, we discuss sex differences inherent to the regulation of energy homeostasis via these two circuitries, as well as the activational effects of sex steroid hormones that bring about these intrinsic disparities between males and females. Finally, we explore the multifaceted role of PACAP in regulating homeostatic and hedonic feeding through its actions in regions like the NAc, BNST, and in particular the ARC, VMN and VTA that occur in sex- and energy status-dependent ways.
Collapse
Affiliation(s)
- Nikki Le
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
| | - Sarah Sayers
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
| | - Veronica Mata-Pacheco
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
| | - Edward J. Wagner
- Graduate College of Biomedical Sciences, Western University of Health Sciences, Pomona, CA, United States
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA, United States
| |
Collapse
|
34
|
Maitiabula G, Tian F, Wang P, Zhang L, Gao X, Wan S, Sun H, Yang J, Zhang Y, Gao T, Xue B, Li C, Li J, Wang X. Liver PP2A-Cα Protects From Parenteral Nutrition-associated Hepatic Steatosis. Cell Mol Gastroenterol Hepatol 2022; 14:669-692. [PMID: 35643235 PMCID: PMC9421584 DOI: 10.1016/j.jcmgh.2022.05.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 05/18/2022] [Accepted: 05/18/2022] [Indexed: 01/07/2023]
Abstract
BACKGROUND & AIMS Parenteral nutrition (PN) is a lifesaving therapy for patients with intestinal failure. Hepatic steatosis is a potentially fatal complication of long-term PN, but the involved pathological mechanisms are incompletely unclarified. Herein, we identify the role of protein phosphatase 2A (PP2A) in the pathogenesis of parenteral nutrition-associated hepatic steatosis (PNAHS). METHODS Proteomic/phosphoproteomic analyses of liver samples from patients with PNAHS were applied to identify the mechanism of PNAHS. Total parenteral nutrition (TPN) mice model, in vivo, and in vitro experiments were used to assess the effect of PP2A-Cα on liver fatty acid metabolism. RESULTS Reduced expression of PP2A-Cα (catalytic subunit) enhanced activation of serine/threonine kinase Akt2 and decreased activation of adenosine monophosphate-activated protein kinase (AMPK) were associated with hepatic steatosis in patients with PNAHS. Mice given PN for 14 days developed hepatic steatosis, down-regulation of PP2A-Cα, activation of Akt2, and inhibition of AMPK. Hepatocyte-specific deletion of PP2A-Cα in mice given PN exacerbated Akt2 activation, AMPK inhibition, and hepatic steatosis through an effect on fatty acid degradation, whereas hepatocyte-specific PP2A-Cα overexpression significantly ameliorated hepatic steatosis accompanied with Akt2 suppression and AMPK activation. Additionally, pharmacological activation of Akt2 in mice overexpressing PP2A-Cα led to the aggravation of hepatic steatosis. CONCLUSIONS Our findings demonstrate that hepatic PP2A-Cα serves as a protective factor of PNAHS due to ameliorating hepatic steatosis and improving liver function. Our study provides a strong rationale that PP2A-Cα may be involved in the pathogenesis of PNAHS.
Collapse
Affiliation(s)
- Gulisudumu Maitiabula
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Feng Tian
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Peng Wang
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Li Zhang
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xuejin Gao
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Songlin Wan
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Haifeng Sun
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Jianbo Yang
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yupeng Zhang
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Tingting Gao
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Bin Xue
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center of the Medical School of Nanjing University, Nanjing, China,Core Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China,Bin Xue, PhD, LongMian Avenue, Nanjing 211166, China. tel: +86-25-87115542
| | - Chaojun Li
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center of the Medical School of Nanjing University, Nanjing, China,Chaojun Li, PhD, Hankou Road, Nanjing, 210093, China. tel: +86-25-83596289.
| | - Jieshou Li
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Xinying Wang
- Department of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China,Correspondence Address correspondence to: Xinying Wang, MD, PhD, Department of General Surgery, Jinling Hospital, Medical School of Nanjing University. 305 East Zhongshan Road, Nanjing, 210002, China. tel: +86-25-80861429
| |
Collapse
|
35
|
Ma S, Dong Z, Huang Y, Liu JY, Zhang JT. Translation initiation factor eIF3a regulates glucose metabolism and cell proliferation via promoting small GTPase Rheb synthesis and AMPK activation. J Biol Chem 2022; 298:102044. [PMID: 35595099 PMCID: PMC9207673 DOI: 10.1016/j.jbc.2022.102044] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 12/05/2022] Open
Abstract
Eukaryotic translation initiation factor 3 subunit A (eIF3a), the largest subunit of the eIF3 complex, has been shown to be overexpressed in malignant cancer cells, potentially making it a proto-oncogene. eIF3a overexpression can drive cancer cell proliferation but contributes to better prognosis. While its contribution to prognosis was previously shown to be due to its function in suppressing synthesis of DNA damage repair proteins, it remains unclear how eIF3a regulates cancer cell proliferation. In this study, we show using genetic approaches that eIF3a controls cell proliferation by regulating glucose metabolism via the phosphorylation and activation of AMP-activated protein kinase alpha (AMPKα) at Thr172 in its kinase activation loop. We demonstrate that eIF3a regulates AMPK activation mainly by controlling synthesis of the small GTPase Rheb, largely independent of the well-known AMPK upstream liver kinase B1 and Ca2+/calmodulin-dependent protein kinase kinase 2, and also independent of mammalian target of rapamycin signaling and glucose levels. Our findings suggest that glucose metabolism in and proliferation of cancer cells may be translationally regulated via a novel eIF3a–Rheb–AMPK signaling axis.
Collapse
Affiliation(s)
- Shijie Ma
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Zizheng Dong
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Yanfei Huang
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Jing-Yuan Liu
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Jian-Ting Zhang
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA.
| |
Collapse
|
36
|
Cheratta AR, Thayyullathil F, Hawley SA, Ross FA, Atrih A, Lamont DJ, Pallichankandy S, Subburayan K, Alakkal A, Rezgui R, Gray A, Hardie DG, Galadari S. Caspase cleavage and nuclear retention of the energy sensor AMPK-α1 during apoptosis. Cell Rep 2022; 39:110761. [PMID: 35508122 PMCID: PMC9108549 DOI: 10.1016/j.celrep.2022.110761] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 02/11/2022] [Accepted: 04/07/2022] [Indexed: 02/09/2023] Open
Abstract
AMP-activated protein kinase (AMPK) coordinates energy homeostasis during metabolic and energy stress. We report that the catalytic subunit isoform AMPK-α1 (but not α2) is cleaved by caspase-3 at an early stage during induction of apoptosis. AMPK-α1 cleavage occurs following Asp529, generating an ∼58-kDa N-terminal fragment (cl-AMPK-α1) and leading to the precise excision of the nuclear export sequence (NES) from the C-terminal end. This cleavage does not affect (1) the stability of pre-formed heterotrimeric complexes, (2) the ability of cl-AMPK-α1 to become phosphorylated and activated by the upstream kinases LKB1 or CaMKK2, or (3) allosteric activation by AMP or A-769662. Importantly, cl-AMPK-α1 is only detectable in the nucleus, consistent with removal of the NES, and ectopic expression of cleavage-resistant D529A-mutant AMPK-α1 promotes cell death induced by cytotoxic agents. Thus, we have elucidated a non-canonical mechanism of AMPK activation within the nucleus, which protects cells against death induced by DNA damage.
Collapse
Affiliation(s)
- Anees Rahman Cheratta
- Cell Death Signaling Laboratory (Division of Science), Experimental Research Building, New York University Abu Dhabi, PO Box 129188, Saadiyat Island, Abu Dhabi, UAE
| | - Faisal Thayyullathil
- Cell Death Signaling Laboratory (Division of Science), Experimental Research Building, New York University Abu Dhabi, PO Box 129188, Saadiyat Island, Abu Dhabi, UAE
| | - Simon A. Hawley
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Dundee, Scotland DD1 5EH, UK
| | - Fiona A. Ross
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Dundee, Scotland DD1 5EH, UK
| | - Abdelmajdid Atrih
- Fingerprints Proteomics Facility, School of Life Sciences, University of Dundee, Dundee, Scotland DD1 5EH, UK
| | - Douglas J. Lamont
- Fingerprints Proteomics Facility, School of Life Sciences, University of Dundee, Dundee, Scotland DD1 5EH, UK
| | - Siraj Pallichankandy
- Cell Death Signaling Laboratory (Division of Science), Experimental Research Building, New York University Abu Dhabi, PO Box 129188, Saadiyat Island, Abu Dhabi, UAE
| | - Karthikeyan Subburayan
- Cell Death Signaling Laboratory (Division of Science), Experimental Research Building, New York University Abu Dhabi, PO Box 129188, Saadiyat Island, Abu Dhabi, UAE
| | - Ameer Alakkal
- Cell Death Signaling Laboratory (Division of Science), Experimental Research Building, New York University Abu Dhabi, PO Box 129188, Saadiyat Island, Abu Dhabi, UAE
| | - Rachid Rezgui
- Core Technology Platform, Experimental Research Building, New York University Abu Dhabi, PO Box 129188, Saadiyat Island, Abu Dhabi, UAE
| | - Alex Gray
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Dundee, Scotland DD1 5EH, UK
| | - D. Grahame Hardie
- Division of Cell Signalling & Immunology, School of Life Sciences, University of Dundee, Dundee, Scotland DD1 5EH, UK,Corresponding author
| | - Sehamuddin Galadari
- Cell Death Signaling Laboratory (Division of Science), Experimental Research Building, New York University Abu Dhabi, PO Box 129188, Saadiyat Island, Abu Dhabi, UAE.
| |
Collapse
|
37
|
Integrating adipocyte insulin signaling and metabolism in the multi-omics era. Trends Biochem Sci 2022; 47:531-546. [PMID: 35304047 DOI: 10.1016/j.tibs.2022.02.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 02/08/2022] [Accepted: 02/21/2022] [Indexed: 12/16/2022]
Abstract
Insulin stimulates glucose uptake into adipocytes via mTORC2/AKT signaling and GLUT4 translocation and directs glucose carbons into glycolysis, glycerol for TAG synthesis, and de novo lipogenesis. Adipocyte insulin resistance is an early indicator of type 2 diabetes in obesity, a worldwide health crisis. Thus, understanding the interplay between insulin signaling and central carbon metabolism pathways that maintains adipocyte function, blood glucose levels, and metabolic homeostasis is critical. While classically viewed through the lens of individual enzyme-substrate interactions, advances in mass spectrometry are beginning to illuminate adipocyte signaling and metabolic networks on an unprecedented scale, yet this is just the tip of the iceberg. Here, we review how 'omics approaches help to elucidate adipocyte insulin action in cellular time and space.
Collapse
|
38
|
Morrison KR, Smiles WJ, Ling NXY, Hoque A, Shea G, Ngoei KRW, Yu D, Murray-Segal L, Scott JW, Galic S, Kemp BE, Petersen J, Oakhill JS. An AMPKα2-specific phospho-switch controls lysosomal targeting for activation. Cell Rep 2022; 38:110365. [PMID: 35172150 DOI: 10.1016/j.celrep.2022.110365] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 10/28/2021] [Accepted: 01/19/2022] [Indexed: 12/12/2022] Open
Abstract
AMP-activated protein kinase (AMPK) and mechanistic target of rapamycin complex 1 (mTORC1) are metabolic kinases that co-ordinate nutrient supply with cell growth. AMPK negatively regulates mTORC1, and mTORC1 reciprocally phosphorylates S345/7 in both AMPK α-isoforms. We report that genetic or torin1-induced loss of α2-S345 phosphorylation relieves suppression of AMPK signaling; however, the regulatory effect does not translate to α1-S347 in HEK293T or MEF cells. Dephosphorylation of α2-S345, but not α1-S347, transiently targets AMPK to lysosomes, a cellular site for activation by LKB1. By mass spectrometry, we find that α2-S345 is basally phosphorylated at 2.5-fold higher stoichiometry than α1-S347 in HEK293T cells and, unlike α1, phosphorylation is partially retained after prolonged mTORC1 inhibition. Loss of α2-S345 phosphorylation in endogenous AMPK fails to sustain growth of MEFs under amino acid starvation conditions. These findings uncover an α2-specific mechanism by which AMPK can be activated at lysosomes in the absence of changes in cellular energy.
Collapse
Affiliation(s)
- Kaitlin R Morrison
- Flinders Health and Medical Research Institute, Flinders University, Adelaide, SA 5042, Australia
| | - William J Smiles
- Metabolic Signalling Laboratory, St Vincent's Institute of Medical Research, School of Medicine, University of Melbourne, Melbourne, VIC 3065, Australia
| | - Naomi X Y Ling
- Metabolic Signalling Laboratory, St Vincent's Institute of Medical Research, School of Medicine, University of Melbourne, Melbourne, VIC 3065, Australia
| | - Ashfaqul Hoque
- Metabolic Signalling Laboratory, St Vincent's Institute of Medical Research, School of Medicine, University of Melbourne, Melbourne, VIC 3065, Australia
| | - Gabrielle Shea
- Flinders Health and Medical Research Institute, Flinders University, Adelaide, SA 5042, Australia
| | - Kevin R W Ngoei
- Protein Chemistry & Metabolism Unit, St Vincent's Institute of Medical Research, School of Medicine, University of Melbourne, Melbourne, VIC 3065, Australia
| | - Dingyi Yu
- Protein Chemistry & Metabolism Unit, St Vincent's Institute of Medical Research, School of Medicine, University of Melbourne, Melbourne, VIC 3065, Australia
| | - Lisa Murray-Segal
- Protein Chemistry & Metabolism Unit, St Vincent's Institute of Medical Research, School of Medicine, University of Melbourne, Melbourne, VIC 3065, Australia
| | - John W Scott
- Protein Chemistry & Metabolism Unit, St Vincent's Institute of Medical Research, School of Medicine, University of Melbourne, Melbourne, VIC 3065, Australia; Mary MacKillop Institute for Health Research, Australian Catholic University, Fitzroy, VIC 3000, Australia; The Florey Institute of Neuroscience and Mental Health, Parkville, Melbourne, VIC 3052, Australia
| | - Sandra Galic
- Protein Chemistry & Metabolism Unit, St Vincent's Institute of Medical Research, School of Medicine, University of Melbourne, Melbourne, VIC 3065, Australia
| | - Bruce E Kemp
- Protein Chemistry & Metabolism Unit, St Vincent's Institute of Medical Research, School of Medicine, University of Melbourne, Melbourne, VIC 3065, Australia; Mary MacKillop Institute for Health Research, Australian Catholic University, Fitzroy, VIC 3000, Australia
| | - Janni Petersen
- Flinders Health and Medical Research Institute, Flinders University, Adelaide, SA 5042, Australia; Nutrition and Metabolism, South Australia Health and Medical Research Institute, Adelaide, SA, Australia.
| | - Jonathan S Oakhill
- Metabolic Signalling Laboratory, St Vincent's Institute of Medical Research, School of Medicine, University of Melbourne, Melbourne, VIC 3065, Australia.
| |
Collapse
|
39
|
Supplementing Soy-Based Diet with Creatine in Rats: Implications for Cardiac Cell Signaling and Response to Doxorubicin. Nutrients 2022; 14:nu14030583. [PMID: 35276943 PMCID: PMC8840593 DOI: 10.3390/nu14030583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 01/16/2022] [Accepted: 01/21/2022] [Indexed: 02/04/2023] Open
Abstract
Nutritional habits can have a significant impact on cardiovascular health and disease. This may also apply to cardiotoxicity caused as a frequent side effect of chemotherapeutic drugs, such as doxorubicin (DXR). The aim of this work was to analyze if diet, in particular creatine (Cr) supplementation, can modulate cardiac biochemical (energy status, oxidative damage and antioxidant capacity, DNA integrity, cell signaling) and functional parameters at baseline and upon DXR treatment. Here, male Wistar rats were fed for 4 weeks with either standard rodent diet (NORMAL), soy-based diet (SOY), or Cr-supplemented soy-based diet (SOY + Cr). Hearts were either freeze-clamped in situ or following ex vivo Langendorff perfusion without or with 25 μM DXR and after recording cardiac function. The diets had distinct cardiac effects. Soy-based diet (SOY vs. NORMAL) did not alter cardiac performance but increased phosphorylation of acetyl-CoA carboxylase (ACC), indicating activation of rather pro-catabolic AMP-activated protein kinase (AMPK) signaling, consistent with increased ADP/ATP ratios and lower lipid peroxidation. Creatine addition to the soy-based diet (SOY + Cr vs. SOY) slightly increased left ventricular developed pressure (LVDP) and contractility dp/dt, as measured at baseline in perfused heart, and resulted in activation of the rather pro-anabolic protein kinases Akt and ERK. Challenging perfused heart with DXR, as analyzed across all nutritional regimens, deteriorated most cardiac functional parameters and also altered activation of the AMPK, ERK, and Akt signaling pathways. Despite partial reprogramming of cell signaling and metabolism in the rat heart, diet did not modify the functional response to supraclinical DXR concentrations in the used acute cardiotoxicity model. However, the long-term effect of these diets on cardiac sensitivity to chronic and clinically relevant DXR doses remains to be established.
Collapse
|
40
|
Emerging Role of cAMP/AMPK Signaling. Cells 2022; 11:cells11020308. [PMID: 35053423 PMCID: PMC8774420 DOI: 10.3390/cells11020308] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 12/20/2022] Open
Abstract
The 5′-Adenosine monophosphate (AMP)-activated protein kinase (AMPK) is a natural energy sensor in mammalian cells that plays a key role in cellular and systemic energy homeostasis. At the cellular level, AMPK supports numerous processes required for energy and redox homeostasis, including mitochondrial biogenesis, autophagy, and glucose and lipid metabolism. Thus, understanding the pathways regulating AMPK activity is crucial for developing strategies to treat metabolic disorders. Mounting evidence suggests the presence of a link between cyclic AMP (cAMP) and AMPK signaling. cAMP signaling is known to be activated in circumstances of physiological and metabolic stress due to the release of stress hormones, such as adrenaline and glucagon, which is followed by activation of membrane-bound adenylyl cyclase and elevation of cellular cAMP. Because the majority of physiological stresses are associated with elevated energy consumption, it is not surprising that activation of cAMP signaling may promote AMPK activity. Aside from the physiological role of the cAMP/AMPK axis, numerous reports have suggested its role in several pathologies, including inflammation, ischemia, diabetes, obesity, and aging. Furthermore, novel reports have provided more mechanistic insight into the regulation of the cAMP/AMPK axis. In particular, the role of distinct cAMP microdomains generated by soluble adenylyl cyclase in regulating basal and induced AMPK activity has recently been demonstrated. In the present review, we discuss current advances in the understanding of the regulation of the cAMP/AMPK axis and its role in cellular homeostasis and explore some translational aspects.
Collapse
|
41
|
Batchuluun B, Pinkosky SL, Steinberg GR. Lipogenesis inhibitors: therapeutic opportunities and challenges. Nat Rev Drug Discov 2022; 21:283-305. [PMID: 35031766 PMCID: PMC8758994 DOI: 10.1038/s41573-021-00367-2] [Citation(s) in RCA: 149] [Impact Index Per Article: 74.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2021] [Indexed: 12/12/2022]
Abstract
Fatty acids are essential for survival, acting as bioenergetic substrates, structural components and signalling molecules. Given their vital role, cells have evolved mechanisms to generate fatty acids from alternative carbon sources, through a process known as de novo lipogenesis (DNL). Despite the importance of DNL, aberrant upregulation is associated with a wide variety of pathologies. Inhibiting core enzymes of DNL, including citrate/isocitrate carrier (CIC), ATP-citrate lyase (ACLY), acetyl-CoA carboxylase (ACC) and fatty acid synthase (FAS), represents an attractive therapeutic strategy. Despite challenges related to efficacy, selectivity and safety, several new classes of synthetic DNL inhibitors have entered clinical-stage development and may become the foundation for a new class of therapeutics. De novo lipogenesis (DNL) is vital for the maintenance of whole-body and cellular homeostasis, but aberrant upregulation of the pathway is associated with a broad range of conditions, including cardiovascular disease, metabolic disorders and cancers. Here, Steinberg and colleagues provide an overview of the physiological and pathological roles of the core DNL enzymes and assess strategies and agents currently in development to therapeutically target them.
Collapse
Affiliation(s)
- Battsetseg Batchuluun
- Centre for Metabolism, Obesity and Diabetes Research, Department of Medicine and Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | | | - Gregory R Steinberg
- Centre for Metabolism, Obesity and Diabetes Research, Department of Medicine and Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
42
|
Abstract
We previously identified genomic variants that are quantitative trait loci for circulating miR-1908-5p and then showed this microRNA to causally associate with plasma levels of LDL-C, fasting blood glucose and HbA1c. The link to LDL-C was subsequently validated and clarified by the identification of a miR1908-5p-TGFB-LDLR regulatory axis. Here, we continue our investigations on miR1908-5p function by leveraging human primary hepatocytes and HuH-7 hepatoma models. Expression of miR1908-5p was shown to be sensitive to glucose and agents affecting glucose metabolism. Transcriptome-wide changes in primary hepatocytes and HuH-7 cells treated with a miR1908-5p mimic were investigated by enrichment approaches to identify targeted transcripts and cognate pathways. Significant pathways included autophagy and increased mitochondrial function. Reduced activation and/or levels of several key energy and metabolic regulators (AKT, mTOR, ME1, G6PD, AMPK and LKB) were subsequently confirmed in mimic treated HuH-7 cells. These effects were associated with reduced NADPH to NADP+ ratio in HuH-7 cells. LKB1 was validated as a direct target of miR1908-5p, the reintroduction of which was however insufficient to compensate for the impact of the miR1908-5p mimic on AMPK and ACC1. These findings implicate miR1908-5p in metabolic and energy regulation in hepatocyte models via multiple, independent, pathways.
Collapse
|
43
|
Zhou B, Zhang Y, Li S, Wu L, Fejes-Toth G, Naray-Fejes-Toth A, Soukas AA. Serum- and glucocorticoid-induced kinase drives hepatic insulin resistance by directly inhibiting AMP-activated protein kinase. Cell Rep 2021; 37:109785. [PMID: 34610303 PMCID: PMC8576737 DOI: 10.1016/j.celrep.2021.109785] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 08/05/2021] [Accepted: 09/10/2021] [Indexed: 12/01/2022] Open
Abstract
A hallmark of type 2 diabetes (T2D) is hepatic resistance to insulin's glucose-lowering effects. The serum- and glucocorticoid-regulated family of protein kinases (SGK) is activated downstream of mechanistic target of rapamycin complex 2 (mTORC2) in response to insulin in parallel to AKT. Surprisingly, despite an identical substrate recognition motif to AKT, which drives insulin sensitivity, pathological accumulation of SGK1 drives insulin resistance. Liver-specific Sgk1-knockout (Sgk1Lko) mice display improved glucose tolerance and insulin sensitivity and are protected from hepatic steatosis when fed a high-fat diet. Sgk1 promotes insulin resistance by inactivating AMP-activated protein kinase (AMPK) via phosphorylation on inhibitory site AMPKαSer485/491. We demonstrate that SGK1 is dominant among SGK family kinases in regulation of insulin sensitivity, as Sgk1, Sgk2, and Sgk3 triple-knockout mice have similar increases in hepatic insulin sensitivity. In aggregate, these data suggest that targeting hepatic SGK1 may have therapeutic potential in T2D.
Collapse
Affiliation(s)
- Ben Zhou
- Department of Medicine, Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA; CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Yuyao Zhang
- Department of Medicine, Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Sainan Li
- Department of Medicine, Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Lianfeng Wu
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, School of Life Sciences, Westlake University, Hangzhou, 310024, China
| | - Geza Fejes-Toth
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA
| | - Aniko Naray-Fejes-Toth
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, 03755, USA
| | - Alexander A Soukas
- Department of Medicine, Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Medicine, Harvard Medical School, Boston, MA 02114, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA.
| |
Collapse
|
44
|
Vásquez-Trincado C, Patel M, Sivaramakrishnan A, Bekeová C, Anderson-Pullinger L, Wang N, Tang HY, Seifert EL. Adaptation of the heart to Frataxin depletion: Evidence that integrated stress response can predominate over mTORC1 activation. Hum Mol Genet 2021; 33:ddab216. [PMID: 34550363 PMCID: PMC11000666 DOI: 10.1093/hmg/ddab216] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 11/12/2022] Open
Abstract
Friedreich's ataxia (FRDA) is an inherited disorder caused by depletion of frataxin (FXN), a mitochondrial protein required for iron-sulfur cluster (ISC) biogenesis. Cardiac dysfunction is the main cause of death. Yet pathogenesis, and, more generally, how the heart adapts to FXN loss, remain poorly understood, though are expected to be linked to an energy deficit. We modified a transgenic (TG) mouse model of inducible FXN depletion that permits phenotypic evaluation of the heart at different FXN levels, and focused on substrate-specific bioenergetics and stress signaling. When FXN protein in the TG heart was 17% of normal, bioenergetics and signaling were not different from control. When, 8 weeks later, FXN was ~ 97% depleted in the heart, TG heart mass and cardiomyocyte cross-sectional area were less, without evidence of fibrosis or apoptosis. mTORC1 signaling was activated, as was the integrated stress response, evidenced by greater phosphorylation of eIF2α relative to total eIF2α, and decreased protein translation. We interpret these results to suggest that, in TG hearts, an anabolic stimulus was constrained by eIF2α phosphorylation. Cardiac contractility was maintained in the 97%-FXN-depleted hearts, possibly contributed by an unexpected preservation of β-oxidation, though pyruvate oxidation was lower. Bioenergetics alterations were matched by changes in the mitochondrial proteome, including a non-uniform decrease in abundance of ISC-containing proteins. Altogether, these findings suggest that the FXN depleted heart can suppress a major ATP demanding process such as protein translation, which, together with some preservation of β-oxidation, could be adaptive, at least in the short term.
Collapse
Affiliation(s)
- César Vásquez-Trincado
- MitoCare Center for Mitochondrial Imaging Research and Diagnostics, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Monika Patel
- MitoCare Center for Mitochondrial Imaging Research and Diagnostics, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Aishwarya Sivaramakrishnan
- MitoCare Center for Mitochondrial Imaging Research and Diagnostics, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Carmen Bekeová
- MitoCare Center for Mitochondrial Imaging Research and Diagnostics, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Lauren Anderson-Pullinger
- MitoCare Center for Mitochondrial Imaging Research and Diagnostics, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Nadan Wang
- Center for Translational Medicine, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, United States
| | - Hsin-Yao Tang
- Proteomics and Metabolomics Facility, The Wistar Institute, Philadelphia, PA 19104, United States
| | - Erin L Seifert
- MitoCare Center for Mitochondrial Imaging Research and Diagnostics, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, United States
| |
Collapse
|
45
|
Chen YC, Chien CY, Hsu CC, Lee CH, Chou YT, Shiah SG, Liu SY, Yen CY, Hsieh ACT, Wabitsch M, Shieh YS. Obesity-associated leptin promotes chemoresistance in colorectal cancer through YAP-dependent AXL upregulation. Am J Cancer Res 2021; 11:4220-4240. [PMID: 34659884 PMCID: PMC8493400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 08/09/2021] [Indexed: 06/13/2023] Open
Abstract
Obesity results from an imbalance between caloric intake and energy expenditure, and it is highly associated with colorectal carcinogenesis and therapeutic resistance in patients with colorectal cancer (CRC). Dysregulation of adipokine production in obesity has been reported to cause malignant behaviors in CRC. Leptin, which is the principal hormone secreted by adipocytes and an obesity-associated adipokine, is significantly overexpressed in CRC tissues. However, the effect of leptin on chemoresistance in CRC is unclear. Therefore, the aim of this study was to clarify the role of leptin and the underlying mechanisms in mediating 5-fluorouracil (5-FU) resistance in CRC. We used palmitate to artificially generate obese adipocytes. As expected, lipid accumulation was significantly increased in obese adipocytes. We demonstrated that CRC cells incubated with conditioned media (CM) harvested from obese adipocytes were associated with increased resistance to 5-FU. Notably, this increase in resistance to 5-FU was through the elevated production and secretion of leptin. Leptin could further stimulate the expression of AXL and activate its downstream signaling molecule, PLCγ, thereby resulting in an increased expression of p-glycoprotein (P-gp) in CRC cells. Mechanistically, leptin induced AXL expression via the inhibition of AMPK and subsequent increase in YAP activation and nuclear translocation. In addition, nuclear YAP interacted with TEAD and promoted the occupancy of TEAD on the AXL promoter, thereby stimulating AXL promoter activity after leptin treatment. Furthermore, leptin neutralization rescued the sensitivity of CRC tumors to 5-FU in mice fed on a high-fat diet (HFD). These results indicated that leptin mediated 5-FU resistance through YAP-dependent AXL overexpression in CRC.
Collapse
Affiliation(s)
- Ying-Chen Chen
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica and Graduate Institute of Life Science, National Defense Medical CenterTaipei 114201, Taiwan
| | - Chu-Yen Chien
- Graduate Institute of Medical Sciences, National Defense Medical CenterTaipei 114201, Taiwan
| | - Chia-Chen Hsu
- Graduate Institute of Medical Sciences, National Defense Medical CenterTaipei 114201, Taiwan
| | - Chien-Hsing Lee
- Department and Graduate Institute of Biochemistry, National Defense Medical CenterTaipei 114201, Taiwan
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical CenterTaipei 114201, Taiwan
| | - Yu-Ting Chou
- Institute of Biotechnology, National Tsing Hua UniversityHsinchu 300044, Taiwan
| | - Shine-Gwo Shiah
- National Institute of Cancer Research, National Health Research InstitutesMiaoli 350, Taiwan
| | - Shyun-Yeu Liu
- Department of Oral and Maxillofacial Surgery, Chi Mei Medical CenterTainan 71004, Taiwan
| | - Ching-Yu Yen
- Department of Oral and Maxillofacial Surgery, Chi Mei Medical CenterTainan 71004, Taiwan
| | | | - Martin Wabitsch
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Endocrinology and Diabetes, Centre for Hormonal Disorders in Children and Adolescents, Ulm University HospitalUlm 89081, Germany
| | - Yi-Shing Shieh
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica and Graduate Institute of Life Science, National Defense Medical CenterTaipei 114201, Taiwan
- Department and Graduate Institute of Biochemistry, National Defense Medical CenterTaipei 114201, Taiwan
- Department of Dentistry, Tri-Service General Hospital, National Defense Medical CenterTaipei 114201, Taiwan
| |
Collapse
|
46
|
Duan L, Calhoun S, Shim D, Perez RE, Blatter LA, Maki CG. Fatty acid oxidation and autophagy promote endoxifen resistance and counter the effect of AKT inhibition in ER-positive breast cancer cells. J Mol Cell Biol 2021; 13:433-444. [PMID: 33755174 PMCID: PMC8436705 DOI: 10.1093/jmcb/mjab018] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 02/22/2021] [Accepted: 02/24/2021] [Indexed: 11/24/2022] Open
Abstract
Tamoxifen (TAM) is the first-line endocrine therapy for estrogen receptor-positive (ER+) breast cancer (BC). However, acquired resistance occurs in ∼50% cases. Meanwhile, although the PI3K/AKT/mTOR pathway is a viable target for treatment of endocrine therapy-refractory patients, complex signaling feedback loops exist, which can counter the effectiveness of inhibitors of this pathway. Here, we analyzed signaling pathways and metabolism in ER+ MCF7 BC cell line and their TAM-resistant derivatives that are co-resistant to endoxifen using immunoblotting, quantitative polymerase chain reaction, and the Agilent Seahorse XF Analyzer. We found that activation of AKT and the energy-sensing kinase AMPK was increased in TAM and endoxifen-resistant cells. Furthermore, ERRα/PGC-1β and their target genes MCAD and CPT-1 were increased and regulated by AMPK, which coincided with increased fatty acid oxidation (FAO) and autophagy in TAM-resistant cells. Inhibition of AKT feedback-activates AMPK and ERRα/PGC-1β-MCAD/CPT-1 with a consequent increase in FAO and autophagy that counters the therapeutic effect of endoxifen and AKT inhibitors. Therefore, our results indicate increased activation of AKT and AMPK with metabolic reprogramming and increased autophagy in TAM-resistant cells. Simultaneous inhibition of AKT and FAO/autophagy is necessary to fully sensitize resistant cells to endoxifen.
Collapse
Affiliation(s)
- Lei Duan
- Department of Cell and Molecular Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Sarah Calhoun
- Department of Cell and Molecular Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Daeun Shim
- Department of Cell and Molecular Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Ricardo E Perez
- Department of Cell and Molecular Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Lothar A Blatter
- Department of Molecular Biophysics and Physiology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Carl G Maki
- Department of Cell and Molecular Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
47
|
Gandhi N, Oturkar CC, Das GM. Estrogen Receptor-Alpha and p53 Status as Regulators of AMPK and mTOR in Luminal Breast Cancer. Cancers (Basel) 2021; 13:cancers13143612. [PMID: 34298826 PMCID: PMC8306694 DOI: 10.3390/cancers13143612] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/10/2021] [Accepted: 07/14/2021] [Indexed: 12/12/2022] Open
Abstract
Luminal breast cancer (LBC) driven by dysregulated estrogen receptor-alpha (ERα) signaling accounts for 70% of the breast cancer cases diagnosed. Although endocrine therapy (ET) is effective against LBC, about one-third of these patients fail to respond to therapy owing to acquired or inherent resistance mechanisms. Aberrant signaling via ERα, oncogenes, growth factor receptors, and mutations in tumor suppressors such as p53 impinge on downstream regulators such as AMPK and mTOR. While both AMPK and mTOR have been reported to play important roles in determining sensitivity of LBC to ET, how the ERα-p53 crosstalk impinges on regulation of AMPK and mTOR, thereby influencing therapeutic efficacy remains unknown. Here, we have addressed this important issue using isogenic breast cancer cell lines, siRNA-mediated RNA knockdown, and different modes of drug treatments. Interaction of p53 with ERα and AMPK was determined by in situ proximity ligation assay (PLA), and endogenous gene transcripts were analyzed by quantitative real-time polymerase chain reaction (qRT-PCR). Further, the effect of concurrent and sequential administration of Fulvestrant-Everolimus combination on colony formation was determined. The studies showed that in cells expressing wild type p53, as well as in cells devoid of p53, ERα represses AMPK, whereas in cells harboring mutant p53, repression of AMPK is sustained even in the absence of ERα. AMPK is a major negative regulator of mTOR, and to our knowledge, this is the first study on the contribution of AMPK-dependent regulation of mTOR by ERα. Furthermore, the studies revealed that independent of the p53 mutation status, combination of Fulvestrant and Everolimus may be a viable first line therapeutic strategy for potentially delaying resistance of ERα+/HER2- LBC to ET.
Collapse
|
48
|
Lee HJ, Donati A, Feliers D, Sun Y, Ding Y, Madesh M, Salmon AB, Ikeno Y, Ross C, O'Connor CL, Ju W, Bitzer M, Chen Y, Choudhury GG, Singh BB, Sharma K, Kasinath BS. Chloride channel accessory 1 integrates chloride channel activity and mTORC1 in aging-related kidney injury. Aging Cell 2021; 20:e13407. [PMID: 34118180 PMCID: PMC8282273 DOI: 10.1111/acel.13407] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 03/30/2021] [Accepted: 05/08/2021] [Indexed: 12/19/2022] Open
Abstract
The mechanism of kidney injury in aging are not well understood. In order to identify hitherto unknown pathways of aging‐related kidney injury, we performed RNA‐Seq on kidney extracts of young and aged mice. Expression of chloride (Cl) channel accessory 1 (CLCA1) mRNA and protein was increased in the kidneys of aged mice. Immunostaining showed a marked increase in CLCLA1 expression in the proximal tubules of the kidney from aged mice. Increased kidney CLCA1 gene expression also correlated with aging in marmosets and in a human cohort. In aging mice, increased renal cortical CLCA1 content was associated with hydrogen sulfide (H2S) deficiency, which was ameliorated by administering sodium hydrosulfide (NaHS), a source of H2S. In order to study whether increased CLCA1 expression leads to injury phenotype and the mechanisms involved, stable transfection of proximal tubule epithelial cells overexpressing human CLCA1 (hCLCA1) was performed. Overexpression of hCLCA1 augmented Cl− current via the Ca++‐dependent Cl− channel TMEM16A (anoctamin‐1) by patch‐clamp studies. hCLCA1 overexpression also increased the expression of fibronectin, a matrix protein, and induced the senescence‐associated secretory phenotype (SASP). Mechanistic studies underlying these changes showed that hCLCA1 overexpression leads to inhibition of AMPK activity and stimulation of mTORC1 as cellular signaling determinants of injury. Both TMEM16A inhibitor and NaHS reversed these signaling events and prevented changes in fibronectin and SASP. We conclude that CLCA1‐TMEM16A‐Cl− current pathway is a novel mediator of kidney injury in aging that is regulated by endogenous H2S.
Collapse
Affiliation(s)
- Hak Joo Lee
- Department of Medicine Center for Renal Precision Medicine University of Texas Health San Antonio TX USA
| | - Andrew Donati
- Department of Medicine Center for Renal Precision Medicine University of Texas Health San Antonio TX USA
| | - Denis Feliers
- Department of Medicine Center for Renal Precision Medicine University of Texas Health San Antonio TX USA
| | - Yuyang Sun
- Department of Periodontics University of Texas Health San Antonio TX USA
| | - Yanli Ding
- Department of Pathology University of Texas Health San Antonio TX USA
| | - Muniswamy Madesh
- Department of Medicine Center for Renal Precision Medicine University of Texas Health San Antonio TX USA
| | - Adam B. Salmon
- Department of Molecular Medicine University of Texas Health San Antonio TX USA
- Barshop Institute for Longevity and Aging Studies University of Texas Health San Antonio TX USA
- South Texas Veterans Health Care System San Antonio TX USA
- Geriatric Research Education & Clinical Center South Texas Veterans Health Care System San Antonio TX USA
| | - Yuji Ikeno
- Department of Pathology University of Texas Health San Antonio TX USA
- Department of Molecular Medicine University of Texas Health San Antonio TX USA
- South Texas Veterans Health Care System San Antonio TX USA
| | - Corinna Ross
- Texas Biomedical Research Institute Southwest National Primate Research Center San Antonio TX USA
- Department of Science and Mathematics Texas A&M University San Antonio San Antonio TX USA
| | | | - Wenjun Ju
- Department of Internal Medicine University of Michigan Ann Arbor MI USA
| | - Markus Bitzer
- Department of Internal Medicine University of Michigan Ann Arbor MI USA
| | - Yidong Chen
- Department of Population Health Sciences University of Texas Health San Antonio TX USA
- Greehey Children's Cancer Research Institute University of Texas Health San Antonio TX USA
| | - Goutam Ghosh Choudhury
- Department of Medicine Center for Renal Precision Medicine University of Texas Health San Antonio TX USA
- South Texas Veterans Health Care System San Antonio TX USA
- Geriatric Research Education & Clinical Center South Texas Veterans Health Care System San Antonio TX USA
| | - Brij B. Singh
- Department of Periodontics University of Texas Health San Antonio TX USA
| | - Kumar Sharma
- Department of Medicine Center for Renal Precision Medicine University of Texas Health San Antonio TX USA
- South Texas Veterans Health Care System San Antonio TX USA
| | - Balakuntalam S. Kasinath
- Department of Medicine Center for Renal Precision Medicine University of Texas Health San Antonio TX USA
- Barshop Institute for Longevity and Aging Studies University of Texas Health San Antonio TX USA
- South Texas Veterans Health Care System San Antonio TX USA
- Geriatric Research Education & Clinical Center South Texas Veterans Health Care System San Antonio TX USA
| |
Collapse
|
49
|
Mygland L, Brinch SA, Strand MF, Olsen PA, Aizenshtadt A, Lund K, Solberg NT, Lycke M, Thorvaldsen TE, Espada S, Misaghian D, Page CM, Agafonov O, Nygård S, Chi NW, Lin E, Tan J, Yu Y, Costa M, Krauss S, Waaler J. Identification of response signatures for tankyrase inhibitor treatment in tumor cell lines. iScience 2021; 24:102807. [PMID: 34337362 PMCID: PMC8313754 DOI: 10.1016/j.isci.2021.102807] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 05/26/2021] [Accepted: 06/28/2021] [Indexed: 02/06/2023] Open
Abstract
Small-molecule tankyrase 1 and tankyrase 2 (TNKS1/2) inhibitors are effective antitumor agents in selected tumor cell lines and mouse models. Here, we characterized the response signatures and the in-depth mechanisms for the antiproliferative effect of tankyrase inhibition (TNKSi). The TNKS1/2-specific inhibitor G007-LK was used to screen 537 human tumor cell lines and a panel of particularly TNKSi-sensitive tumor cell lines was identified. Transcriptome, proteome, and bioinformatic analyses revealed the overall TNKSi-induced response signatures in the selected panel. TNKSi-mediated inhibition of wingless-type mammary tumor virus integration site/β-catenin, yes-associated protein 1 (YAP), and phosphatidylinositol-4,5-bisphosphate 3-kinase/AKT signaling was validated and correlated with lost expression of the key oncogene MYC and impaired cell growth. Moreover, we show that TNKSi induces accumulation of TNKS1/2-containing β-catenin degradasomes functioning as core complexes interacting with YAP and angiomotin proteins during attenuation of YAP signaling. These findings provide a contextual and mechanistic framework for using TNKSi in anticancer treatment that warrants further comprehensive preclinical and clinical evaluations. TNKSi-responding tumor cell lines were identified TNKSi targets WNT/β-catenin, YAP, and PI3K/AKT signaling Reduced MYC expression leads to impaired tumor cell growth
Collapse
Affiliation(s)
- Line Mygland
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, P.O. Box 4950 Nydalen, Oslo 0424, Norway.,Hybrid Technology Hub - Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, P.O. Box 1110 Blindern, 0317 Oslo, Norway
| | - Shoshy Alam Brinch
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, P.O. Box 4950 Nydalen, Oslo 0424, Norway.,Hybrid Technology Hub - Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, P.O. Box 1110 Blindern, 0317 Oslo, Norway
| | - Martin Frank Strand
- School of Health Sciences, Kristiania University College, P.O. Box 1190 Sentrum, 0107 Oslo, Norway
| | - Petter Angell Olsen
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, P.O. Box 4950 Nydalen, Oslo 0424, Norway.,Hybrid Technology Hub - Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, P.O. Box 1110 Blindern, 0317 Oslo, Norway
| | - Aleksandra Aizenshtadt
- Hybrid Technology Hub - Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, P.O. Box 1110 Blindern, 0317 Oslo, Norway
| | - Kaja Lund
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, P.O. Box 4950 Nydalen, Oslo 0424, Norway
| | - Nina Therese Solberg
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, P.O. Box 4950 Nydalen, Oslo 0424, Norway
| | - Max Lycke
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, P.O. Box 4950 Nydalen, Oslo 0424, Norway
| | - Tor Espen Thorvaldsen
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0379 Oslo, Norway
| | - Sandra Espada
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, P.O. Box 4950 Nydalen, Oslo 0424, Norway.,Hybrid Technology Hub - Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, P.O. Box 1110 Blindern, 0317 Oslo, Norway
| | - Dorna Misaghian
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, P.O. Box 4950 Nydalen, Oslo 0424, Norway
| | - Christian M Page
- Center for Fertility and Health, Norwegian Institute of Public Health, P.O. Box 222 Skøyen, 0213 Oslo, Norway.,Oslo Centre for Biostatistics and Epidemiology, Oslo University Hospital, P.O. Box 4950 Nydalen, 0424 Oslo, Norway
| | - Oleg Agafonov
- Bioinformatics Core Facility, Department of Core Facilities, Institute for Cancer Research, Oslo University Hospital, Ullernchausseen 70, 0379 Oslo, Norway
| | - Ståle Nygård
- Department of Informatics, University of Oslo, P.O. box 080 Blindern, 0316 Oslo, Norway
| | - Nai-Wen Chi
- Endocrine Service, VA San Diego Healthcare System, 3350 La Jolla Village Dr., San Diego, CA 92161, USA
| | - Eva Lin
- Department of Discovery Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Jenille Tan
- Department of Discovery Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Yihong Yu
- Department of Discovery Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Mike Costa
- Department of Discovery Oncology, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | - Stefan Krauss
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, P.O. Box 4950 Nydalen, Oslo 0424, Norway.,Hybrid Technology Hub - Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, P.O. Box 1110 Blindern, 0317 Oslo, Norway
| | - Jo Waaler
- Department of Immunology and Transfusion Medicine, Oslo University Hospital, P.O. Box 4950 Nydalen, Oslo 0424, Norway.,Hybrid Technology Hub - Centre of Excellence, Institute of Basic Medical Sciences, University of Oslo, P.O. Box 1110 Blindern, 0317 Oslo, Norway
| |
Collapse
|
50
|
Dai X, Bu X, Gao Y, Guo J, Hu J, Jiang C, Zhang Z, Xu K, Duan J, He S, Zhang J, Wan L, Liu T, Zhou X, Hung MC, Freeman GJ, Wei W. Energy status dictates PD-L1 protein abundance and anti-tumor immunity to enable checkpoint blockade. Mol Cell 2021; 81:2317-2331.e6. [PMID: 33909988 PMCID: PMC8178223 DOI: 10.1016/j.molcel.2021.03.037] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 10/26/2020] [Accepted: 03/22/2021] [Indexed: 02/06/2023]
Abstract
Aberrant energy status contributes to multiple metabolic diseases, including obesity, diabetes, and cancer, but the underlying mechanism remains elusive. Here, we report that ketogenic-diet-induced changes in energy status enhance the efficacy of anti-CTLA-4 immunotherapy by decreasing PD-L1 protein levels and increasing expression of type-I interferon (IFN) and antigen presentation genes. Mechanistically, energy deprivation activates AMP-activated protein kinase (AMPK), which in turn, phosphorylates PD-L1 on Ser283, thereby disrupting its interaction with CMTM4 and subsequently triggering PD-L1 degradation. In addition, AMPK phosphorylates EZH2, which disrupts PRC2 function, leading to enhanced IFNs and antigen presentation gene expression. Through these mechanisms, AMPK agonists or ketogenic diets enhance the efficacy of anti-CTLA-4 immunotherapy and improve the overall survival rate in syngeneic mouse tumor models. Our findings reveal a pivotal role for AMPK in regulating the immune response to immune-checkpoint blockade and advocate for combining ketogenic diets or AMPK agonists with anti-CTLA4 immunotherapy to combat cancer.
Collapse
Affiliation(s)
- Xiaoming Dai
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Xia Bu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Yang Gao
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA; Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Jianping Guo
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Jia Hu
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Cong Jiang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Zhao Zhang
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Kexin Xu
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Jinzhi Duan
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Shaohui He
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Jinfang Zhang
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | - Lixin Wan
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Tianjie Liu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiaobo Zhou
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences and Center for Molecular Medicine, China Medical University, Taichung 404, Taiwan
| | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|