51
|
Schaf J, Shinhmar S, Zeng Q, Pardo OE, Beesley P, Syed N, Williams RSB. Enhanced Sestrin expression through Tanshinone 2A treatment improves PI3K-dependent inhibition of glioma growth. Cell Death Discov 2023; 9:172. [PMID: 37202382 DOI: 10.1038/s41420-023-01462-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 04/28/2023] [Accepted: 05/03/2023] [Indexed: 05/20/2023] Open
Abstract
Glioblastomas are a highly aggressive cancer type which respond poorly to current pharmaceutical treatments, thus novel therapeutic approaches need to be investigated. One such approach involves the use of the bioactive natural product Tanshinone IIA (T2A) derived from the Chinese herb Danshen, where mechanistic insight for this anti-cancer agent is needed to validate its use. Here, we employ a tractable model system, Dictyostelium discoideum, to provide this insight. T2A potently inhibits cellular proliferation of Dictyostelium, suggesting molecular targets in this model. We show that T2A rapidly reduces phosphoinositide 3 kinase (PI3K) and protein kinase B (PKB) activity, but surprisingly, the downstream complex mechanistic target of rapamycin complex 1 (mTORC1) is only inhibited following chronic treatment. Investigating regulators of mTORC1, including PKB, tuberous sclerosis complex (TSC), and AMP-activated protein kinase (AMPK), suggests these enzymes were not responsible for this effect, implicating an additional molecular mechanism of T2A. We identify this mechanism as the increased expression of sestrin, a negative regulator of mTORC1. We further show that combinatory treatment using a PI3K inhibitor and T2A gives rise to a synergistic inhibition of cell proliferation. We then translate our findings to human and mouse-derived glioblastoma cell lines, where both a PI3K inhibitor (Paxalisib) and T2A reduces glioblastoma proliferation in monolayer cultures and in spheroid expansion, with combinatory treatment significantly enhancing this effect. Thus, we propose a new approach for cancer treatment, including glioblastomas, through combinatory treatment with PI3K inhibitors and T2A.
Collapse
Affiliation(s)
- Judith Schaf
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham, TW20 0EX, UK
| | - Sonia Shinhmar
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham, TW20 0EX, UK
| | - Qingyu Zeng
- John Fulcher Neuro-Oncology Laboratory, Imperial College London, Hammersmith Hospital, London, UK
| | - Olivier E Pardo
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Philip Beesley
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham, TW20 0EX, UK
| | - Nelofer Syed
- John Fulcher Neuro-Oncology Laboratory, Imperial College London, Hammersmith Hospital, London, UK
| | - Robin S B Williams
- Centre for Biomedical Sciences, School of Biological Sciences, Royal Holloway University of London, Egham, TW20 0EX, UK.
| |
Collapse
|
52
|
Hwang I, Kim M. Muscular Sestrins: Roles in Exercise Physiology and Stress Resistance. Biomolecules 2023; 13:722. [PMID: 37238592 PMCID: PMC10216764 DOI: 10.3390/biom13050722] [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: 03/21/2023] [Revised: 04/15/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023] Open
Abstract
Sestrins are a family of stress-inducible proteins that are critical for stress adaptation and the maintenance of metabolic homeostasis. High expression of Sestrins is observed in skeletal and cardiac muscle tissues, suggesting their significance in the physiological homeostasis of these organs. Furthermore, expression of Sestrins is dynamically controlled in the tissues, based on the level of physical activity and the presence or absence of stress insults. Genetic studies in model organisms have shown that muscular Sestrin expression is critical for metabolic homeostasis, exercise adaptation, stress resistance, and repair and may mediate the beneficial effects of some available therapeutics. The current minireview summarizes and discusses recent findings that shed light on the role of Sestrins in regulating muscle physiology and homeostasis.
Collapse
Affiliation(s)
| | - Myungjin Kim
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
53
|
Seike M, Asahara SI, Inoue H, Kudo M, Kanno A, Yokoi A, Suzuki H, Kimura-Koyanagi M, Kido Y, Ogawa W. l-Asparaginase regulates mTORC1 activity via a TSC2-dependent pathway in pancreatic beta cells. Biochem Biophys Res Commun 2023; 652:121-130. [PMID: 36842323 DOI: 10.1016/j.bbrc.2023.02.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023]
Abstract
Eif2ak4, a susceptibility gene for type 2 diabetes, encodes GCN2, a molecule activated by amino acid deficiency. Mutations or deletions in GCN2 in pancreatic β-cells increase mTORC1 activity by decreasing Sestrin2 expression in a TSC2-independent manner. In this study, we searched for molecules downstream of GCN2 that suppress mTORC1 activity in a TSC2-dependent manner. To do so, we used a pull-down assay to identify molecules that competitively inhibit the binding of the T1462 phosphorylation site of TSC2 to 14-3-3. l-asparaginase was identified. Although l-asparaginase is frequently used as an anticancer drug for acute lymphoblastic leukemia, little is known about endogenous l-asparaginase. l-Asparaginase, which is expressed downstream of GCN2, was found to bind 14-3-3 and thereby to inhibit its binding to the T1462 phosphorylation site of TSC2 and contribute to TSC2 activation and mTORC1 inactivation upon TSC2 dephosphorylation. Further investigation of the regulation of mTORC1 activity in pancreatic β-cells by l-asparaginase should help to elucidate the mechanism of diabetes and insulin secretion failure during anticancer drug use.
Collapse
Affiliation(s)
- Masako Seike
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan.
| | - Shun-Ichiro Asahara
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan.
| | - Hiroyuki Inoue
- Division of Medical Chemistry, Department of Metabolism and Diseases, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, Hyogo, 654-0142, Japan.
| | - Michiyo Kudo
- Division of Medical Chemistry, Department of Metabolism and Diseases, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, Hyogo, 654-0142, Japan.
| | - Ayumi Kanno
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan.
| | - Aisha Yokoi
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan.
| | - Hirotaka Suzuki
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan.
| | - Maki Kimura-Koyanagi
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan.
| | - Yoshiaki Kido
- Division of Medical Chemistry, Department of Metabolism and Diseases, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, Hyogo, 654-0142, Japan.
| | - Wataru Ogawa
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan.
| |
Collapse
|
54
|
Papež M, Jiménez Lancho V, Eisenhut P, Motheramgari K, Borth N. SLAM-seq reveals early transcriptomic response mechanisms upon glutamine deprivation in Chinese hamster ovary cells. Biotechnol Bioeng 2023; 120:970-986. [PMID: 36575109 DOI: 10.1002/bit.28320] [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: 09/14/2022] [Revised: 11/30/2022] [Accepted: 12/25/2022] [Indexed: 12/29/2022]
Abstract
Mammalian cells frequently encounter subtle perturbations during recombinant protein production. Identifying the genetic factors that govern the cellular stress response can facilitate targeted genetic engineering to obtain production cell lines that demonstrate a higher stress tolerance. To simulate nutrient stress, Chinese hamster ovary (CHO) cells were transferred into a glutamine(Q)-free medium and transcriptional dynamics using thiol(SH)-linked alkylation for the metabolic sequencing of RNA (SLAM-seq) along with standard RNA-seq of stressed and unstressed cells were investigated. The SLAM-seq method allows differentiation between actively transcribed, nascent mRNA, and total (previously present) mRNA in the sample, adding an additional, time-resolved layer to classic RNA-sequencing. The cells tackle amino acid (AA) limitation by inducing the integrated stress response (ISR) signaling pathway, reflected in Atf4 overexpression in the early hours post Q deprivation, leading to subsequent activation of its targets, Asns, Atf3, Ddit3, Eif4ebp1, Gpt2, Herpud1, Slc7a1, Slc7a11, Slc38a2, Trib3, and Vegfa. The GCN2-eIF2α-ATF4 pathway is confirmed by a significant halt in transcription of translation-related genes at 24 h post Q deprivation. The downregulation of lipid synthesis indicates the inhibition of the mTOR pathway, further confirmed by overexpression of Sesn2. Furthermore, SLAM-seq detects short-lived transcription factors, such as Egr1, that would have been missed in standard experimental designs with RNA-seq. Our results describe the successful establishment of SLAM-seq in CHO cells and therefore facilitate its future use in other scenarios where dynamic transcriptome profiling in CHO cells is essential.
Collapse
Affiliation(s)
- Maja Papež
- Austrian Centre of Industrial Biotechnology (acib GmbH), Graz, Austria
| | | | - Peter Eisenhut
- Austrian Centre of Industrial Biotechnology (acib GmbH), Graz, Austria
| | | | - Nicole Borth
- Austrian Centre of Industrial Biotechnology (acib GmbH), Graz, Austria
- University of Natural Resources and Life Sciences (BOKU), Vienna, Austria
| |
Collapse
|
55
|
Roberson PA, Kincheloe GN, Welles JE, Xu D, Sam-Clarke M, MacLean PS, Lang CH, Jefferson LS, Kimball SR. Glucose-Induced Activation of mTORC1 is Associated with Hexokinase2 Binding to Sestrins in HEK293T Cells. J Nutr 2023; 153:988-998. [PMID: 37061344 PMCID: PMC10273196 DOI: 10.1016/j.tjnut.2022.11.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/25/2022] [Accepted: 11/23/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Sestrins (SESN1-3) act as proximal sensors in leucine-induced activation of the protein kinase mechanistic target of rapamycin (mTOR) in complex 1 (mTORC1), a key regulator of cell growth and metabolism. OBJECTIVE In the present study, the hypothesis that SESNs also mediate glucose-induced activation of mTORC1 was tested. METHODS Rats underwent overnight fasting, and in the morning, either saline or a glucose solution (4 g⋅kg-1 BW/10 mL⋅kg-1) was administered by oral gavage; mTORC1 activation in the tibialis anterior muscle was assessed. To further assess the mechanism through which glucose promotes mTORC1 activation, wild-type (WT) HEK293T and HEK293T cells lacking either all 3 SESNs (SESNTKO) or hexokinase 2 (HK2KO) were deprived of glucose, followed by glucose addback, and mTORC1 activation was assessed. In addition, glucose-induced changes in the association of the SESNs with components of the GAP activity toward the Rags (GATOR2) complex and with hexokinase 2 (HK2) were assessed by co-immunoprecipitation. One- and two-way ANOVA with Tukey post hoc comparisons were used. RESULTS Glucose administration to fasted rats promoted mTORC1 activation. Similarly, glucose readdition (GluAB) to the medium of glucose-deprived WT cells also promoted mTORC1 activation. By contrast, SESNTKO cells demonstrated attenuated mTORC1 activation following GluAB compared with WT cells. Interestingly, HK2 associated with all 3 SESNs in a glucose-dependent manner, i.e., HK2 abundance in SESN immunoprecipitates was high in cells deprived of glucose and decreased in response to GluAB. Moreover, similar to SESNTKO cells, the sensitivity of mTORC1 to GluAB was attenuated in HK2KO cells compared with WT cells. CONCLUSIONS The results of this study demonstrate that the SESNs and HK2 play important roles in glucose-induced mTORC1 activation in HEK293T cells. However, unlike leucine-induced mTORC1 activation, the effect was independent of the changes in SESN-GATOR2 interaction, and instead, it was associated with alterations in the association of SESNs with HK2.
Collapse
Affiliation(s)
- Paul A Roberson
- Penn State College of Medicine, Department of Cellular and Molecular Physiology, Hershey, PA, USA
| | - Gregory N Kincheloe
- Penn State College of Medicine, Department of Cellular and Molecular Physiology, Hershey, PA, USA
| | - Jaclyn E Welles
- Penn State College of Medicine, Department of Cellular and Molecular Physiology, Hershey, PA, USA
| | - Dandan Xu
- Penn State College of Medicine, Department of Cellular and Molecular Physiology, Hershey, PA, USA
| | - Mahalia Sam-Clarke
- Penn State College of Medicine, Department of Cellular and Molecular Physiology, Hershey, PA, USA
| | - Paul S MacLean
- Divisions of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Charles H Lang
- Penn State College of Medicine, Department of Cellular and Molecular Physiology, Hershey, PA, USA
| | - Leonard S Jefferson
- Penn State College of Medicine, Department of Cellular and Molecular Physiology, Hershey, PA, USA
| | - Scot R Kimball
- Penn State College of Medicine, Department of Cellular and Molecular Physiology, Hershey, PA, USA.
| |
Collapse
|
56
|
Regulation of mTORC1 by the Rag GTPases. Biochem Soc Trans 2023; 51:655-664. [PMID: 36929165 DOI: 10.1042/bst20210038] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 02/21/2023] [Accepted: 02/23/2023] [Indexed: 03/18/2023]
Abstract
The Rag GTPases are an evolutionarily conserved family that play a crucial role in amino acid sensing by the mammalian target of rapamycin complex 1 (mTORC1). mTORC1 is often referred to as the master regulator of cell growth. mTORC1 hyperactivation is observed in multiple diseases such as cancer, obesity, metabolic disorders, and neurodegeneration. The Rag GTPases sense amino acid levels and form heterodimers, where RagA or RagB binds to RagC or RagD, to recruit mTORC1 to the lysosome where it becomes activated. Here, we review amino acid signaling to mTORC1 through the Rag GTPases.
Collapse
|
57
|
Influence of mTOR-regulated anabolic pathways on equine skeletal muscle health. J Equine Vet Sci 2023; 124:104281. [PMID: 36905972 DOI: 10.1016/j.jevs.2023.104281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/04/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023]
Abstract
Skeletal muscle is a highly dynamic organ that is essential for locomotion as well as endocrine regulation in all populations of horses. However, despite the importance of adequate muscle development and maintenance, the mechanisms underlying protein anabolism in horses on different diets, exercise programs, and at different life stages remain obscure. Mechanistic target of rapamycin (mTOR) is a key component of the protein synthesis pathway and is regulated by biological factors such as insulin and amino acid availability. Providing a diet ample in vital amino acids, such as leucine and glutamine, is essential in activating sensory pathways that recruit mTOR to the lysosome and assist in the translation of important downstream targets. When the diet is well balanced, mitochondrial biogenesis and protein synthesis are activated in response to increased exercise bouts in the performing athlete. It is important to note that the mTOR kinase pathways are multi-faceted and very complex, with several binding partners and targets that lead to specific functions in protein turnover of the cell, and ultimately, the capacity to maintain or grow muscle mass. Further, these pathways are likely altered across the lifespan, with an emphasis of growth in young horses while decreases in musculature with aged horses appears to be attributable to degradation or other regulators of protein synthesis rather than alterations in the mTOR pathway. Previous work has begun to pinpoint ways in which the mTOR pathway is influenced by diet, exercise, and age; however, future research is warranted to quantify the functional outcomes related to changes in mTOR. Promisingly, this could provide direction on appropriate management techniques to support skeletal muscle growth and maximize athletic potential in differing equine populations.
Collapse
|
58
|
Sestrin2 as a Protective Shield against Cardiovascular Disease. Int J Mol Sci 2023; 24:ijms24054880. [PMID: 36902310 PMCID: PMC10003517 DOI: 10.3390/ijms24054880] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/22/2023] [Accepted: 01/23/2023] [Indexed: 03/06/2023] Open
Abstract
A timely and adequate response to stress is inherently present in each cell and is important for maintaining the proper functioning of the cell in changing intracellular and extracellular environments. Disruptions in the functioning or coordination of defense mechanisms against cellular stress can reduce the tolerance of cells to stress and lead to the development of various pathologies. Aging also reduces the effectiveness of these defense mechanisms and results in the accumulation of cellular lesions leading to senescence or death of the cells. Endothelial cells and cardiomyocytes are particularly exposed to changing environments. Pathologies related to metabolism and dynamics of caloric intake, hemodynamics, and oxygenation, such as diabetes, hypertension, and atherosclerosis, can overwhelm endothelial cells and cardiomyocytes with cellular stress to produce cardiovascular disease. The ability to cope with stress depends on the expression of endogenous stress-inducible molecules. Sestrin2 (SESN2) is an evolutionary conserved stress-inducible cytoprotective protein whose expression is increased in response to and defend against different types of cellular stress. SESN2 fights back the stress by increasing the supply of antioxidants, temporarily holding the stressful anabolic reactions, and increasing autophagy while maintaining the growth factor and insulin signaling. If the stress and the damage are beyond repair, SESN2 can serve as a safety valve to signal apoptosis. The expression of SESN2 decreases with age and its levels are associated with cardiovascular disease and many age-related pathologies. Maintaining sufficient levels or activity of SESN2 can in principle prevent the cardiovascular system from aging and disease.
Collapse
|
59
|
Lu C, Jiang Y, Xu W, Bao X. Sestrin2: multifaceted functions, molecular basis, and its implications in liver diseases. Cell Death Dis 2023; 14:160. [PMID: 36841824 PMCID: PMC9968343 DOI: 10.1038/s41419-023-05669-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/26/2023]
Abstract
Sestrin2 (SESN2), a highly conserved stress-responsive protein, can be triggered by various noxious stimuli, such as hypoxia, DNA damage, oxidative stress, endoplasmic reticulum (ER) stress, and inflammation. Multiple transcription factors regulate SESN2 expression, including hypoxia-inducible factor 1 (HIF-1), p53, nuclear factor E2-related factor 2 (Nrf2), activating transcription factor 4 (ATF4), ATF6, etc. Upon induction, SESN2 generally leads to activation of adenosine monophosphate-activated protein kinase (AMPK) and inhibition of mechanistic target of rapamycin complex 1 (mTORC1). To maintain cellular homeostasis, SESN2 and its downstream molecules directly scavenge reactive oxygen species or indirectly influence the expression patterns of key genes associated with redox, macroautophagy, mitophagy, ER stress, apoptosis, protein synthesis, and inflammation. In liver diseases including acute liver injury, fatty liver diseases, hepatic fibrosis, and hepatocellular carcinoma (HCC), SESN2 is abnormally expressed and correlated with disease progression. In NAFLD, SESN2 helps with postponing disease progression through balancing glycolipid metabolism and macroautophagy (lipophagy), and rectifying oxidative damage and ER stress. During hepatic fibrosis, SESN2 represses HSCs activation and intrahepatic inflammation, hindering the occurrence and progress of fibrogenesis. However, the role of SESN2 in HCC is controversial due to its paradoxical pro-autophagic and anti-apoptotic effects. In conclusion, this review summarizes the biological functions of SESN2 in hypoxia, genotoxic stress, oxidative stress, ER stress, and inflammation, and specifically emphasizes the pathophysiological significance of SESN2 in liver diseases, aiming to providing a comprehensive understanding for SESN2 as a potential therapeutic target in liver diseases.
Collapse
Affiliation(s)
- Chunfeng Lu
- grid.260483.b0000 0000 9530 8833School of Pharmacy, Nantong University, 226001 Nantong, Jiangsu China
| | - Yiming Jiang
- grid.260483.b0000 0000 9530 8833School of Pharmacy, Nantong University, 226001 Nantong, Jiangsu China
| | - Wenxuan Xu
- School of Life Science and Technology, China Pharmaceutical University, 210009, Nanjing, Jiangsu, China.
| | - Xiaofeng Bao
- School of Pharmacy, Nantong University, 226001, Nantong, Jiangsu, China.
| |
Collapse
|
60
|
Deleyto-Seldas N, Efeyan A. AMPK knocks at the gate of GATOR. Nat Metab 2023; 5:197-198. [PMID: 36732623 DOI: 10.1038/s42255-022-00729-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
| | - Alejo Efeyan
- Spanish National Cancer Research Center, Madrid, Spain.
| |
Collapse
|
61
|
Leucine ingestion promotes mTOR translocation to the periphery and enhances total and peripheral RPS6 phosphorylation in human skeletal muscle. Amino Acids 2023; 55:253-261. [PMID: 36474017 DOI: 10.1007/s00726-022-03221-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022]
Abstract
The activation of the mechanistic target of rapamycin complex 1 (mTORC1), a master regulator of protein synthesis, by anabolic stimuli (such as muscle contraction or essential amino acids) involves its translocation to the cell periphery. Leucine is generally considered the most anabolic of amino acids for its ability to independently modulate muscle protein synthesis. However, it is currently unknown if free leucine impacts region-specific mTORC1-mediated phosphorylation events and protein-protein interactions. In this clinical trial (NCT03952884; registered May 16, 2019), we used immunofluorescence methods to investigate the role of dietary leucine on the postprandial regulation of mTORC1 and ribosomal protein S6 (RPS6), an important downstream readout of mTORC1 activity. Eight young, healthy, recreationally active males (n = 8; 23 ± 3 yrs) ingested 2 g of leucine with vastus lateralis biopsies collected at baseline, 30, 60, and 180 min postprandial. Leucine promoted mTOR translocation to the periphery (~ 18-29%; p ≤ 0.012) and enhanced mTOR localization with the lysosome (~ 16%; both p = 0.049) at 30 and 60 min post-feeding. p-RPS6Ser240/244 staining intensity, a readout of mTORC1 activity, was significantly elevated at all postprandial timepoints in both the total fiber (~ 14-30%; p ≤ 0.032) and peripheral regions (~ 16-33%; p ≤ 0.014). Additionally, total and peripheral p-RPS6Ser240/244 staining intensity at 60 min was positively correlated (r = 0.74, p = 0.036; r = 0.80, p = 0.016, respectively) with rates of myofibrillar protein synthesis over 180 min. The ability of leucine to activate mTORC1 in peripheral regions favors an enhanced rate of MPS, as this is the intracellular space thought to be replete with the cellular machinery that facilitates this anabolic process.
Collapse
|
62
|
Rudar M, Suryawan A, Nguyen HV, Chacko SK, Vonderohe C, Stoll B, Burrin DG, Fiorotto ML, Davis TA. Regulation of skeletal muscle protein synthesis in the preterm pig by intermittent leucine pulses during continuous parenteral feeding. JPEN J Parenter Enteral Nutr 2023; 47:276-286. [PMID: 36128996 PMCID: PMC10621874 DOI: 10.1002/jpen.2450] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/31/2022] [Accepted: 09/15/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND Extrauterine growth restriction is a common complication of preterm birth. Leucine (Leu) is an agonist for the mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) signaling pathway that regulates translation initiation and protein synthesis in skeletal muscle. Previously, we showed that intermittent intravenous pulses of Leu to neonatal pigs born at term receiving continuous enteral nutrition increases muscle protein synthesis and lean mass accretion. Our objective was to determine the impact of intermittent intravenous pulses of Leu on muscle protein anabolism in preterm neonatal pigs administered continuous parenteral nutrition. METHODS Following preterm delivery (on day 105 of 115 gestation), pigs were fitted with umbilical artery and jugular vein catheters and provided continuous parenteral nutrition. Four days after birth, pigs were assigned to receive intermittent Leu (1600 µmol kg-1 h-1 ; n = 8) or alanine (1600 µmol kg-1 h-1 ; n = 8) parenteral pulses every 4 h for 28 h. Anabolic signaling and fractional protein synthesis were determined in skeletal muscle. RESULTS Leu concentration in the longissimus dorsi and gastrocnemius muscles increased in the leucine (LEU) group compared with the alanine (ALA) group (P < 0.0001). Despite the Leu-induced disruption of the Sestrin2·GATOR2 complex, which inhibits mTORC1 activation, in these muscles (P < 0.01), the abundance of mTOR·RagA and mTOR·RagC was not different. Accordingly, mTORC1-dependent activation of 4EBP1, S6K1, eIF4E·eIF4G, and protein synthesis were not different in any muscle between the LEU and ALA groups. CONCLUSION Intermittent pulses of Leu do not enhance muscle protein anabolism in preterm pigs supplied continuous parenteral nutrition.
Collapse
Affiliation(s)
- Marko Rudar
- Department of Animal Sciences, Auburn University, Auburn, Alabama, USA
| | - Agus Suryawan
- USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, Texas, USA
| | - Hanh V. Nguyen
- USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, Texas, USA
| | - Shaji K. Chacko
- USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, Texas, USA
| | - Caitlin Vonderohe
- USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, Texas, USA
| | - Barbara Stoll
- USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, Texas, USA
| | - Douglas G. Burrin
- USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, Texas, USA
| | - Marta L. Fiorotto
- USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, Texas, USA
| | - Teresa A. Davis
- USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
63
|
Rudar M, Naberhuis JK, Suryawan A, Nguyen HV, Fiorotto ML, Davis TA. Prematurity blunts protein synthesis in skeletal muscle independently of body weight in neonatal pigs. Pediatr Res 2023:10.1038/s41390-022-02456-3. [PMID: 36627358 DOI: 10.1038/s41390-022-02456-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 11/30/2022] [Accepted: 12/21/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND Postnatal growth failure in premature infants is associated with reduced lean mass accretion. Prematurity impairs the feeding-induced stimulation of translation initiation and protein synthesis in the skeletal muscle of neonatal pigs. The objective was to determine whether body weight independently contributes to the blunted postprandial protein synthesis. METHODS Preterm and term pigs that were either fasted or fed were stratified into quartiles according to birth weight to yield preterm and term groups of similar body weight; first and second quartiles of preterm pigs and third and fourth quartiles of term pigs were compared (preterm-fasted, n = 23; preterm-fed, n = 25; term-fasted, n = 21; term-fed, n = 21). Protein synthesis rates and mechanistic target of rapamycin complex 1 (mTORC1) activation in skeletal muscle were determined. RESULTS Relative body weight gain was lower in preterm compared to term pigs. Prematurity attenuated the feeding-induced increase in mTORC1 activation in longissimus dorsi and gastrocnemius muscles (P < 0.05). Protein synthesis in gastrocnemius (P < 0.01), but not in longissimus dorsi muscle, was blunted by preterm birth. CONCLUSION A lower capacity of skeletal muscle to respond adequately to feeding may contribute to reduced body weight gain and lean mass accretion in preterm infants. IMPACT This study has shown that the feeding-induced increase in protein synthesis of skeletal and cardiac muscle is blunted in neonatal pigs born preterm compared to pigs born at term independently of birth weight. These findings support the notion that preterm birth, and not low birth weight, impairs the capacity of skeletal and cardiac muscle to upregulate mechanistic target of rapamycin-dependent anabolic signaling pathways and protein synthesis in response to the postprandial increase in insulin and amino acids. These observations suggest that a blunted anabolic response to feeding contributes to reduced lean mass accretion and altered body composition in preterm infants.
Collapse
Affiliation(s)
- Marko Rudar
- Department of Animal Sciences, Auburn University, Auburn, AL, USA
| | - Jane K Naberhuis
- United States Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Agus Suryawan
- United States Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Hanh V Nguyen
- United States Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Marta L Fiorotto
- United States Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Teresa A Davis
- United States Department of Agriculture/Agricultural Research Service Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
64
|
Wang G, Zhang J, Wu S, Qin S, Zheng Y, Xia C, Geng H, Yao J, Deng L. The mechanistic target of rapamycin complex 1 pathway involved in hepatic gluconeogenesis through peroxisome-proliferator-activated receptor γ coactivator-1α. ANIMAL NUTRITION 2022; 11:121-131. [PMID: 36204284 PMCID: PMC9516411 DOI: 10.1016/j.aninu.2022.07.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 07/18/2022] [Accepted: 07/27/2022] [Indexed: 11/29/2022]
Abstract
Cattle can efficiently perform de novo generation of glucose through hepatic gluconeogenesis to meet post-weaning glucose demand. Substantial evidence points to cattle and non-ruminant animals being characterized by phylogenetic features in terms of their differing capacity for hepatic gluconeogenesis, a process that is highly efficient in cattle yet the underlying mechanism remains unclear. Here we used a variety of transcriptome data, as well as tissue and cell-based methods to uncover the mechanisms of high-efficiency hepatic gluconeogenesis in cattle. We showed that cattle can efficiently convert propionate into pyruvate, at least partly, via high expression of acyl-CoA synthetase short-chain family member 1 (ACSS1), propionyl-CoA carboxylase alpha chain (PCCA), methylmalonyl-CoA epimerase (MCEE), methylmalonyl-CoA mutase (MMUT), and succinate-CoA ligase (SUCLG2) genes in the liver (P < 0.01). Moreover, higher expression of the rate-limiting enzymes of gluconeogenesis, such as phosphoenolpyruvate carboxykinase (PCK) and fructose 1,6-bisphosphatase (FBP), ensures the efficient operation of hepatic gluconeogenesis in cattle (P < 0.01). Mechanistically, we found that cattle liver exhibits highly active mechanistic target of rapamycin complex 1 (mTORC1), and the expressions of PCCA, MMUT, SUCLG2, PCK, and FBP genes are regulated by the activation of mTORC1 (P < 0.001). Finally, our results showed that mTORC1 promotes hepatic gluconeogenesis in a peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) dependent manner. Collectively, our results not only revealed an important mechanism responsible for the quantitative differences in the efficiency of hepatic gluconeogenesis in cattle versus non-ruminant animals, but also established that mTORC1 is indeed involved in the regulation of hepatic gluconeogenesis through PGC-1α. These results provide a novel potential insight into promoting hepatic gluconeogenesis through activated mTORC1 in both ruminants and mammals.
Collapse
|
65
|
Tinline-Goodfellow CT, Lees MJ, Hodson N. The skeletal muscle fiber periphery: A nexus of mTOR-related anabolism. SPORTS MEDICINE AND HEALTH SCIENCE 2022; 5:10-19. [PMID: 36994172 PMCID: PMC10040390 DOI: 10.1016/j.smhs.2022.11.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/16/2022] [Accepted: 11/23/2022] [Indexed: 12/02/2022] Open
Abstract
Skeletal muscle anabolism is driven by numerous stimuli such as growth factors, nutrients (i.e., amino acids, glucose), and mechanical stress. These stimuli are integrated by the mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) signal transduction cascade. In recent years, work from our laboratory and elsewhere has sought to unravel the molecular mechanisms underpinning the mTOR-related activation of muscle protein synthesis (MPS), as well as the spatial regulation of these mechanisms within the skeletal muscle cell. These studies have suggested that the skeletal muscle fiber periphery is a region of central importance in anabolism (i.e., growth/MPS). Indeed, the fiber periphery is replete with the substrates, molecular machinery, and translational apparatus necessary to facilitate MPS. This review provides a summary of the mechanisms underpinning the mTOR-associated activation of MPS from cell, rodent, and human studies. It also presents an overview of the spatial regulation of mTORC1 in response to anabolic stimuli and outlines the factors that distinguish the periphery of the cell as a highly notable region of skeletal muscle for the induction of MPS. Future research should seek to further explore the nutrient-induced activation of mTORC1 at the periphery of skeletal muscle fibers.
Collapse
Affiliation(s)
| | - Matthew J. Lees
- Faculty of Kinesiology and Physical Education, University of Toronto, Canada
| | - Nathan Hodson
- Faculty of Kinesiology and Physical Education, University of Toronto, Canada
- Institute of Sport, Manchester Metropolitan University, Manchester, United Kingdom
- Department of Sport and Exercise Sciences, Manchester Metropolitan University, Manchester, United Kingdom
- Corresponding author. Faculty of Kinesiology and Physical Education, University of Toronto, Canada.
| |
Collapse
|
66
|
Selection-free endogenous tagging of cell lines by bicistronic co-expression of the surface antigen NGFR. MethodsX 2022; 9:101929. [DOI: 10.1016/j.mex.2022.101929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/13/2022] [Indexed: 11/19/2022] Open
|
67
|
Napolitano G, Di Malta C, Ballabio A. Non-canonical mTORC1 signaling at the lysosome. Trends Cell Biol 2022; 32:920-931. [PMID: 35654731 DOI: 10.1016/j.tcb.2022.04.012] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 01/21/2023]
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) signaling hub integrates multiple environmental cues to modulate cell growth and metabolism. Over the past decade considerable knowledge has been gained on the mechanisms modulating mTORC1 lysosomal recruitment and activation. However, whether and how mTORC1 is able to elicit selective responses to diverse signals has remained elusive until recently. We discuss emerging evidence for a 'non-canonical' mTORC1 signaling pathway that controls the function of microphthalmia/transcription factor E (MiT-TFE) transcription factors, key regulators of cell metabolism. This signaling pathway is mediated by a specific mechanism of substrate recruitment, and responds to stimuli that appear to converge on the lysosomal surface. We discuss the relevance of this pathway in physiological and disease conditions.
Collapse
Affiliation(s)
- Gennaro Napolitano
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli, Naples, Italy; Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, Via Pansini 5, 80131 Naples, Italy.
| | - Chiara Di Malta
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli, Naples, Italy; Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, Via Pansini 5, 80131 Naples, Italy.
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli, Naples, Italy; Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, Via Pansini 5, 80131 Naples, Italy; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA.
| |
Collapse
|
68
|
Doolittle WKL, Park S, Lee SG, Jeong S, Lee G, Ryu D, Schoonjans K, Auwerx J, Lee J, Jo YS. Non-genomic activation of the AKT-mTOR pathway by the mitochondrial stress response in thyroid cancer. Oncogene 2022; 41:4893-4904. [PMID: 36195659 DOI: 10.1038/s41388-022-02484-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 09/18/2022] [Accepted: 09/22/2022] [Indexed: 11/09/2022]
Abstract
Cancer progression is associated with metabolic reprogramming and causes significant intracellular stress; however, the mechanisms that link cellular stress and growth signalling are not fully understood. Here, we identified a mechanism that couples the mitochondrial stress response (MSR) with tumour progression. We demonstrated that the MSR is activated in a significant proportion of human thyroid cancers via the upregulation of heat shock protein D family members and the mitokine, growth differentiation factor 15. Our study also revealed that MSR triggered AKT/S6K signalling by activating mTORC2 via activating transcription factor 4/sestrin 2 activation whilst promoting leucine transporter and nutrient-induced mTORC1 activation. Importantly, we found that an increase in mtDNA played an essential role in MSR-induced mTOR activation and that crosstalk between MYC and MSR potentiated mTOR activation. Together, these findings suggest that the MSR could be a predictive marker for aggressive human thyroid cancer as well as a useful therapeutic target.
Collapse
Affiliation(s)
- Woo Kyung Lee Doolittle
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.,Department of Medicine, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Sunmi Park
- Department of Internal Medicine, Open NBI Convergence Technology Research Laboratory, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Seul Gi Lee
- Department of Surgery, Eulji University School of Medicine, Daejeon, 34824, South Korea
| | - Seonhyang Jeong
- Department of Internal Medicine, Open NBI Convergence Technology Research Laboratory, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Gibbeum Lee
- Department of Surgery, Open NBI Convergence Technology Research Laboratory, Yonsei Cancer Center, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, South Korea
| | - Dongryeol Ryu
- Laboratory of Molecular and Integrative Biology, Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, 16419, South Korea
| | - Kristina Schoonjans
- Laboratory of Metabolic Signaling, École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne, Lausanne, 1015, Switzerland
| | - Jandee Lee
- Department of Surgery, Open NBI Convergence Technology Research Laboratory, Yonsei Cancer Center, Severance Hospital, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| | - Young Suk Jo
- Department of Internal Medicine, Open NBI Convergence Technology Research Laboratory, Yonsei University College of Medicine, Seoul, 03722, South Korea.
| |
Collapse
|
69
|
Phillips SM, Lau KJ, D'Souza AC, Nunes EA. An umbrella review of systematic reviews of β-hydroxy-β-methyl butyrate supplementation in ageing and clinical practice. J Cachexia Sarcopenia Muscle 2022; 13:2265-2275. [PMID: 35818771 PMCID: PMC9530546 DOI: 10.1002/jcsm.13030] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 04/05/2022] [Accepted: 05/16/2022] [Indexed: 01/06/2023] Open
Abstract
The compound β-hydroxy-β-methyl butyrate (HMB) is proposed to increase or mitigate the loss of skeletal muscle and improve muscle function. We undertook a review of systematic reviews of HMB supplementation to promote gains or mitigate muscle loss in ageing and clinical populations. Following PRISMA guidelines, we searched for systematic reviews reporting the effect of HMB in our target populations. Dual-energy X-ray absorptiometry (DXA) measured lean soft-tissue mass (LSTM) was accepted as a proxy for muscle. We identified 15 systematic reviews that met our inclusion criteria, which were independently evaluated. The methodological quality of the reviews was assessed using A Measurement Tool to Assess Systematic Reviews (AMSTAR), and standardized effectiveness statements were generated. Five of 15 studies found some evidence that HMB augmented LSTM; the remaining 10 studies reported some evidence favouring no difference (6/10 studies) or insufficient evidence to determine an effect (4/10 studies). Of the 12 studies that evaluated strength, 4/12 found some evidence, 5/12 found some evidence of no effect with one article finding some evidence in favour of patients in peri-hospitalized and no evidence for those that are community-dwelling, 4/12 had insufficient evidence to determine an effect, and 1/12 had insufficient evidence. No]study reported a positive effect of HMB on physical function; however, 2/10 studies found some evidence favouring no effect, and 7/10 studies reported insufficient evidence to determine an effect. The effectiveness of HMB supplementation in augmenting LSTM was heterogeneous, with most reviews finding no effect or inconclusive evidence to determine an effect. Most reviews concluded that HMB supplementation did not affect strength outcome measures or studies were inconclusive. The current evidence is insufficient to assess the impact of HMB supplementation on functional outcome measures. Our analysis shows minor, inconsistent support for HMB as part of an oral nutritional supplement or as a stand-alone supplement (or combined with other amino acids) to increase or promote retention of LSTM, improve strength, and no evidence that it improves physical function in older persons or clinical populations.
Collapse
Affiliation(s)
| | | | | | - Everson A. Nunes
- McMaster UniversityHamiltonOntarioCanada
- Federal University of Santa CatarinaFlorianópolisBrazil
| |
Collapse
|
70
|
Singh B, Kumar Rai A. Loss of immune regulation in aged T-cells: A metabolic review to show lack of ability to control responses within the self. Hum Immunol 2022; 83:808-817. [DOI: 10.1016/j.humimm.2022.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 09/19/2022] [Accepted: 10/04/2022] [Indexed: 11/04/2022]
|
71
|
Chao X, Williams SN, Ding WX. Role of mechanistic target of rapamycin in autophagy and alcohol-associated liver disease. Am J Physiol Cell Physiol 2022; 323:C1100-C1111. [PMID: 36062877 PMCID: PMC9550572 DOI: 10.1152/ajpcell.00281.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/26/2022] [Accepted: 08/26/2022] [Indexed: 11/22/2022]
Abstract
Mechanistic target of rapamycin (mTOR) is a serine-threonine kinase and a cellular sensor for nutrient and energy status, which is critical in regulating cell metabolism and growth by governing the anabolic (protein and lipid synthesis) and catabolic process (autophagy). Alcohol-associated liver disease (ALD) is a major chronic liver disease worldwide that carries a huge financial burden. The spectrum of the pathogenesis of ALD includes steatosis, fibrosis, inflammation, ductular reaction, and eventual hepatocellular carcinoma, which is closely associated with metabolic changes that are regulated by mTOR. In this review, we summarized recent progress of alcohol consumption on the changes of mTORC1 and mTORC2 activity, the potential mechanisms and possible impact of the mTORC1 changes on autophagy in ALD. We also discussed the potential beneficial effects and limitations of targeting mTORC1 against ALD.
Collapse
Affiliation(s)
- Xiaojuan Chao
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas
| | - Sha Neisha Williams
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas
| | - Wen-Xing Ding
- Department of Pharmacology, Toxicology and Therapeutics, The University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
72
|
Melnik BC, Schmitz G. Milk Exosomal microRNAs: Postnatal Promoters of β Cell Proliferation but Potential Inducers of β Cell De-Differentiation in Adult Life. Int J Mol Sci 2022; 23:ijms231911503. [PMID: 36232796 PMCID: PMC9569743 DOI: 10.3390/ijms231911503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/15/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Pancreatic β cell expansion and functional maturation during the birth-to-weaning period is driven by epigenetic programs primarily triggered by growth factors, hormones, and nutrients provided by human milk. As shown recently, exosomes derived from various origins interact with β cells. This review elucidates the potential role of milk-derived exosomes (MEX) and their microRNAs (miRs) on pancreatic β cell programming during the postnatal period of lactation as well as during continuous cow milk exposure of adult humans to bovine MEX. Mechanistic evidence suggests that MEX miRs stimulate mTORC1/c-MYC-dependent postnatal β cell proliferation and glycolysis, but attenuate β cell differentiation, mitochondrial function, and insulin synthesis and secretion. MEX miR content is negatively affected by maternal obesity, gestational diabetes, psychological stress, caesarean delivery, and is completely absent in infant formula. Weaning-related disappearance of MEX miRs may be the critical event switching β cells from proliferation to TGF-β/AMPK-mediated cell differentiation, whereas continued exposure of adult humans to bovine MEX miRs via intake of pasteurized cow milk may reverse β cell differentiation, promoting β cell de-differentiation. Whereas MEX miR signaling supports postnatal β cell proliferation (diabetes prevention), persistent bovine MEX exposure after the lactation period may de-differentiate β cells back to the postnatal phenotype (diabetes induction).
Collapse
Affiliation(s)
- Bodo C. Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, D-49076 Osnabrück, Germany
- Correspondence: ; Tel.: +49-52-4198-8060
| | - Gerd Schmitz
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital of Regensburg, University of Regensburg, D-93053 Regensburg, Germany
| |
Collapse
|
73
|
Obeng KA, Mochizuki S, Koike S, Toyoshima Y, Sato Y, Yoshizawa F. Analysis of the Stimulative Effect of Tryptophan on Hepatic Protein Synthesis in Rats. J Nutr Sci Vitaminol (Tokyo) 2022; 68:312-319. [PMID: 36047103 DOI: 10.3177/jnsv.68.312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Tryptophan is an essential amino acid important as a protein building block, but it also serves as substrate for the generation of several bioactive compounds with important physiological roles. Furthermore, tryptophan has been reported to have a unique role as a nutritional signaling molecule that regulates protein synthesis in mouse and rat liver. In the present study, the acute effects of tryptophan on protein synthesis were confirmed and compared with those of leucine in rats. Eighteen hours fasted rats were orally administered of tryptophan or leucine at a dose of 135 mg/100 g body weight by gavage and then sacrificed 1 h after administration. The effects of tryptophan and leucine on the rate of protein synthesis were evaluated by the surface sensing of translation (SUnSET) method. We also examined the ability of tryptophan to induce activation of the mTOR pathway by measuring phosphorylation of 4E-BP1 and S6K1. Oral administration of tryptophan led to a stimulation of the rate of protein synthesis concomitant with activation of mTOR pathway in the liver, but not in skeletal muscle. We also investigated the sensitivity of liver protein synthesis to tryptophan administration. The half-maximal effective doses (ED50) of tryptophan in stimulating 4E-BP1 and S6K1 phosphorylation were both about 60% of daily intake. The effect of tryptophan on hepatic protein synthesis was similar to that of leucine on muscle protein synthesis, and the sensitivity of liver protein synthesis to tryptophan administration appeared to be almost the same or slightly lower than that of muscle protein synthesis to leucine administration.
Collapse
Affiliation(s)
- Kodwo Amuzuah Obeng
- Department of Biological Production Science, The United Graduate School of Agricultural Science, Tokyo University of Agriculture and Technology
| | - Shinji Mochizuki
- Department of Agrobiology and Bioresources, School of Agriculture, Utsunomiya University
| | - Shinichiro Koike
- Department of Biological Production Science, The United Graduate School of Agricultural Science, Tokyo University of Agriculture and Technology
| | - Yuka Toyoshima
- Department of Agrobiology and Bioresources, School of Agriculture, Utsunomiya University
| | - Yusuke Sato
- Department of Biological Production Science, The United Graduate School of Agricultural Science, Tokyo University of Agriculture and Technology.,Department of Agrobiology and Bioresources, School of Agriculture, Utsunomiya University
| | - Fumiaki Yoshizawa
- Department of Biological Production Science, The United Graduate School of Agricultural Science, Tokyo University of Agriculture and Technology.,Department of Agrobiology and Bioresources, School of Agriculture, Utsunomiya University
| |
Collapse
|
74
|
The role of branched chain amino acids metabolic disorders in tumorigenesis and progression. Biomed Pharmacother 2022; 153:113390. [DOI: 10.1016/j.biopha.2022.113390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/04/2022] [Accepted: 07/07/2022] [Indexed: 11/20/2022] Open
|
75
|
St-Cyr S, Child DD, Giaime E, Smith AR, Pascua CJ, Hahm S, Saiah E, Davidson BL. Huntington’s disease phenotypes are improved via mTORC1 modulation by small molecule therapy. PLoS One 2022; 17:e0273710. [PMID: 36037192 PMCID: PMC9423655 DOI: 10.1371/journal.pone.0273710] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 08/11/2022] [Indexed: 11/23/2022] Open
Abstract
Huntington’s Disease (HD) is a dominantly inherited neurodegenerative disease for which the major causes of mortality are neurodegeneration-associated aspiration pneumonia followed by cardiac failure. mTORC1 pathway perturbations are present in HD models and human tissues. Amelioration of mTORC1 deficits by genetic modulation improves disease phenotypes in HD models, is not a viable therapeutic strategy. Here, we assessed a novel small molecule mTORC1 pathway activator, NV-5297, for its improvement of the disease phenotypes in the N171-82Q HD mouse model. Oral dosing of NV-5297 over 6 weeks activated mTORC1, increased striatal volume, improved motor learning and heart contractility. Further, the heart contractility, heart fibrosis, and survival were improved in response to the cardiac stressor isoprenaline when compared to vehicle-treated mice. Cummulatively, these data support mTORC1 activation as a therapeutic target in HD and consolidates NV-5297 as a promising drug candidate for treating central and peripheral HD phenotypes and, more generally, mTORC1-deficit related diseases.
Collapse
Affiliation(s)
- Sophie St-Cyr
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Daniel D. Child
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
- The Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, United States of America
| | - Emilie Giaime
- Navitor Pharmaceuticals Inc., Cambridge, MA, United States of America
| | - Alicia R. Smith
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Christine J. Pascua
- Division of Cardiology, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
| | - Seung Hahm
- Navitor Pharmaceuticals Inc., Cambridge, MA, United States of America
| | - Eddine Saiah
- Navitor Pharmaceuticals Inc., Cambridge, MA, United States of America
- * E-mail: (BLD); (ES)
| | - Beverly L. Davidson
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States of America
- The Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA, United States of America
- * E-mail: (BLD); (ES)
| |
Collapse
|
76
|
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) senses nutrient levels in the cell and based on the availability, regulates cellular growth and proliferation. Its activity is tightly modulated by two GTPase units, the Rag GTPases and the Rheb GTPase. The Rag GTPases are the central hub of amino acid sensing as they summarize the amino acid signals from upstream regulators and control the subcellular localization of mTORC1. Unique from canonical signaling GTPases, the Rag GTPases are obligatory heterodimers, and the two subunits coordinate their nucleotide loading states to regulate their functional states. Robust biochemical analysis is indispensable to understanding the molecular mechanism governing the GTPase cycle. This chapter discusses protocols for purifying and biochemically characterizing the Rag GTPase heterodimer. We described two purification protocols to recombinantly produce the Rag GTPase heterodimer in large quantities. We then described assays to quantitatively measure the nucleotide binding and hydrolysis by the Rag GTPases. These assays allow for a thorough investigation of this unique heterodimeric GTPase, and they could be applicable to investigations of other noncanonical GTPases.
Collapse
Affiliation(s)
- Dylan D Doxsey
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, United States
| | - Kuang Shen
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, United States.
| |
Collapse
|
77
|
Gu X, Jouandin P, Lalgudi PV, Binari R, Valenstein ML, Reid MA, Allen AE, Kamitaki N, Locasale JW, Perrimon N, Sabatini DM. Sestrin mediates detection of and adaptation to low-leucine diets in Drosophila. Nature 2022; 608:209-216. [PMID: 35859173 PMCID: PMC10112710 DOI: 10.1038/s41586-022-04960-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 06/09/2022] [Indexed: 12/28/2022]
Abstract
Mechanistic target of rapamycin complex 1 (mTORC1) regulates cell growth and metabolism in response to multiple nutrients, including the essential amino acid leucine1. Recent work in cultured mammalian cells established the Sestrins as leucine-binding proteins that inhibit mTORC1 signalling during leucine deprivation2,3, but their role in the organismal response to dietary leucine remains elusive. Here we find that Sestrin-null flies (Sesn-/-) fail to inhibit mTORC1 or activate autophagy after acute leucine starvation and have impaired development and a shortened lifespan on a low-leucine diet. Knock-in flies expressing a leucine-binding-deficient Sestrin mutant (SesnL431E) have reduced, leucine-insensitive mTORC1 activity. Notably, we find that flies can discriminate between food with or without leucine, and preferentially feed and lay progeny on leucine-containing food. This preference depends on Sestrin and its capacity to bind leucine. Leucine regulates mTORC1 activity in glial cells, and knockdown of Sesn in these cells reduces the ability of flies to detect leucine-free food. Thus, nutrient sensing by mTORC1 is necessary for flies not only to adapt to, but also to detect, a diet deficient in an essential nutrient.
Collapse
Affiliation(s)
- Xin Gu
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA.
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Patrick Jouandin
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
- Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA.
| | - Pranav V Lalgudi
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Rich Binari
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA
| | - Max L Valenstein
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Michael A Reid
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Annamarie E Allen
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Nolan Kamitaki
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Jason W Locasale
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Norbert Perrimon
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
- Howard Hughes Medical Institute, Harvard Medical School, Boston, MA, USA.
| | | |
Collapse
|
78
|
Gardner G, Moradi F, Moffatt C, Cliche M, Garlisi B, Gratton J, Mehmood F, Stuart JA. Rapid nutrient depletion to below the physiological range by cancer cells cultured in Plasmax. Am J Physiol Cell Physiol 2022; 323:C823-C834. [DOI: 10.1152/ajpcell.00403.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mammalian cell culture is a fundamental tool used to study living cells. Presently, the standard protocol for performing cell culture involves the use of commercial media that contain an excess of nutrients. While this reduces the likelihood of cell starvation, it creates non-physiologic culture conditions that have been shown to 're-wire' cellular metabolism. Recently, researchers have developed new media like Plasmax, formulated to approximate the nutrient composition of human blood plasma. Although this represents an improvement in cell culture practice, physiologic media may be vulnerable to nutrient depletion. In this study we directly addressed this concern by measuring the rates of glucose and amino acid depletion from Plasmax in several cancer cell lines (PC-3, LNCaP, MCF-7, SH-SY5Y) over 48 hours. In all cell lines, depletion of glucose from Plasmax was rapid such that, by 48h, cells were hypoglycemic (<2mM glucose). Most amino acids were similarly rapidly depleted to sub-physiological levels by 48h. In contrast, glucose and most amino acids remained within the physiological range at 24h. When the experiment was done at physiological oxygen (5%) versus standard (18%) with LNCaP cells, no effect on glucose or amino acid consumption was observed. Using RNA sequencing, we show that this nutrient depletion is associated with enrichment of starvation responses, apoptotic signalling, and endoplasmic reticulum stress. A shift from glycolytic metabolism to mitochondrial respiration at 5% O2 was also measured using Seahorse analysis. Taken together, these results exemplify the metabolic considerations for Plasmax, highlighting that cell culture in Plasmax requires daily media exchange.
Collapse
Affiliation(s)
- Georgina Gardner
- Department of Biological Sciences, Brock University, St Catharines, Ontario, Canada
| | - Feresteh Moradi
- Department of Biological Sciences, Brock University, St Catharines, Ontario, Canada
| | - Christopher Moffatt
- Department of Biological Sciences, Brock University, St Catharines, Ontario, Canada
| | - Meagan Cliche
- Department of Biological Sciences, Brock University, St Catharines, Ontario, Canada
| | - Bianca Garlisi
- Department of Biological Sciences, Brock University, St Catharines, Ontario, Canada
| | - John Gratton
- Department of Biological Sciences, Brock University, St Catharines, Ontario, Canada
| | - Fatima Mehmood
- Department of Biological Sciences, Brock University, St Catharines, Ontario, Canada
| | - Jeffrey A. Stuart
- Department of Biological Sciences, Brock University, St Catharines, Ontario, Canada
| |
Collapse
|
79
|
mTORC1 and mTORC2 Complexes Regulate the Untargeted Metabolomics and Amino Acid Metabolites Profile through Mitochondrial Bioenergetic Functions in Pancreatic Beta Cells. Nutrients 2022; 14:nu14153022. [PMID: 35893876 PMCID: PMC9332257 DOI: 10.3390/nu14153022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 07/15/2022] [Accepted: 07/18/2022] [Indexed: 02/04/2023] Open
Abstract
Background: Pancreatic beta cells regulate bioenergetics efficiency and secret insulin in response to glucose and nutrient availability. The mechanistic Target of Rapamycin (mTOR) network orchestrates pancreatic progenitor cell growth and metabolism by nucleating two complexes, mTORC1 and mTORC2. Objective: To determine the impact of mTORC1/mTORC2 inhibition on amino acid metabolism in mouse pancreatic beta cells (Beta-TC-6 cells, ATCC-CRL-11506) using high-resolution metabolomics (HRM) and live-mitochondrial functions. Methods: Pancreatic beta TC-6 cells were incubated for 24 h with either: RapaLink-1 (RL); Torin-2 (T); rapamycin (R); metformin (M); a combination of RapaLink-1 and metformin (RLM); Torin-2 and metformin (TM); compared to the control. We applied high-resolution mass spectrometry (HRMS) LC-MS/MS untargeted metabolomics to compare the twenty natural amino acid profiles to the control. In addition, we quantified the bioenergetics dynamics and cellular metabolism by live-cell imaging and the MitoStress Test XF24 (Agilent, Seahorse). The real-time, live-cell approach simultaneously measures the oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) to determine cellular respiration and metabolism. Statistical significance was assessed using ANOVA on Ranks and post-hoc Welch t-Tests. Results: RapaLink-1, Torin-2, and rapamycin decreased L-aspartate levels compared to the control (p = 0.006). Metformin alone did not affect L-aspartate levels. However, L-asparagine levels decreased with all treatment groups compared to the control (p = 0.03). On the contrary, L-glutamate and glycine levels were reduced only by mTORC1/mTORC2 inhibitors RapaLink-1 and Torin-2, but not by rapamycin or metformin. The metabolic activity network model predicted that L-aspartate and AMP interact within the same activity network. Live-cell bioenergetics revealed that ATP production was significantly reduced in RapaLink-1 (122.23 ± 33.19), Torin-2 (72.37 ± 17.33) treated cells, compared to rapamycin (250.45 ± 9.41) and the vehicle control (274.23 ± 38.17), p < 0.01. However, non-mitochondrial oxygen consumption was not statistically different between RapaLink-1 (67.17 ± 3.52), Torin-2 (55.93 ± 8.76), or rapamycin (80.01 ± 4.36, p = 0.006). Conclusions: Dual mTORC1/mTORC2 inhibition by RapaLink-1 and Torin-2 differentially altered the amino acid profile and decreased mitochondrial respiration compared to rapamycin treatment which only blocks the FRB domain on mTOR. Third-generation mTOR inhibitors may alter the mitochondrial dynamics and reveal a bioenergetics profile that could be targeted to reduce mitochondrial stress.
Collapse
|
80
|
Vitto VAM, Bianchin S, Zolondick AA, Pellielo G, Rimessi A, Chianese D, Yang H, Carbone M, Pinton P, Giorgi C, Patergnani S. Molecular Mechanisms of Autophagy in Cancer Development, Progression, and Therapy. Biomedicines 2022; 10:1596. [PMID: 35884904 PMCID: PMC9313210 DOI: 10.3390/biomedicines10071596] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/25/2022] [Accepted: 06/30/2022] [Indexed: 01/18/2023] Open
Abstract
Autophagy is an evolutionarily conserved and tightly regulated process that plays an important role in maintaining cellular homeostasis. It involves regulation of various genes that function to degrade unnecessary or dysfunctional cellular components, and to recycle metabolic substrates. Autophagy is modulated by many factors, such as nutritional status, energy level, hypoxic conditions, endoplasmic reticulum stress, hormonal stimulation and drugs, and these factors can regulate autophagy both upstream and downstream of the pathway. In cancer, autophagy acts as a double-edged sword depending on the tissue type and stage of tumorigenesis. On the one hand, autophagy promotes tumor progression in advanced stages by stimulating tumor growth. On the other hand, autophagy inhibits tumor development in the early stages by enhancing its tumor suppressor activity. Moreover, autophagy drives resistance to anticancer therapy, even though in some tumor types, its activation induces lethal effects on cancer cells. In this review, we summarize the biological mechanisms of autophagy and its dual role in cancer. In addition, we report the current understanding of autophagy in some cancer types with markedly high incidence and/or lethality, and the existing therapeutic strategies targeting autophagy for the treatment of cancer.
Collapse
Affiliation(s)
- Veronica Angela Maria Vitto
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (V.A.M.V.); (S.B.); (G.P.); (A.R.); (D.C.); (P.P.)
| | - Silvia Bianchin
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (V.A.M.V.); (S.B.); (G.P.); (A.R.); (D.C.); (P.P.)
| | - Alicia Ann Zolondick
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI 96816, USA; (A.A.Z.); (H.Y.); (M.C.)
- Department of Molecular Biosciences and Bioengineering, University of Hawai’i at Manoa, Honolulu, HI 96816, USA
| | - Giulia Pellielo
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (V.A.M.V.); (S.B.); (G.P.); (A.R.); (D.C.); (P.P.)
| | - Alessandro Rimessi
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (V.A.M.V.); (S.B.); (G.P.); (A.R.); (D.C.); (P.P.)
| | - Diego Chianese
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (V.A.M.V.); (S.B.); (G.P.); (A.R.); (D.C.); (P.P.)
| | - Haining Yang
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI 96816, USA; (A.A.Z.); (H.Y.); (M.C.)
| | - Michele Carbone
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, HI 96816, USA; (A.A.Z.); (H.Y.); (M.C.)
| | - Paolo Pinton
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (V.A.M.V.); (S.B.); (G.P.); (A.R.); (D.C.); (P.P.)
| | - Carlotta Giorgi
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (V.A.M.V.); (S.B.); (G.P.); (A.R.); (D.C.); (P.P.)
| | - Simone Patergnani
- Laboratory for Technologies of Advanced Therapies (LTTA), Department of Medical Science, University of Ferrara, 44121 Ferrara, Italy; (V.A.M.V.); (S.B.); (G.P.); (A.R.); (D.C.); (P.P.)
| |
Collapse
|
81
|
Lv X, Zhou C, Yan Q, Tan Z, Kang J, Tang S. Elucidating the underlying mechanism of amino acids to regulate muscle protein synthesis: impact on human health. Nutrition 2022; 103-104:111797. [DOI: 10.1016/j.nut.2022.111797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 10/31/2022]
|
82
|
Yue S, Li G, He S, Li T. The central role of mTORC1 in amino acid sensing. Cancer Res 2022; 82:2964-2974. [PMID: 35749594 DOI: 10.1158/0008-5472.can-21-4403] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 04/28/2022] [Accepted: 06/17/2022] [Indexed: 11/16/2022]
Abstract
The mechanistic target of rapamycin (mTOR) is a master regulator of cell growth that controls cell homeostasis in response to nutrients, growth factors, and other environmental cues. Recent studies have emphasized the importance of lysosomes as a hub for nutrient sensing, especially amino acid sensing by mTORC1. This review highlights recent advances in understanding the amino acid-mTORC1 signaling axis and the role of mTORC1 in cancer.
Collapse
|
83
|
Trautman ME, Richardson NE, Lamming DW. Protein restriction and branched-chain amino acid restriction promote geroprotective shifts in metabolism. Aging Cell 2022; 21:e13626. [PMID: 35526271 PMCID: PMC9197406 DOI: 10.1111/acel.13626] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/13/2022] [Accepted: 04/21/2022] [Indexed: 01/20/2023] Open
Abstract
The proportion of humans suffering from age‐related diseases is increasing around the world, and creative solutions are needed to promote healthy longevity. Recent work has clearly shown that a calorie is not just a calorie—and that low protein diets are associated with reduced mortality in humans and promote metabolic health and extended lifespan in rodents. Many of the benefits of protein restriction on metabolism and aging are the result of decreased consumption of the three branched‐chain amino acids (BCAAs), leucine, isoleucine, and valine. Here, we discuss the emerging evidence that BCAAs are critical modulators of healthy metabolism and longevity in rodents and humans, as well as the physiological and molecular mechanisms that may drive the benefits of BCAA restriction. Our results illustrate that protein quality—the specific composition of dietary protein—may be a previously unappreciated driver of metabolic dysfunction and that reducing dietary BCAAs may be a promising new approach to delay and prevent diseases of aging.
Collapse
Affiliation(s)
- Michaela E. Trautman
- Department of Medicine University of Wisconsin‐Madison Madison Wisconsin USA
- William S. Middleton Memorial Veterans Hospital Madison Wisconsin USA
- Interdepartmental Graduate Program in Nutritional Sciences University of Wisconsin‐Madison Madison Wisconsin USA
| | - Nicole E. Richardson
- Department of Medicine University of Wisconsin‐Madison Madison Wisconsin USA
- William S. Middleton Memorial Veterans Hospital Madison Wisconsin USA
- Endocrinology and Reproductive Physiology Graduate Training Program University of Wisconsin‐Madison Madison Wisconsin USA
| | - Dudley W. Lamming
- Department of Medicine University of Wisconsin‐Madison Madison Wisconsin USA
- William S. Middleton Memorial Veterans Hospital Madison Wisconsin USA
- Endocrinology and Reproductive Physiology Graduate Training Program University of Wisconsin‐Madison Madison Wisconsin USA
| |
Collapse
|
84
|
Egri SB, Ouch C, Chou HT, Yu Z, Song K, Xu C, Shen K. Cryo-EM structures of the human GATOR1-Rag-Ragulator complex reveal a spatial-constraint regulated GAP mechanism. Mol Cell 2022; 82:1836-1849.e5. [PMID: 35338845 PMCID: PMC9133170 DOI: 10.1016/j.molcel.2022.03.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 12/09/2021] [Accepted: 02/28/2022] [Indexed: 12/20/2022]
Abstract
mTORC1 controls cellular metabolic processes in response to nutrient availability. Amino acid signals are transmitted to mTORC1 through the Rag GTPases, which are localized on the lysosomal surface by the Ragulator complex. The Rag GTPases receive amino acid signals from multiple upstream regulators. One negative regulator, GATOR1, is a GTPase activating protein (GAP) for RagA. GATOR1 binds to the Rag GTPases via two modes: an inhibitory mode and a GAP mode. How these two binding interactions coordinate to process amino acid signals is unknown. Here, we resolved three cryo-EM structural models of the GATOR1-Rag-Ragulator complex, with the Rag-Ragulator subcomplex occupying the inhibitory site, the GAP site, and both binding sites simultaneously. When the Rag GTPases bind to GATOR1 at the GAP site, both Rag subunits contact GATOR1 to coordinate their nucleotide loading states. These results reveal a potential GAP mechanism of GATOR1 during the mTORC1 inactivation process.
Collapse
Affiliation(s)
- Shawn B Egri
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, 373 Plantation St, Worcester, MA 01605, USA
| | - Christna Ouch
- Department of Biochemistry & Molecular Pharmacology, University of Massachusetts Chan Medical School, 364 Plantation St, Worcester, MA 01605, USA
| | - Hui-Ting Chou
- Howard Hughes Medical Institute, Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Zhiheng Yu
- Howard Hughes Medical Institute, Janelia Research Campus, 19700 Helix Drive, Ashburn, VA 20147, USA
| | - Kangkang Song
- Department of Biochemistry & Molecular Pharmacology, University of Massachusetts Chan Medical School, 364 Plantation St, Worcester, MA 01605, USA
| | - Chen Xu
- Department of Biochemistry & Molecular Pharmacology, University of Massachusetts Chan Medical School, 364 Plantation St, Worcester, MA 01605, USA
| | - Kuang Shen
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, 373 Plantation St, Worcester, MA 01605, USA.
| |
Collapse
|
85
|
Sawa R, Ohnishi A, Ohno M, Nagata M, Wake I, Okimura Y. Specific amino acids regulate Sestrin2 mRNA and protein levels in an ATF4-dependent manner in C2C12 myocytes. Biochim Biophys Acta Gen Subj 2022; 1866:130174. [PMID: 35597502 DOI: 10.1016/j.bbagen.2022.130174] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 04/29/2022] [Accepted: 05/16/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Sestrin2 is a conserved protein in several species, and its expression is upregulated in cells under various environmental stresses. Sestrin2 content is involved in the function of mechanistic target of rapamycin complex 1 (mTORC1) in mouse embryonic fibroblasts and C2C12 cells. METHODS C2C12 cells were treated with amino acid-free DMEM (AF-DMEM) for 5 h. The effects of the addition of specific amino acids to AF-DMEM on Sestrin2 mRNA and protein expression were examined using RT-qPCR and western blotting, respectively. The mechanism by which amino acids regulate Sestrin2 mRNA expression was examined using blocking and siRNA experiments. RESULTS AF-DMEM increased the mRNA and protein levels of both Sestrin2 and activating transcription factor 4 (ATF4). The addition of a specific amino acid changed Sestrin2 mRNA and protein levels. The response pattern of Sestrin2 to specific amino acids was similar to that of ATF4. ATF4 siRNA reduced Sestrin2 mRNA levels. AF-DMEM increased eukaryotic initiation factor 2α (eIF2α) phosphorylation as early as 10 min after the treatment; however, ATF4 and Sestrin2 were increased 300 min after the treatment. Nuclear factor erythroid 2-related factor 2 and pancreatic and duodenal homeobox 1 siRNA did not affect ATF4 and Sestrin2 mRNA expression. CONCLUSIONS Specific Amino acids regulate Sestrin2 levels in an ATF4-dependent manner in C2C12 cells. GENERAL SIGNIFICANCE The results of the present study indicate that amino acids regulate levels of Sestrin2, which might cause phenotypic alterations, including mTORC1 activity, in C2C12 cells.
Collapse
Affiliation(s)
- Ran Sawa
- Department of Nutrition and Food Science, Kobe Women's University Graduate School of Life Sciences, 2-1 Higashisuma-aoyama, Suma-ku, Kobe 654-8585, Japan
| | - Ayaka Ohnishi
- Department of Nutrition and Food Science, Kobe Women's University Graduate School of Life Sciences, 2-1 Higashisuma-aoyama, Suma-ku, Kobe 654-8585, Japan
| | - Maya Ohno
- Department of Nutrition and Food Science, Kobe Women's University Graduate School of Life Sciences, 2-1 Higashisuma-aoyama, Suma-ku, Kobe 654-8585, Japan
| | - Maika Nagata
- Department of Nutrition and Food Science, Kobe Women's University Graduate School of Life Sciences, 2-1 Higashisuma-aoyama, Suma-ku, Kobe 654-8585, Japan
| | - Ikumi Wake
- Department of Nutrition and Food Science, Kobe Women's University Graduate School of Life Sciences, 2-1 Higashisuma-aoyama, Suma-ku, Kobe 654-8585, Japan
| | - Yasuhiko Okimura
- Department of Nutrition and Food Science, Kobe Women's University Graduate School of Life Sciences, 2-1 Higashisuma-aoyama, Suma-ku, Kobe 654-8585, Japan.
| |
Collapse
|
86
|
Meng D, Yang Q, Jeong MH, Curukovic A, Tiwary S, Melick CH, Lama-Sherpa TD, Wang H, Huerta-Rosario M, Urquhart G, Zacharias LG, Lewis C, DeBerardinis RJ, Jewell JL. SNAT7 regulates mTORC1 via macropinocytosis. Proc Natl Acad Sci U S A 2022; 119:e2123261119. [PMID: 35561222 PMCID: PMC9171778 DOI: 10.1073/pnas.2123261119] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 03/13/2022] [Indexed: 11/30/2022] Open
Abstract
Mammalian target of rapamycin complex 1 (mTORC1) senses amino acids to control cell growth, metabolism, and autophagy. Some amino acids signal to mTORC1 through the Rag GTPase, whereas glutamine and asparagine activate mTORC1 through a Rag GTPase-independent pathway. Here, we show that the lysosomal glutamine and asparagine transporter SNAT7 activates mTORC1 after extracellular protein, such as albumin, is macropinocytosed. The N terminus of SNAT7 forms nutrient-sensitive interaction with mTORC1 and regulates mTORC1 activation independently of the Rag GTPases. Depletion of SNAT7 inhibits albumin-induced mTORC1 lysosomal localization and subsequent activation. Moreover, SNAT7 is essential to sustain KRAS-driven pancreatic cancer cell growth through mTORC1. Thus, SNAT7 links glutamine and asparagine signaling from extracellular protein to mTORC1 independently of the Rag GTPases and is required for macropinocytosis-mediated mTORC1 activation and pancreatic cancer cell growth.
Collapse
Affiliation(s)
- Delong Meng
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Qianmei Yang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Mi-Hyeon Jeong
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Adna Curukovic
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Shweta Tiwary
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Chase H. Melick
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Tshering D. Lama-Sherpa
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Huanyu Wang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Mariela Huerta-Rosario
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Greg Urquhart
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Lauren G. Zacharias
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Cheryl Lewis
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Ralph J. DeBerardinis
- Children's Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Jenna L. Jewell
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
87
|
Hisaoka M, Schott J, Bortecen T, Lindner D, Krijgsveld J, Stoecklin G. Preferential translation of p53 target genes. RNA Biol 2022; 19:437-452. [PMID: 35388737 PMCID: PMC8993080 DOI: 10.1080/15476286.2022.2048562] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The transcription factor p53 exerts its tumour suppressive effect through transcriptional activation of numerous target genes controlling cell cycle arrest, apoptosis, cellular senescence and DNA repair. In addition, there is evidence that p53 influences the translation of specific mRNAs, including translational inhibition of ribosomal protein synthesis and translational activation of MDM2. A challenge in the analysis of translational control is that changes in mRNA abundance exert a kinetic (passive) effect on ribosome densities. In order to separate these passive effects from active regulation of translation efficiency in response to p53 activation, we conducted a comprehensive analysis of translational regulation by comparative analysis of mRNA levels and ribosome densities upon DNA damage induced by neocarzinostatin in wild-type and TP53−/− HCT116 colorectal carcinoma cells. Thereby, we identified a specific group of mRNAs that are preferentially translated in response to p53 activation, many of which correspond to p53 target genes including MDM2, SESN1 and CDKN1A. By subsequent polysome profile analysis of SESN1 and CDKN1A mRNA, we could demonstrate that p53-dependent translational activation relies on a combination of inducing the expression of translationally advantageous isoforms and trans-acting mechanisms that further enhance the translation of these mRNAs.
Collapse
Affiliation(s)
- Miharu Hisaoka
- Division of Biochemistry Mannheim Institute for Innate Immunoscience (MI3) and Mannheim Cancer Center (MCC), Medical Faculty Mannheim of Heidelberg University, Mannheim, Germany.,Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBHAlliance, Heidelberg, Germany.,National Center for Tumor Diseases (NCT) partner site, Heidelberg, Germany
| | - Johanna Schott
- Division of Biochemistry Mannheim Institute for Innate Immunoscience (MI3) and Mannheim Cancer Center (MCC), Medical Faculty Mannheim of Heidelberg University, Mannheim, Germany.,Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBHAlliance, Heidelberg, Germany
| | - Toman Bortecen
- Division of Proteomics of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Bioscience, Heidelberg University, Heidelberg, Germany
| | - Doris Lindner
- Division of Biochemistry Mannheim Institute for Innate Immunoscience (MI3) and Mannheim Cancer Center (MCC), Medical Faculty Mannheim of Heidelberg University, Mannheim, Germany.,Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBHAlliance, Heidelberg, Germany
| | - Jeroen Krijgsveld
- Division of Proteomics of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Medical Faculty, Heidelberg University, Heidelberg, Germany
| | - Georg Stoecklin
- Division of Biochemistry Mannheim Institute for Innate Immunoscience (MI3) and Mannheim Cancer Center (MCC), Medical Faculty Mannheim of Heidelberg University, Mannheim, Germany.,Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBHAlliance, Heidelberg, Germany.,National Center for Tumor Diseases (NCT) partner site, Heidelberg, Germany
| |
Collapse
|
88
|
Xu C, Yang J, Cao J, Jiang N, Zhou Y, Zeng L, Zhong Q, Fan Y. The quantitative proteomic analysis of rare minnow, Gobiocypris rarus, infected with virulent and attenuated isolates of grass carp reovirus genotype Ⅱ. FISH & SHELLFISH IMMUNOLOGY 2022; 123:142-151. [PMID: 35219830 DOI: 10.1016/j.fsi.2022.02.037] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/16/2022] [Accepted: 02/18/2022] [Indexed: 06/14/2023]
Abstract
Grass carp reovirus genotype Ⅱ (GCRV II) causes severe hemorrhagic disease in grass carp and affects the aquaculture industry in China. GCRV Ⅱ isolates have been collected from different epidemic areas in China, and these isolates can lead to different degrees of hemorrhagic symptoms in grass carp. Rare minnow (Gobiocypris rarus) is widely used as a model fish to study the mechanism of hemorrhagic disease because of its high sensitivity to GCRV. In this study, the protein levels in the spleen of rare minnow after infection with GCRV virulent isolate JZ809 and attenuated isolate XT422 were investigated using isobaric tags for relative and absolute quantitation (iTRAQ)-based quantitative proteomics. 109 and 50 differentially expressed proteins (DEPs) in the virulent and attenuated infection groups were obtained, respectively, among which 40 DEPs were identified in both groups. Combining protein expression profiling with gene ontology (GO) annotation, the responses of rare minnow to the two genotypes GCRV Ⅱ in terms of upregulated proteins were similar, focusing on ATP synthesis, in which ATP can serve as a "danger" signal to activate an immunoreaction in eukaryotes. Meanwhile, the virulent genotype JZ809 induced more immunoproteins and increased the levels of ubiquitin-proteasome system members to adapt to virus infection. However, together with a persistent and excessive inflammatory response and declining carbon metabolism, rare minnow presented more severe hemorrhagic disease and mortality after infection with virulent JZ809 than with attenuated XT422. The results provide a valuable information that will increase our understanding of the pathogenesis of viruses with different levels of virulence and the mechanism of interaction between the virus and host. Furthermore, the 6 proteins that were only significantly upregulated in the XT422 infection group all belonged to cluster 2, and 28 of 30 proteins that were only upregulated in JZ809 infection group were clustered into cluster 1. For the downregulated proteins, all DEPs in the XT422 infection group were clustered into cluster 4, and 25 of 39 proteins that were only significantly downregulated in the JZ809 infection group belonged to cluster 3. The results indicated that the DEPs in the attenuated XT422 infection group might be sensitive and their abundance changed more quickly when fish experienced virus infection.
Collapse
Affiliation(s)
- Chen Xu
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, 430223, China.
| | - Jie Yang
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, 430223, China; College of Biological Science and Engineering, Jiangxi Agricultural University, Nanchang, 330045, China.
| | - JiaJia Cao
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, 430223, China; College of Biological Science and Engineering, Jiangxi Agricultural University, Nanchang, 330045, China.
| | - Nan Jiang
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, 430223, China.
| | - Yong Zhou
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, 430223, China.
| | - Lingbing Zeng
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, 430223, China.
| | - Qiwang Zhong
- College of Biological Science and Engineering, Jiangxi Agricultural University, Nanchang, 330045, China.
| | - Yuding Fan
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan, 430223, China; College of Biological Science and Engineering, Jiangxi Agricultural University, Nanchang, 330045, China.
| |
Collapse
|
89
|
Transcriptome analysis of HEK 293T cells revealed different significance of the depletion of DNA-dependent protein kinase subunits, Ku70, Ku80, and DNA-PKcs. Biochimie 2022; 199:139-149. [DOI: 10.1016/j.biochi.2022.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/17/2022] [Accepted: 04/12/2022] [Indexed: 01/08/2023]
|
90
|
Simcox J, Lamming DW. The central moTOR of metabolism. Dev Cell 2022; 57:691-706. [PMID: 35316619 PMCID: PMC9004513 DOI: 10.1016/j.devcel.2022.02.024] [Citation(s) in RCA: 74] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/20/2022] [Accepted: 02/24/2022] [Indexed: 12/21/2022]
Abstract
The protein kinase mechanistic target of rapamycin (mTOR) functions as a central regulator of metabolism, integrating diverse nutritional and hormonal cues to control anabolic processes, organismal physiology, and even aging. This review discusses the current state of knowledge regarding the regulation of mTOR signaling and the metabolic regulation of the four macromolecular building blocks of the cell: carbohydrate, nucleic acid, lipid, and protein by mTOR. We review the role of mTOR in the control of organismal physiology and aging through its action in key tissues and discuss the potential for clinical translation of mTOR inhibition for the treatment and prevention of diseases of aging.
Collapse
Affiliation(s)
- Judith Simcox
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA.
| | - Dudley W Lamming
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA; Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
91
|
ROCK ‘n TOR: An Outlook on Keratinocyte Stem Cell Expansion in Regenerative Medicine via Protein Kinase Inhibition. Cells 2022; 11:cells11071130. [PMID: 35406693 PMCID: PMC8997668 DOI: 10.3390/cells11071130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 12/13/2022] Open
Abstract
Keratinocyte stem cells play a fundamental role in homeostasis and repair of stratified epithelial tissues. Transplantation of cultured keratinocytes autografts provides a landmark example of successful cellular therapies by restoring durable integrity in stratified epithelia lost to devastating tissue conditions. Despite the overall success of such procedures, failures still occur in case of paucity of cultured stem cells in therapeutic grafts. Strategies aiming at a further amplification of stem cells during keratinocyte ex vivo expansion may thus extend the applicability of these treatments to subjects in which endogenous stem cells pools are depauperated by aging, trauma, or disease. Pharmacological targeting of stem cell signaling pathways is recently emerging as a powerful strategy for improving stem cell maintenance and/or amplification. Recent experimental data indicate that pharmacological inhibition of two prominent keratinocyte signaling pathways governed by apical mTOR and ROCK protein kinases favor stem cell maintenance and/or amplification ex vivo and may improve the effectiveness of stem cell-based therapeutic procedures. In this review, we highlight the pathophysiological roles of mTOR and ROCK in keratinocyte biology and evaluate existing pre-clinical data on the effects of their inhibition in epithelial stem cell expansion for transplantation purposes.
Collapse
|
92
|
Xie Z, Zhang W, Zhang Y. Loss of Slc38a4 imprinting is a major cause of mouse placenta hyperplasia in somatic cell nuclear transferred embryos at late gestation. Cell Rep 2022; 38:110407. [PMID: 35196486 PMCID: PMC8919768 DOI: 10.1016/j.celrep.2022.110407] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 11/02/2021] [Accepted: 01/26/2022] [Indexed: 12/12/2022] Open
Abstract
Placenta hyperplasia is commonly observed in cloned animals and is believed to impede the proper development of cloned embryos. However, the mechanism underlying this phenomenon is largely unknown. Here, we show that placenta hyperplasia of cloned mouse embryos occurs in both middle and late gestation. Interestingly, restoring paternal-specific expression of an amino acid transporter Slc38a4, which loses maternal H3K27me3-dependent imprinting and becomes biallelically expressed in cloned placentae, rescues the overgrowth of cloned placentae at late gestation. Molecular analyses reveal that loss of Slc38a4 imprinting leads to over-activation of the mechanistic target of rapamycin complex 1 (mTORC1) signaling pathway in cloned placentae, which is likely due to the increased amino acids transport by SLC38A4. Collectively, our study not only reveals loss of Slc38a4 imprinting is responsible for overgrowth of cloned placentae at late gestation but also suggests the underlying mechanism involves increased amino acid transport and over-activation of mTORC1.
Collapse
Affiliation(s)
- Zhenfei Xie
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA; The Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Wenhao Zhang
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA
| | - Yi Zhang
- Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA 02115, USA; Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA; Division of Hematology/Oncology, Department of Pediatrics, Boston Children's Hospital, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, WAB-149G, 200 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
93
|
Nowosad A, Besson A. Lysosomes at the Crossroads of Cell Metabolism, Cell Cycle, and Stemness. Int J Mol Sci 2022; 23:ijms23042290. [PMID: 35216401 PMCID: PMC8879101 DOI: 10.3390/ijms23042290] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 02/11/2022] [Accepted: 02/16/2022] [Indexed: 02/07/2023] Open
Abstract
Initially described as lytic bodies due to their degradative and recycling functions, lysosomes play a critical role in metabolic adaptation to nutrient availability. More recently, the contribution of lysosomal proteins to cell signaling has been established, and lysosomes have emerged as signaling hubs that regulate diverse cellular processes, including cell proliferation and cell fate. Deciphering these signaling pathways has revealed an extensive crosstalk between the lysosomal and cell cycle machineries that is only beginning to be understood. Recent studies also indicate that a number of lysosomal proteins are involved in the regulation of embryonic and adult stem cell fate and identity. In this review, we will focus on the role of the lysosome as a signaling platform with an emphasis on its function in integrating nutrient sensing with proliferation and cell cycle progression, as well as in stemness-related features, such as self-renewal and quiescence.
Collapse
Affiliation(s)
- Ada Nowosad
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), University of Toulouse, CNRS, UPS, 31062 Toulouse, France;
- Department of Oncology, KULeuven, Laboratory for Molecular Cancer Biology, Center for Cancer Biology, VIB, 3000 Leuven, Belgium
| | - Arnaud Besson
- Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), University of Toulouse, CNRS, UPS, 31062 Toulouse, France;
- Correspondence: ; Tel.: +33-561558486
| |
Collapse
|
94
|
Wang D, Xu C, Yang W, Chen J, Ou Y, Guan Y, Guan J, Liu Y. E3 ligase RNF167 and deubiquitinase STAMBPL1 modulate mTOR and cancer progression. Mol Cell 2022; 82:770-784.e9. [PMID: 35114100 DOI: 10.1016/j.molcel.2022.01.002] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 11/15/2021] [Accepted: 12/29/2021] [Indexed: 12/16/2022]
Abstract
The mTOR complex 1 (mTORC1) is an essential metabolic hub that coordinates cellular metabolism with the availability of nutrients, including amino acids. Sestrin2 has been identified as a cytosolic leucine sensor that transmits leucine status signals to mTORC1. In this study, we identify an E3 ubiquitin ligase RING finger protein 167 (RNF167) and a deubiquitinase STAMBPL1 that function in concert to control the polyubiquitination level of Sestrin2 in response to leucine availability. Ubiquitination of Sestrin2 promotes its interaction with GATOR2 and inhibits mTORC1 signaling. Bioinformatic analysis reveals decreased RNF167 expression and increased STAMBPL1 expression in gastric and colorectal tumors. Knockout of STAMBPL1 or correction of the heterozygous STAMBPL1 mutation in a human colon cancer cell line suppresses xenograft tumor growth. Lastly, a cell-permeable peptide that blocks the STAMBPL1-Sestrin2 interaction inhibits mTORC1 and provides a potential option for cancer therapy.
Collapse
Affiliation(s)
- Dong Wang
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Chenchen Xu
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Wenyu Yang
- Yuan Pei College, Peking University, Beijing 100871, China
| | - Jie Chen
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Yuhui Ou
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Yuanyuan Guan
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
| | - Jialiang Guan
- PKU-Tsinghua-NIBS Graduate Program, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Ying Liu
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China; Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China; Beijing Advanced Innovation Center for Genomics, Beijing 100871, China.
| |
Collapse
|
95
|
Khalil H, Abd ElHady A, Elawdan KA, Mohamed D, Mohamed DD, Abd El Maksoud AI, El-Chennawi FA, El-Fikiy B, El-Sayed IH. The Mechanical Autophagy as a Part of Cellular Immunity; Facts and Features in Treating the Medical Disorders. Immunol Invest 2022; 51:266-289. [PMID: 32993405 DOI: 10.1080/08820139.2020.1828453] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Autophagy is a cellular housekeeping process that incorporates lysosomal-degradation to maintain cell survival and energy sources. In recent decades, the role of autophagy has implicated in the initiation and development of many diseases that affect humanity. Among these diseases are autoimmune diseases and neurodegenerative diseases, which connected with the lacking autophagy. Other diseases are connected with the increasing levels of autophagy such as cancers and infectious diseases. Therefore, controlling autophagy with sufficient regulators could represent an effective strategy to overcome such diseases. Interestingly, targeting autophagy can also provide a sufficient method to combat the current epidemic caused by the ongoing coronavirus. In this review, we aim to highlight the physiological function of the autophagic process to understand the circumstances surrounding its role in the cellular immunity associated with the development of human diseases.
Collapse
Affiliation(s)
- Hany Khalil
- Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | - Amira Abd ElHady
- Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | - Khaled A Elawdan
- Department of Molecular Biology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | - Dalia Mohamed
- Industrial Biotechnology Department, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | - Doaa D Mohamed
- Industrial Biotechnology Department, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | - Ahmed I Abd El Maksoud
- Industrial Biotechnology Department, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | - Farha A El-Chennawi
- Clinical Pathology Department, Faculty of Medicine, Mansora University, Mansora, Egypt
| | - Bhgat El-Fikiy
- Department of Animal Biotechnology, Genetic Engineering and Biotechnology Research Institute, University of Sadat City, Sadat City, Egypt
| | - Ibrahim H El-Sayed
- Chemistry Department, Faculty of Science, Kafrelsheikh University, Kafrelsheikh, Egypt
| |
Collapse
|
96
|
Oyabu M, Takigawa K, Mizutani S, Hatazawa Y, Fujita M, Ohira Y, Sugimoto T, Suzuki O, Tsuchiya K, Suganami T, Ogawa Y, Ishihara K, Miura S, Kamei Y. FOXO1 cooperates with C/EBPδ and ATF4 to regulate skeletal muscle atrophy transcriptional program during fasting. FASEB J 2022; 36:e22152. [DOI: 10.1096/fj.202101385rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 12/22/2021] [Accepted: 12/23/2021] [Indexed: 12/14/2022]
Affiliation(s)
- Mamoru Oyabu
- Laboratory of Molecular Nutrition Graduate School of Life and Environmental Sciences Kyoto Prefectural University Kyoto Japan
| | - Kaho Takigawa
- Laboratory of Molecular Nutrition Graduate School of Life and Environmental Sciences Kyoto Prefectural University Kyoto Japan
| | - Sako Mizutani
- Laboratory of Molecular Nutrition Graduate School of Life and Environmental Sciences Kyoto Prefectural University Kyoto Japan
| | - Yukino Hatazawa
- Laboratory of Molecular Nutrition Graduate School of Life and Environmental Sciences Kyoto Prefectural University Kyoto Japan
| | - Mariko Fujita
- Laboratory of Molecular Nutrition Graduate School of Life and Environmental Sciences Kyoto Prefectural University Kyoto Japan
| | - Yuto Ohira
- Laboratory of Molecular Nutrition Graduate School of Life and Environmental Sciences Kyoto Prefectural University Kyoto Japan
| | - Takumi Sugimoto
- Laboratory of Molecular Nutrition Graduate School of Life and Environmental Sciences Kyoto Prefectural University Kyoto Japan
| | - Osamu Suzuki
- Laboratory of Animal Models for Human Diseases National Institutes of Biomedical Innovation, Health and Nutrition Osaka Japan
| | - Kyoichiro Tsuchiya
- Third Department of Internal Medicine Interdisciplinary Graduate School of Medicine and Engineering University of Yamanashi Yamanashi Japan
| | - Takayoshi Suganami
- Department of Molecular Medicine and Metabolism Research Institute of Environmental Medicine Nagoya University Nagoya Japan
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Kengo Ishihara
- Department of Food Science and Human Nutrition Faculty of Agriculture Ryukoku University Shiga Japan
| | - Shinji Miura
- Graduate School of Nutritional and Environmental Sciences University of Shizuoka Shizuoka Japan
| | - Yasutomi Kamei
- Laboratory of Molecular Nutrition Graduate School of Life and Environmental Sciences Kyoto Prefectural University Kyoto Japan
| |
Collapse
|
97
|
Sanz B, Rezola-Pardo C, Arrieta H, Fraile-Bermúdez AB, Alonso-Puyo J, Molano I, Rodriguez-Larrad A, Irazusta J. Serum Sestrin-1 Concentration Is Higher in Frail than Non-Frail Older People Living in Nursing Homes. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:ijerph19031079. [PMID: 35162104 PMCID: PMC8834059 DOI: 10.3390/ijerph19031079] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/11/2022] [Accepted: 01/15/2022] [Indexed: 02/04/2023]
Abstract
Given the increasing prevalence of frailty and its implications for public health, the identification of biomarkers to detect frailty is essential. Sestrin-1 is a protein with a protective role in muscle function. This study aimed to determine whether the serum sestrin-1 concentration differed between frail and non-frail populations and to investigate its association with frailty-related variables in 225 older women and men living in nursing homes (Gipuzkoa, Spain). Serum sestrin-1 concentration was measured by ELISA. Frailty, dependence, anthropometry, physical function, and physical activity were determined by validated tests and tools. The associations between sestrin-1 concentration and the other variables were determined using generalized linear models. The differences between frail and non-frail individuals were analyzed by the Mann–Whitney U-test, and receiver operating characteristic (ROC) curves were constructed to calculate the capability of sestrin-1 to detect frailty. Unexpectedly, frail individuals—according to the Fried Frailty Phenotype or the Clinical Frailty Scale—had higher serum sestrin-1 concentrations than non-frail individuals. Furthermore, the higher serum sestrin-1 concentration was associated with the increased frailty scores and dependence as well as the poorer physical function and the less physical activity. Given the contradictory results regarding serum sestrin-1 and frailty, further investigation is required to propose it as a molecular biomarker of frailty.
Collapse
Affiliation(s)
- Begoña Sanz
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 489040 Leioa, Bizkaia, Spain; (C.R.-P.); (J.A.-P.); (A.R.-L.); (J.I.)
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Bizkaia, Spain
- Correspondence: ; Tel.: +34-946013307
| | - Chloe Rezola-Pardo
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 489040 Leioa, Bizkaia, Spain; (C.R.-P.); (J.A.-P.); (A.R.-L.); (J.I.)
| | - Haritz Arrieta
- Department of Nursing II, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 20014 Donostia-San Sebastián, Gipuzkoa, Spain;
| | - Ana Belén Fraile-Bermúdez
- Department of Nursing I, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 48940 Leioa, Bizkaia, Spain;
| | - Janire Alonso-Puyo
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 489040 Leioa, Bizkaia, Spain; (C.R.-P.); (J.A.-P.); (A.R.-L.); (J.I.)
| | - Irene Molano
- Residencia Sanmarcosene, Carretera de San Marcos, s/n, 20100 Errenteria, Gipuzkoa, Spain;
| | - Ana Rodriguez-Larrad
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 489040 Leioa, Bizkaia, Spain; (C.R.-P.); (J.A.-P.); (A.R.-L.); (J.I.)
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Bizkaia, Spain
| | - Jon Irazusta
- Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), 489040 Leioa, Bizkaia, Spain; (C.R.-P.); (J.A.-P.); (A.R.-L.); (J.I.)
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Bizkaia, Spain
| |
Collapse
|
98
|
Yu Z, Yang Z, Ren G, Wang Y, Luo X, Zhu F, Yu S, Jia L, Chen M, Worley PF, Xiao B. GATOR2 complex-mediated amino acid signaling regulates brain myelination. Proc Natl Acad Sci U S A 2022; 119:e2110917119. [PMID: 35022234 PMCID: PMC8784133 DOI: 10.1073/pnas.2110917119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 11/16/2021] [Indexed: 02/05/2023] Open
Abstract
Amino acids are essential for cell growth and metabolism. Amino acid and growth factor signaling pathways coordinately regulate the mechanistic target of rapamycin complex 1 (mTORC1) kinase in cell growth and organ development. While major components of amino acid signaling mechanisms have been identified, their biological functions in organ development are unclear. We aimed to understand the functions of the critically positioned amino acid signaling complex GAP activity towards Rags 2 (GATOR2) in brain development. GATOR2 mediates amino acid signaling to mTORC1 by directly linking the amino acid sensors for arginine and leucine to downstream signaling complexes. Now, we report a role of GATOR2 in oligodendrocyte myelination in postnatal brain development. We show that the disruption of GATOR2 complex by genetic deletion of meiosis regulator for oocyte development (Mios, encoding a component of GATOR2) selectively impairs the formation of myelinating oligodendrocytes, thus brain myelination, without apparent effects on the formation of neurons and astrocytes. The loss of Mios impairs cell cycle progression of oligodendrocyte precursor cells, leading to their reduced proliferation and differentiation. Mios deletion manifests a cell type-dependent effect on mTORC1 in the brain, with oligodendroglial mTORC1 selectively affected. However, the role of Mios/GATOR2 in oligodendrocyte formation and myelination involves mTORC1-independent function. This study suggests that GATOR2 coordinates amino acid and growth factor signaling to regulate oligodendrocyte myelination.
Collapse
Affiliation(s)
- Zongyan Yu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150000, People's Republic of China
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518005, People's Republic of China
- Department of Biology, School of Life Sciences, Brain Research Center, Southern University of Science and Technology, Shenzhen 518000, People's Republic of China
| | - Zhiwen Yang
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518005, People's Republic of China
- Department of Biology, School of Life Sciences, Brain Research Center, Southern University of Science and Technology, Shenzhen 518000, People's Republic of China
| | - Guoru Ren
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518005, People's Republic of China
- Department of Biology, School of Life Sciences, Brain Research Center, Southern University of Science and Technology, Shenzhen 518000, People's Republic of China
| | - Yingjie Wang
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518005, People's Republic of China
- Department of Biology, School of Life Sciences, Brain Research Center, Southern University of Science and Technology, Shenzhen 518000, People's Republic of China
| | - Xiang Luo
- School of Life Science and Technology, Harbin Institute of Technology, Harbin 150000, People's Republic of China
- Department of Biology, School of Life Sciences, Brain Research Center, Southern University of Science and Technology, Shenzhen 518000, People's Republic of China
| | - Feiyan Zhu
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518005, People's Republic of China
- Department of Biology, School of Life Sciences, Brain Research Center, Southern University of Science and Technology, Shenzhen 518000, People's Republic of China
| | - Shouyang Yu
- Neuroscience & Metabolism Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, People's Republic of China
| | - Lanlan Jia
- Neuroscience & Metabolism Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, People's Republic of China
| | - Mina Chen
- Neuroscience & Metabolism Research, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu 610041, People's Republic of China
| | - Paul F Worley
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Bo Xiao
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518005, People's Republic of China;
- Department of Biology, School of Life Sciences, Brain Research Center, Southern University of Science and Technology, Shenzhen 518000, People's Republic of China
| |
Collapse
|
99
|
Sebestyén A, Dankó T, Sztankovics D, Moldvai D, Raffay R, Cervi C, Krencz I, Zsiros V, Jeney A, Petővári G. The role of metabolic ecosystem in cancer progression — metabolic plasticity and mTOR hyperactivity in tumor tissues. Cancer Metastasis Rev 2022; 40:989-1033. [PMID: 35029792 PMCID: PMC8825419 DOI: 10.1007/s10555-021-10006-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/26/2021] [Indexed: 12/14/2022]
Abstract
Despite advancements in cancer management, tumor relapse and metastasis are associated with poor outcomes in many cancers. Over the past decade, oncogene-driven carcinogenesis, dysregulated cellular signaling networks, dynamic changes in the tissue microenvironment, epithelial-mesenchymal transitions, protein expression within regulatory pathways, and their part in tumor progression are described in several studies. However, the complexity of metabolic enzyme expression is considerably under evaluated. Alterations in cellular metabolism determine the individual phenotype and behavior of cells, which is a well-recognized hallmark of cancer progression, especially in the adaptation mechanisms underlying therapy resistance. In metabolic symbiosis, cells compete, communicate, and even feed each other, supervised by tumor cells. Metabolic reprogramming forms a unique fingerprint for each tumor tissue, depending on the cellular content and genetic, epigenetic, and microenvironmental alterations of the developing cancer. Based on its sensing and effector functions, the mechanistic target of rapamycin (mTOR) kinase is considered the master regulator of metabolic adaptation. Moreover, mTOR kinase hyperactivity is associated with poor prognosis in various tumor types. In situ metabolic phenotyping in recent studies highlights the importance of metabolic plasticity, mTOR hyperactivity, and their role in tumor progression. In this review, we update recent developments in metabolic phenotyping of the cancer ecosystem, metabolic symbiosis, and plasticity which could provide new research directions in tumor biology. In addition, we suggest pathomorphological and analytical studies relating to metabolic alterations, mTOR activity, and their associations which are necessary to improve understanding of tumor heterogeneity and expand the therapeutic management of cancer.
Collapse
|
100
|
Chen Y, Huang T, Yu Z, Yu Q, Wang Y, Hu J, Shi J, Yang G. The functions and roles of sestrins in regulating human diseases. Cell Mol Biol Lett 2022; 27:2. [PMID: 34979914 PMCID: PMC8721191 DOI: 10.1186/s11658-021-00302-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022] Open
Abstract
Sestrins (Sesns), highly conserved stress-inducible metabolic proteins, are known to protect organisms against various noxious stimuli including DNA damage, oxidative stress, starvation, endoplasmic reticulum (ER) stress, and hypoxia. Sesns regulate metabolism mainly through activation of the key energy sensor AMP-dependent protein kinase (AMPK) and inhibition of mammalian target of rapamycin complex 1 (mTORC1). Sesns also play pivotal roles in autophagy activation and apoptosis inhibition in normal cells, while conversely promoting apoptosis in cancer cells. The functions of Sesns in diseases such as metabolic disorders, neurodegenerative diseases, cardiovascular diseases, and cancer have been broadly investigated in the past decades. However, there is a limited number of reviews that have summarized the functions of Sesns in the pathophysiological processes of human diseases, especially musculoskeletal system diseases. One aim of this review is to discuss the biological functions of Sesns in the pathophysiological process and phenotype of diseases. More significantly, we include some new evidence about the musculoskeletal system. Another purpose is to explore whether Sesns could be potential biomarkers or targets in the future diagnostic and therapeutic process.
Collapse
Affiliation(s)
- Yitong Chen
- Department of Orthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, Zhejiang, China
| | - Tingben Huang
- Department of Implantology, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, Zhejiang, China
| | - Zhou Yu
- Department of Implantology, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, Zhejiang, China
| | - Qiong Yu
- Department of Implantology, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, Zhejiang, China
| | - Ying Wang
- Department of Oral Medicine, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, Zhejiang, China
| | - Ji'an Hu
- Department of Oral Pathology, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, Zhejiang, China.
| | - Jiejun Shi
- Department of Orthodontics, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, Zhejiang, China.
| | - Guoli Yang
- Department of Implantology, Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, Zhejiang, China.
| |
Collapse
|