1
|
Keary KM, Gu QH, Chen J, Li Z. Dendritic distribution of autophagosomes underlies pathway-selective induction of LTD. Cell Rep 2023; 42:112898. [PMID: 37516958 PMCID: PMC10528062 DOI: 10.1016/j.celrep.2023.112898] [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: 11/02/2022] [Revised: 05/31/2023] [Accepted: 07/13/2023] [Indexed: 08/01/2023] Open
Abstract
The mechanism of long-term depression (LTD), a cellular substrate for learning, memory, and behavioral flexibility, is extensively studied in Schaffer collateral (SC) synapses, with inhibition of autophagy identified as a key factor. SC inputs terminate at basal and proximal apical dendrites, whereas distal apical dendrites receive inputs from the temporoammonic pathway (TAP). Here, we demonstrate that TAP and SC synapses have a shared LTD mechanism reliant on NMDA receptors, caspase-3, and autophagy inhibition. Despite this shared LTD mechanism, proximal apical dendrites contain more autophagosomes than distal apical dendrites. Additionally, unlike SC LTD, which diminishes with age, TAP LTD persists into adulthood. Our previous study shows that the high autophagy in adulthood disallows SC LTD induction. The reduction of autophagosomes from proximal to distal dendrites, combined with distinct LTD inducibility at SC and TAP synapses, suggests a model where the differential distribution of autophagosomes in dendrites gates LTD inducibility at specific circuits.
Collapse
Affiliation(s)
- Kevin M Keary
- Section on Synapse Development Plasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA; Department of Neuroscience, Brown University, Providence, RI 02912, USA
| | - Qin-Hua Gu
- Section on Synapse Development Plasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jiji Chen
- Advanced Imaging and Microscopy (AIM) Resource, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892, USA
| | - Zheng Li
- Section on Synapse Development Plasticity, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
2
|
Artoni F, Grützmacher N, Demetriades C. Unbiased evaluation of rapamycin's specificity as an mTOR inhibitor. Aging Cell 2023; 22:e13888. [PMID: 37222020 PMCID: PMC10410055 DOI: 10.1111/acel.13888] [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: 12/22/2022] [Revised: 05/02/2023] [Accepted: 05/07/2023] [Indexed: 05/25/2023] Open
Abstract
Rapamycin is a macrolide antibiotic that functions as an immunosuppressive and anti-cancer agent, and displays robust anti-ageing effects in multiple organisms including humans. Importantly, rapamycin analogues (rapalogs) are of clinical importance against certain cancer types and neurodevelopmental diseases. Although rapamycin is widely perceived as an allosteric inhibitor of mTOR (mechanistic target of rapamycin), the master regulator of cellular and organismal physiology, its specificity has not been thoroughly evaluated so far. In fact, previous studies in cells and in mice hinted that rapamycin may be also acting independently from mTOR to influence various cellular processes. Here, we generated a gene-edited cell line that expresses a rapamycin-resistant mTOR mutant (mTORRR ) and assessed the effects of rapamycin treatment on the transcriptome and proteome of control or mTORRR -expressing cells. Our data reveal a striking specificity of rapamycin towards mTOR, demonstrated by virtually no changes in mRNA or protein levels in rapamycin-treated mTORRR cells, even following prolonged drug treatment. Overall, this study provides the first unbiased and conclusive assessment of rapamycin's specificity, with potential implications for ageing research and human therapeutics.
Collapse
Affiliation(s)
- Filippo Artoni
- Max Planck Institute for Biology of Ageing (MPI‐AGE)CologneGermany
- Cologne Graduate School of Ageing Research (CGA)CologneGermany
| | - Nina Grützmacher
- Max Planck Institute for Biology of Ageing (MPI‐AGE)CologneGermany
| | - Constantinos Demetriades
- Max Planck Institute for Biology of Ageing (MPI‐AGE)CologneGermany
- Cologne Graduate School of Ageing Research (CGA)CologneGermany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD)University of CologneCologneGermany
| |
Collapse
|
3
|
Li M, Lan Y, Gao J, Yuan S, Hou S, Guo T, Zhao F, Wang Y, Yuan W, Wang X. Rapamycin Promotes the Expansion of Myeloid Cells by Increasing G-CSF Expression in Mesenchymal Stem Cells. Front Cell Dev Biol 2022; 10:779159. [PMID: 35372343 PMCID: PMC8969869 DOI: 10.3389/fcell.2022.779159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 02/23/2022] [Indexed: 11/13/2022] Open
Abstract
Rapamycin, also known as sirolimus, an inhibitor of mammalian target of rapamycin (mTOR), is a regulatory kinase responsible for multiple signal transduction pathways. Although rapamycin has been widely used in treating various hematologic diseases, the effects of rapamycin are still not fully understood. Here we found that both oral and intraperitoneal administration of rapamycin led to the expansion of myeloid lineage, while intraperitoneal administration of rapamycin impaired granulocyte differentiation in mice. Rapamycin induced bone marrow mesenchymal stem cells to produce more G-CSF in vitro and in vivo, and promoted the myeloid cells expansion. Our results thus demonstrated that intraperitoneal administration of rapamycin might promote the expansion of myeloid lineage while impair myeloid cell differentiation in vivo.
Collapse
Affiliation(s)
- Minghao Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Shanghai Blood Center, Shanghai, China
| | - Yanjie Lan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Juan Gao
- Tianjin Eye Hospital, Tianjin Key Laboratory of Ophthalmology and Visual Science, Tianjin Eye Institute, Clinical College of Ophthalmology, Tianjin Medical University, Nankai University Affiliated Eye Hospital, Tianjin, China
| | - Shengnan Yuan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Shuaibing Hou
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Tengxiao Guo
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Fei Zhao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yuxia Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Weiping Yuan
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Xiaomin Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- *Correspondence: Xiaomin Wang,
| |
Collapse
|
4
|
Chen S, Lu XT, He TT, Yishake D, Tan XY, Hou MJ, Luo Y, Long JA, Tang ZH, Zhong RH, Fang AP, Zhu HL. Betaine Delayed Muscle Loss by Attenuating Samtor Complex Inhibition for mTORC1 Signaling Via Increasing SAM Level. Mol Nutr Food Res 2021; 65:e2100157. [PMID: 34061446 DOI: 10.1002/mnfr.202100157] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 04/29/2021] [Indexed: 01/26/2023]
Abstract
SCOPE The muscle loss during aging results from the blunt of protein synthesis and poses threat to the elderly health. This study aims to investigate whether betaine affects muscle loss by improving protein synthesis. METHODS AND RESULTS Male C57BL/6J mice are raised from age 12 or 15 months. Mice are fed with AIN-93M diet without or with 2% w/v betaine in distilled water as control group or betaine intervention group (Bet), respectively. Betaine supplementation to mice demonstrates better body composition, grip strength, and motor function. Muscle morphology upregulates expression of myogenic regulate factors, and elevates myosin heavy chain and also improves in Bet group. Betaine promotes muscle protein synthesis via tethering mammalian target of rapamycin complex1 protein kinase (mTORC1) on the lysosomal membrane thereby activating mTORC1 signaling. All these effects aforementioned are time-dependent (p < 0.05). Ultrahigh-performance liquid chromatography results show that betaine increases S-adenosyl-l-methionine (SAM) via methionine cycle. SAM sensor-Samtor-overexpression in C2C12 cells could displace mTORC1 from lysosome thereby inhibiting the mTORC1 signaling. Addition of betaine attenuates this inhibition by increasing SAM level and then disrupting interaction of Samtor complex. CONCLUSIONS These observations indicate that betaine could promisingly promote protein synthesis to delay age-related muscle loss.
Collapse
Affiliation(s)
- Si Chen
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, China
| | - Xiao-Ting Lu
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, China
| | - Tong-Tong He
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, China
| | - Dinuerguli Yishake
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, China
| | - Xu-Yin Tan
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, China
| | - Meng-Jun Hou
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, China
| | - Yun Luo
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, China
| | - Jing-An Long
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, China
| | - Zhi-Hong Tang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, China
| | - Rong-Huan Zhong
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, China
| | - Ai-Ping Fang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, China
| | - Hui-Lian Zhu
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, China
| |
Collapse
|
5
|
D’Hulst G, Masschelein E, De Bock K. Dampened Muscle mTORC1 Response Following Ingestion of High-Quality Plant-Based Protein and Insect Protein Compared to Whey. Nutrients 2021; 13:1396. [PMID: 33919313 PMCID: PMC8143359 DOI: 10.3390/nu13051396] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/09/2021] [Accepted: 04/14/2021] [Indexed: 12/19/2022] Open
Abstract
Increased amino acid availability acutely stimulates protein synthesis partially via activation of mechanistic target of rapamycin complex 1 (mTORC1). Plant-and insect-based protein sources matched for total protein and/or leucine to animal proteins induce a lower postprandial rise in amino acids, but their effects on mTOR activation in muscle are unknown. C57BL/6J mice were gavaged with different protein solutions: whey, a pea-rice protein mix matched for total protein or leucine content to whey, worm protein matched for total protein, or saline. Blood was drawn 30, 60, 105 and 150 min after gavage and muscle samples were harvested 60 min and 150 min after gavage to measure key components of the mTORC1 pathway. Ingestion of plant-based proteins induced a lower rise in blood leucine compared to whey, which coincided with a dampened mTORC1 activation, both acutely and 150 min after administration. Matching total leucine content to whey did not rescue the reduced rise in plasma amino acids, nor the lower increase in mTORC1 compared to whey. Insect protein elicits a similar activation of downstream mTORC1 kinases as plant-based proteins, despite lower postprandial aminoacidemia. The mTORC1 response following ingestion of high-quality plant-based and insect proteins is dampened compared to whey in mouse skeletal muscle.
Collapse
Affiliation(s)
- Gommaar D’Hulst
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, 8603 Zurich, Switzerland; (E.M.); (K.D.B.)
- Laboratory of Regenerative and Movement Biology, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, 8093 Zurich, Switzerland
| | - Evi Masschelein
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, 8603 Zurich, Switzerland; (E.M.); (K.D.B.)
| | - Katrien De Bock
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, 8603 Zurich, Switzerland; (E.M.); (K.D.B.)
| |
Collapse
|
6
|
Torres W, Nava M, Galbán N, Gómez Y, Morillo V, Rojas M, Cano C, Chacín M, D Marco L, Herazo Y, Velasco M, Bermúdez V, Rojas-Quintero J. Anti-Aging Effect of Metformin: A Molecular and Therapeutical Perspective. Curr Pharm Des 2021; 26:4496-4508. [PMID: 32674728 DOI: 10.2174/1381612826666200716161610] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 06/09/2020] [Indexed: 12/25/2022]
Abstract
Aging is a time-dependent inevitable process, in which cellular homeostasis is affected, which has an impact on tissue function. This represents a risk factor for the development of numerous non-transmissible diseases. In consequence, the scientific community continues to search for therapeutic measures capable of improving quality of life and delaying cellular aging. At the center of this research is metformin, a widely used drug in Type 2 Diabetes Mellitus treatment that has a reduced adverse effects profile. Furthermore, there is evidence that this drug has beneficial health effects that go beyond its anti-hyperglycemic properties. Among these effects, its geronto-protection capability stands out. There is growing evidence that points out to an increased life expectancy as well as the quality of life in model organisms treated with metformin. Therefore, there is an abundance of research centered on elucidating the mechanism through which metformin has its anti-aging effects. Among these, the AMPK, mTORC1, SIRT1, FOXO, NF.kB, and DICER1 pathways can be mentioned. Furthermore, studies have highlighted the possibility of a role for the gut microbiome in these processes. The next step is the design of clinical essays that have as a goal evaluating the efficacy and safety of metformin as an anti-aging drug in humans to create a paradigm in the medical horizon. The question being if metformin is, in fact, the new antiaging therapy in humans?
Collapse
Affiliation(s)
- Wheeler Torres
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Manuel Nava
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Nestor Galbán
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Yosselin Gómez
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Valery Morillo
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Milagros Rojas
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Clímaco Cano
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Maricarmen Chacín
- Universidad Simón Bolívar, Facultad de Ciencias de la Salud, Barranquilla, Colombia
| | - Luis D Marco
- Hospital Clínico Universitario, INCLIVA, Nephrology Department, Valencia, España
| | - Yaneth Herazo
- Universidad Simón Bolívar, Facultad de Ciencias de la Salud, Barranquilla, Colombia
| | - Manuel Velasco
- Clinical Pharmacologic Unit, Vargas School of Medicine, Universidad Central de Venezuela, Caracas,
Venezuela
| | - Valmore Bermúdez
- Universidad Simón Bolívar, Facultad de Ciencias de la Salud, Barranquilla, Colombia
| | - Joselyn Rojas-Quintero
- Pulmonary and Critical Care Medicine Department, Brigham and Women’s Hospital, Harvard Medical School,
Boston, MA 02115, USA
| |
Collapse
|
7
|
Landh E, M Moir L, Bradbury P, Traini D, M Young P, Ong HX. Properties of rapamycin solid lipid nanoparticles for lymphatic access through the lungs & part I: the effect of size. Nanomedicine (Lond) 2020; 15:1927-1945. [PMID: 32820673 DOI: 10.2217/nnm-2020-0077] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background: Lymphangioleiomyomatosis (LAM) is characterized by growth of smooth muscle-like cells in the lungs that spread to other organs via lymphatic vessels. Current oral rapamycin treatment is limited by low bioavailability of approximately 15%. Aim: The effect of inhaled rapamycin solid lipid nanoparticles (Rapa-SLNs) size on its penetration through the lymphatics. Method: Three Rapa-SLN formulations (200-1000 nm) were produced and assessed for particle characteristics and further for toxicity and performance in vitro. Results: Rapa-SLNs of 200 nm inhibited proliferation in TSC2-negative mouse embryonic fibroblast cells and penetrated the respiratory epithelium and lymphatic endothelium significantly faster compared with free rapamycin and larger Rapa-SLNs. Conclusion: Rapa-SLN approximately 200 nm allows efficient entry of rapamycin into the lymphatic system and is therefore a promising treatment for LAM patients.
Collapse
Affiliation(s)
- Emelie Landh
- Respiratory Technology, Woolcock Institute of Medical Research, Glebe, NSW, 2037, Australia.,Discipline of Pharmacology, Faculty of Medicine & Health, Sydney, 2006, Australia
| | - Lyn M Moir
- Respiratory Technology, Woolcock Institute of Medical Research, Glebe, NSW, 2037, Australia.,Discipline of Pharmacology, Faculty of Medicine & Health, Sydney, 2006, Australia
| | - Peta Bradbury
- Respiratory Technology, Woolcock Institute of Medical Research, Glebe, NSW, 2037, Australia
| | - Daniela Traini
- Respiratory Technology, Woolcock Institute of Medical Research, Glebe, NSW, 2037, Australia.,Discipline of Pharmacology, Faculty of Medicine & Health, Sydney, 2006, Australia
| | - Paul M Young
- Respiratory Technology, Woolcock Institute of Medical Research, Glebe, NSW, 2037, Australia.,Discipline of Pharmacology, Faculty of Medicine & Health, Sydney, 2006, Australia
| | - Hui Xin Ong
- Respiratory Technology, Woolcock Institute of Medical Research, Glebe, NSW, 2037, Australia.,Discipline of Pharmacology, Faculty of Medicine & Health, Sydney, 2006, Australia
| |
Collapse
|
8
|
Abstract
Organismal aging is accompanied by a host of progressive metabolic alterations and an accumulation of senescent cells, along with functional decline and the appearance of multiple diseases. This implies that the metabolic features of cell senescence may contribute to the organism’s metabolic changes and be closely linked to age-associated diseases, especially metabolic syndromes. However, there is no clear understanding of senescent metabolic characteristics. Here, we review key metabolic features and regulators of cellular senescence, focusing on mitochondrial dysfunction and anabolic deregulation, and their link to other senescence phenotypes and aging. We further discuss the mechanistic involvement of the metabolic regulators mTOR, AMPK, and GSK3, proposing them as key metabolic switches for modulating senescence.
Collapse
Affiliation(s)
- So Mee Kwon
- Departments of Biochemistry, Ajou University School of Medicine, Suwon 16499, Korea
| | - Sun Mi Hong
- Departments of Biochemistry and Biomedical Sciences (BK21 Plus), Ajou University School of Medicine, Suwon 16499, Korea
| | - Young-Kyoung Lee
- Departments of Biochemistry, Ajou University School of Medicine, Suwon 16499, Korea
| | - Seongki Min
- Departments of Biochemistry and Biomedical Sciences (BK21 Plus), Ajou University School of Medicine, Suwon 16499, Korea
| | - Gyesoon Yoon
- Departments of Biochemistry and Biomedical Sciences (BK21 Plus), Ajou University School of Medicine, Suwon 16499, Korea
| |
Collapse
|
9
|
Lei HW, Cai J, Li CM, Yang F, Shi WQ, Shi WQ, Wang LP, Feng YY. Rapamycin Combi with TAE on the Growth, Metastasis, and Prognosis of Hepatocellular Carcinoma in Rat Models. Ann Hepatol 2018; 17:645-654. [PMID: 29893708 DOI: 10.5604/01.3001.0012.0948] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
INTRODUCTION AND AIM To investigate the effect of mTOR inhibitor Rapamycin combined with transcatheter arterial embolization (TAE) on the growth, metastasis, and prognosis of hepatocellular carcinoma (HCC) in rat model. MATERIAL AND METHOD McARH7777 cells were used to construct rat models of HCC, which were randomly divided into Model, Rapamycin, TAE, and Rapamycin + TAE groups. Quantitative reverse transcription-PCR (qRT-PCR) and Western Blot were used to detect the expression of Epithelial-Mesenchymal Transition (EMT)-related molecules, and immunohistochemical staining to determine the expression of EMTrelated proteins, angiogenic factors as well as microvessel density (MVD)-CD34. RESULTS The hepatic tumor volume of rats in the other three groups were all significantly smaller than the Model group on the 7th, 14th, and 21st day after treatment and the combination treatment was apparently more effective than either treatment alone. Besides, both the number and the size of metastatic nodules of HCC rats after combination treatment were remarkably reduced. In addition, compared with rats in the Rapamycin + TAE group, N-cadherin, Vimentin, HIF-1α, VEGF, and MVD-CD34 were obviously enhanced, while E-cadherin was lowered in those TAE group, which were the complete opposite to the Rapamycin group. Besides, the median survival time of rats in the Rapamycin + TAE group was evidently longer than the resting groups. CONCLUSION Rapamycin combined with TAE may effectively suppress the EMT formation and angiogenesis, thereby inhibiting the growth and lung metastasis of HCC rats, which provides a new idea for countering the recurrence and metastasis of HCC.
Collapse
Affiliation(s)
- Hong-Wei Lei
- Department of Interventional Radiology, the First People's Hospital of Jingzhou, Jingzhou, Hubei, P.R. China First Hospital affiliated to Yangtze University, Jingzhou, Hubei Province, China
| | - Jie Cai
- Department of Interventional Radiology, the First People's Hospital of Jingzhou, Jingzhou, Hubei, P.R. China First Hospital affiliated to Yangtze University, Jingzhou, Hubei Province, China
| | - Cheng-Ming Li
- Department of Interventional Radiology, the First People's Hospital of Jingzhou, Jingzhou, Hubei, P.R. China First Hospital affiliated to Yangtze University, Jingzhou, Hubei Province, China
| | - Fang Yang
- Department of Interventional Radiology, the First People's Hospital of Jingzhou, Jingzhou, Hubei, P.R. China First Hospital affiliated to Yangtze University, Jingzhou, Hubei Province, China
| | - Wan-Qing Shi
- Department of Interventional Radiology, the First People's Hospital of Jingzhou, Jingzhou, Hubei, P.R. China First Hospital affiliated to Yangtze University, Jingzhou, Hubei Province, China
| | - Wan-Qing Shi
- Department of Interventional Radiology, the First People's Hospital of Jingzhou, Jingzhou, Hubei, P.R. China First Hospital affiliated to Yangtze University, Jingzhou, Hubei Province, China
| | - Li-Ping Wang
- Department of Interventional Radiology, the First People's Hospital of Jingzhou, Jingzhou, Hubei, P.R. China First Hospital affiliated to Yangtze University, Jingzhou, Hubei Province, China
| | - You-Ying Feng
- Department of Central Sterile Supply, the First People's Hospital of Jingzhou, Jingzhou, Hubei, P.R. China First Hospital affiliated to Yangtze University, Jingzhou, Hubei Province, China
| |
Collapse
|
10
|
|
11
|
Esnault S, Shen ZJ, Malter JS. Protein Translation and Signaling in Human Eosinophils. Front Med (Lausanne) 2017; 4:150. [PMID: 28971096 PMCID: PMC5609579 DOI: 10.3389/fmed.2017.00150] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 09/01/2017] [Indexed: 01/01/2023] Open
Abstract
We have recently reported that, unlike IL-5 and GM-CSF, IL-3 induces increased translation of a subset of mRNAs. In addition, we have demonstrated that Pin1 controls the activity of mRNA binding proteins, leading to enhanced mRNA stability, GM-CSF protein production and prolonged eosinophil (EOS) survival. In this review, discussion will include an overview of cap-dependent protein translation and its regulation by intracellular signaling pathways. We will address the more general process of mRNA post-transcriptional regulation, especially regarding mRNA binding proteins, which are critical effectors of protein translation. Furthermore, we will focus on (1) the roles of IL-3-driven sustained signaling on enhanced protein translation in EOS, (2) the mechanisms regulating mRNA binding proteins activity in EOS, and (3) the potential targeting of IL-3 signaling and the signaling leading to mRNA binding activity changes to identify therapeutic targets to treat EOS-associated diseases.
Collapse
Affiliation(s)
- Stephane Esnault
- Department of Medicine, Allergy, Pulmonary, and Critical Care Medicine Division, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, United States
| | - Zhong-Jian Shen
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - James S Malter
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
12
|
Shao P, Ma L, Ren Y, Liu H. Modulation of the immune response in rheumatoid arthritis with strategically released rapamycin. Mol Med Rep 2017; 16:5257-5262. [PMID: 28849205 PMCID: PMC5647081 DOI: 10.3892/mmr.2017.7285] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 06/27/2017] [Indexed: 01/01/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease, which is associated with symptoms, including synovial membrane inflammatory pain, joint synovitis and stiffness. However, there are no effective methods available to cure this disease. In the present study, rapamycin was used to modulate immunity in RA. To limit the cytotoxicity of rapamycin, rapamycin was loaded into well-characterized biocompatible nanoparticles. In vitro, rapamycin particles downregulated the activation of dendritic cell surface markers, including CD80+ and CD40+, upon interacting with macrophages. The rapamycin particles reduced the secretion of inflammatory cytokines, including interleukin (IL)-6, tumor necrosis factor (TNF) and IL-1β, which are characteristic of RA. In vivo, the rapamycin particles decreased the symptoms of RA in mice, and the production of inflammatory cytokines was associated with the occurrence of RA. The present study partially revealed the interactions between rapamycin and two types of immune cell in RA disease, and may potentially offer a solution to improve the treatment of RA.
Collapse
Affiliation(s)
- Ping Shao
- Department of Rheumatology, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, P.R. China
| | - Linxiao Ma
- Department of Rheumatology, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, P.R. China
| | - Yile Ren
- Department of Rheumatology, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, P.R. China
| | - Huijie Liu
- Department of Rheumatology, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, P.R. China
| |
Collapse
|
13
|
Abstract
The mammalian target of rapamycin, mTOR, plays key roles in cell growth and proliferation, acting at the catalytic subunit of two protein kinase complexes: mTOR complexes 1 and 2 (mTORC1/2). mTORC1 signaling is switched on by several oncogenic signaling pathways and is accordingly hyperactive in the majority of cancers. Inhibiting mTORC1 signaling has therefore attracted great attention as an anti-cancer therapy. However, progress in using inhibitors of mTOR signaling as therapeutic agents in oncology has been limited by a number of factors, including the fact that the classic mTOR inhibitor, rapamycin, inhibits only some of the effects of mTOR; the existence of several feedback loops; and the crucial importance of mTOR in normal physiology.
Collapse
Affiliation(s)
- Jianling Xie
- Nutrition and Metabolism, South Australian Health and Medical research Institute, Adelaide, SA, Australia
| | - Xuemin Wang
- Nutrition and Metabolism, South Australian Health and Medical research Institute, Adelaide, SA, Australia; School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| | - Christopher G Proud
- Nutrition and Metabolism, South Australian Health and Medical research Institute, Adelaide, SA, Australia; School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia
| |
Collapse
|
14
|
Chronic rapamycin treatment on the nutrient utilization and metabolism of juvenile turbot (Psetta maxima). Sci Rep 2016; 6:28068. [PMID: 27305975 PMCID: PMC4910097 DOI: 10.1038/srep28068] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 05/31/2016] [Indexed: 12/04/2022] Open
Abstract
High dietary protein inclusion is necessary in fish feeds and also represents a major cost in the aquaculture industry, which demands improved dietary conversion into body proteins in fish. In mammals, the target of rapamycin (TOR) is a key nutritionally responsive molecule governing postprandial anabolism. However, its physiological significance in teleosts has not been fully examined. In the present study, we examined the nutritional physiology of turbot after chronic rapamycin inhibition. Our results showed that a 6-week inhibition of TOR using dietary rapamycin inclusion (30 mg/kg diet) reduced growth performance and feed utilization. The rapamycin treatment inhibited TOR signaling and reduced expression of key enzymes in glycolysis, lipogenesis, cholesterol biosynthesis, while increasing the expression of enzymes involved in gluconeogenesis. Furthermore, rapamycin treatment increased intestinal goblet cell number in turbot, while the expressions of Notch and Hes1 were down regulated. It was possible that stimulated goblet cell differentiation by rapamycin was mediated through Notch-Hes1 pathway. Therefore, our results demonstrate the important role of TOR signaling in fish nutritional physiology.
Collapse
|
15
|
Granata S, Dalla Gassa A, Carraro A, Brunelli M, Stallone G, Lupo A, Zaza G. Sirolimus and Everolimus Pathway: Reviewing Candidate Genes Influencing Their Intracellular Effects. Int J Mol Sci 2016; 17:ijms17050735. [PMID: 27187382 PMCID: PMC4881557 DOI: 10.3390/ijms17050735] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 04/21/2016] [Accepted: 05/06/2016] [Indexed: 02/07/2023] Open
Abstract
Sirolimus (SRL) and everolimus (EVR) are mammalian targets of rapamycin inhibitors (mTOR-I) largely employed in renal transplantation and oncology as immunosuppressive/antiproliferative agents. SRL was the first mTOR-I produced by the bacterium Streptomyces hygroscopicus and approved for several medical purposes. EVR, derived from SRL, contains a 2-hydroxy-ethyl chain in the 40th position that makes the drug more hydrophilic than SRL and increases oral bioavailability. Their main mechanism of action is the inhibition of the mTOR complex 1 and the regulation of factors involved in a several crucial cellular functions including: protein synthesis, regulation of angiogenesis, lipid biosynthesis, mitochondrial biogenesis and function, cell cycle, and autophagy. Most of the proteins/enzymes belonging to the aforementioned biological processes are encoded by numerous and tightly regulated genes. However, at the moment, the polygenic influence on SRL/EVR cellular effects is still not completely defined, and its comprehension represents a key challenge for researchers. Therefore, to obtain a complete picture of the cellular network connected to SRL/EVR, we decided to review major evidences available in the literature regarding the genetic influence on mTOR-I biology/pharmacology and to build, for the first time, a useful and specific “SRL/EVR genes-focused pathway”, possibly employable as a starting point for future in-depth research projects.
Collapse
Affiliation(s)
- Simona Granata
- Renal Unit, Department of Medicine, University/Hospital of Verona, 37126 Verona, Italy.
| | | | - Amedeo Carraro
- Liver Transplant Unit, Department of General Surgery and Odontoiatrics, University/Hospital of Verona, 37126 Verona, Italy.
| | - Matteo Brunelli
- Department of Pathology and Diagnostics, University of Verona, Azienda Ospedaliera Universitaria Integrata, 37126 Verona, Italy.
| | - Giovanni Stallone
- Nephrology, Dialysis and Transplantation Unit, University of Foggia, 71122 Foggia, Italy.
| | - Antonio Lupo
- Renal Unit, Department of Medicine, University/Hospital of Verona, 37126 Verona, Italy.
| | - Gianluigi Zaza
- Renal Unit, Department of Medicine, University/Hospital of Verona, 37126 Verona, Italy.
| |
Collapse
|
16
|
Hussein NA, Delaney TL, Tounsel BL, Liebl FLW. The Extracellular-Regulated Kinase Effector Lk6 is Required for Glutamate Receptor Localization at the Drosophila Neuromuscular Junction. J Exp Neurosci 2016; 10:77-91. [PMID: 27199570 PMCID: PMC4866800 DOI: 10.4137/jen.s32840] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 03/14/2016] [Accepted: 03/18/2016] [Indexed: 11/16/2022] Open
Abstract
The proper localization and synthesis of postsynaptic glutamate receptors are essential for synaptic plasticity. Synaptic translation initiation is thought to occur via the target of rapamycin (TOR) and mitogen-activated protein kinase signal-integrating kinase (Mnk) signaling pathways, which is downstream of extracellular-regulated kinase (ERK). We used the model glutamatergic synapse, the Drosophila neuromuscular junction, to better understand the roles of the Mnk and TOR signaling pathways in synapse development. These synapses contain non-NMDA receptors that are most similar to AMPA receptors. Our data show that Lk6, the Drosophila homolog of Mnk1 and Mnk2, is required in either presynaptic neurons or postsynaptic muscle for the proper localization of the GluRIIA glutamate receptor subunit. Lk6 may signal through eukaryotic initiation factor (eIF) 4E to regulate the synaptic levels of GluRIIA as either interfering with eIF4E binding to eIF4G or expression of a nonphosphorylatable isoform of eIF4E resulted in a significant reduction in GluRIIA at the synapse. We also find that Lk6 and TOR may independently regulate synaptic levels of GluRIIA.
Collapse
Affiliation(s)
- Nizar A Hussein
- Department of Biological Sciences, Southern Illinois University Edwardsville, Edwardsville, IL, USA
| | - Taylor L Delaney
- Department of Biological Sciences, Southern Illinois University Edwardsville, Edwardsville, IL, USA
| | - Brittany L Tounsel
- Department of Biological Sciences, Southern Illinois University Edwardsville, Edwardsville, IL, USA
| | - Faith L W Liebl
- Department of Biological Sciences, Southern Illinois University Edwardsville, Edwardsville, IL, USA
| |
Collapse
|
17
|
VCP and ATL1 regulate endoplasmic reticulum and protein synthesis for dendritic spine formation. Nat Commun 2016; 7:11020. [PMID: 26984393 PMCID: PMC4800434 DOI: 10.1038/ncomms11020] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 02/12/2016] [Indexed: 12/29/2022] Open
Abstract
Imbalanced protein homeostasis, such as excessive protein synthesis and protein aggregation, is a pathogenic hallmark of a range of neurological disorders. Here, using expression of mutant proteins, a knockdown approach and disease mutation knockin mice, we show that VCP (valosin-containing protein), together with its cofactor P47 and the endoplasmic reticulum (ER) morphology regulator ATL1 (Atlastin-1), regulates tubular ER formation and influences the efficiency of protein synthesis to control dendritic spine formation in neurons. Strengthening the significance of protein synthesis in dendritic spinogenesis, the translation blocker cyclohexamide and the mTOR inhibitor rapamycin reduce dendritic spine density, while a leucine supplement that increases protein synthesis ameliorates the dendritic spine defects caused by Vcp and Atl1 deficiencies. Because VCP and ATL1 are the causative genes of several neurodegenerative and neurodevelopmental disorders, we suggest that impaired ER formation and inefficient protein synthesis are significant in the pathogenesis of multiple neurological disorders. Protein homeostasis is crucial for maintaining a variety of cellular functions. Here the authors show that valosin-containing protein and its cofactors regulate tubular ER formation and protein synthesis efficiency, thereby control dendritic spine formation in neurons.
Collapse
|
18
|
Rapamycin inhibits mSin1 phosphorylation independently of mTORC1 and mTORC2. Oncotarget 2015; 6:4286-98. [PMID: 25738366 PMCID: PMC4414190 DOI: 10.18632/oncotarget.3006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 12/21/2014] [Indexed: 02/05/2023] Open
Abstract
Current knowledge indicates that the mammalian target of rapamycin (mTOR) functions as two complexes, mTORC1 and mTORC2, regulating cell growth, proliferation, survival, differentiation, and motility. Recently mSin1 has been identified as a critical component of mTORC2, which is essential for phosphorylation of Akt and other signaling molecules. Studies have shown that rapamycin inhibits phosphorylation of mSin1. However, the underlying mechanism is unknown. Here we found that rapamycin inhibited phosphorylation of mSin1 potently and rapidly. Expression of rapamycin-resistant mutant of mTOR (mTOR-T), but not rapamycin-resistant and kinase dead mutant of mTOR (mTOR-TE), prevented rapamycin from inhibiting mSin1 phosphorylation, suggesting that rapamycin-induced dephosphorylation of mSin1 is mTOR-dependent. Surprisingly, ectopic expression of rapamycin-resistant and constitutively active p70 S6 kinase 1 (S6K1) did not confer resistance to rapamycin-induced dephosphorylation of mSin1. Furthermore, disruption of mTORC1 and mTORC2 by silencing raptor and rictor, respectively, or downregulation of S6K1 or Akt did not induce the dephosphorylation of mSin1 as rapamycin did. However, silencing mTOR or mLST8 mimicked the effect of rapamycin, inhibiting mSin1 phosphorylation. Our findings suggest that rapamycin inhibits mSin1 phosphorylation, which is independent of mTORC1 and mTORC2, but is possibly dependent on a new mTOR complex, which at least contains mTOR and mLST8.
Collapse
|
19
|
Leung EY, Askarian-Amiri M, Finlay GJ, Rewcastle GW, Baguley BC. Potentiation of Growth Inhibitory Responses of the mTOR Inhibitor Everolimus by Dual mTORC1/2 Inhibitors in Cultured Breast Cancer Cell Lines. PLoS One 2015; 10:e0131400. [PMID: 26148118 PMCID: PMC4492962 DOI: 10.1371/journal.pone.0131400] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Accepted: 06/01/2015] [Indexed: 11/19/2022] Open
Abstract
The mammalian target of rapamycin (mTOR), a vital component of signaling pathways involving PI3K/AKT, is an attractive therapeutic target in breast cancer. Everolimus, an allosteric mTOR inhibitor that inhibits the mTOR functional complex mTORC1, is approved for treatment of estrogen receptor positive (ER+) breast cancer. Other mTOR inhibitors show interesting differences in target specificities: BEZ235 and GSK2126458 are ATP competitive mTOR inhibitors targeting both PI3K and mTORC1/2; AZD8055, AZD2014 and KU-0063794 are ATP competitive mTOR inhibitors targeting both mTORC1 and mTORC2; and GDC-0941 is a pan-PI3K inhibitor. We have addressed the question of whether mTOR inhibitors may be more effective in combination than singly in inhibiting the proliferation of breast cancer cells. We selected a panel of 30 human breast cancer cell lines that included ER and PR positive, HER2 over-expressing, and “triple negative” variants, and determined whether signaling pathway utilization was related to drug-induced inhibition of proliferation. A significant correlation (p = 0.005) was found between everolimus IC50 values and p70S6K phosphorylation, but not with AKT or ERK phosphorylation, consistent with the mTOR pathway being a principal target. We then carried out combination studies with four everolimus resistant triple-negative breast cancer cell lines, and found an unexpectedly high degree of synergy between everolimus and the other inhibitors tested. The level of potentiation of everolimus inhibitory activity (measured by IC50 values) was found to be cell line-specific for all the kinase inhibitors tested. The results suggest that judicious combination of mTOR inhibitors with different modes of action could have beneficial effects in the treatment of breast cancer.
Collapse
Affiliation(s)
- Euphemia Y. Leung
- Auckland Cancer Society Research Centre, University of Auckland, Grafton, Auckland, New Zealand
- Department of Molecular Medicine and Pathology, University of Auckland, Grafton, Auckland, New Zealand
- * E-mail: (EL); (BB)
| | - Marjan Askarian-Amiri
- Auckland Cancer Society Research Centre, University of Auckland, Grafton, Auckland, New Zealand
- Department of Molecular Medicine and Pathology, University of Auckland, Grafton, Auckland, New Zealand
| | - Graeme J. Finlay
- Auckland Cancer Society Research Centre, University of Auckland, Grafton, Auckland, New Zealand
- Department of Molecular Medicine and Pathology, University of Auckland, Grafton, Auckland, New Zealand
| | - Gordon W. Rewcastle
- Auckland Cancer Society Research Centre, University of Auckland, Grafton, Auckland, New Zealand
| | - Bruce C. Baguley
- Auckland Cancer Society Research Centre, University of Auckland, Grafton, Auckland, New Zealand
- * E-mail: (EL); (BB)
| |
Collapse
|
20
|
Tischner C, Hofer A, Wulff V, Stepek J, Dumitru I, Becker L, Haack T, Kremer L, Datta AN, Sperl W, Floss T, Wurst W, Chrzanowska-Lightowlers Z, De Angelis MH, Klopstock T, Prokisch H, Wenz T. MTO1 mediates tissue specificity of OXPHOS defects via tRNA modification and translation optimization, which can be bypassed by dietary intervention. Hum Mol Genet 2015; 24:2247-66. [PMID: 25552653 PMCID: PMC4380071 DOI: 10.1093/hmg/ddu743] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 12/12/2014] [Accepted: 12/22/2014] [Indexed: 11/15/2022] Open
Abstract
Mitochondrial diseases often exhibit tissue-specific pathologies, but this phenomenon is poorly understood. Here we present regulation of mitochondrial translation by the Mitochondrial Translation Optimization Factor 1, MTO1, as a novel player in this scenario. We demonstrate that MTO1 mediates tRNA modification and controls mitochondrial translation rate in a highly tissue-specific manner associated with tissue-specific OXPHOS defects. Activation of mitochondrial proteases, aberrant translation products, as well as defects in OXPHOS complex assembly observed in MTO1 deficient mice further imply that MTO1 impacts translation fidelity. In our mouse model, MTO1-related OXPHOS deficiency can be bypassed by feeding a ketogenic diet. This therapeutic intervention is independent of the MTO1-mediated tRNA modification and involves balancing of mitochondrial and cellular secondary stress responses. Our results thereby establish mammalian MTO1 as a novel factor in the tissue-specific regulation of OXPHOS and fine tuning of mitochondrial translation accuracy.
Collapse
Affiliation(s)
- Christin Tischner
- Institute for Genetics and Cluster of Excellence: Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Zülpicher Str. 47A, Cologne 50674, Germany
| | - Annette Hofer
- Institute for Genetics and Cluster of Excellence: Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Zülpicher Str. 47A, Cologne 50674, Germany
| | - Veronika Wulff
- Institute for Genetics and Cluster of Excellence: Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Zülpicher Str. 47A, Cologne 50674, Germany
| | - Joanna Stepek
- Institute for Genetics and Cluster of Excellence: Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Zülpicher Str. 47A, Cologne 50674, Germany
| | - Iulia Dumitru
- Institute for Genetics and Cluster of Excellence: Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Zülpicher Str. 47A, Cologne 50674, Germany
| | - Lore Becker
- Department of Neurology, Friedrich-Baur-Institute, Ludwig-Maximilians-University, Munich 80336, Germany, German Mouse Clinic, Institute of Experimental Genetics
| | - Tobias Haack
- Institute of Human Genetics, German Network for Mitochondrial Disorders (mitoNET), Germany
| | - Laura Kremer
- Institute of Human Genetics, German Network for Mitochondrial Disorders (mitoNET), Germany
| | - Alexandre N Datta
- Division of Neuropediatrics and Developmental Medicine, University Children's Hospital Basel (UKBB), University of Basel, Basel 4031, Switzerland
| | - Wolfgang Sperl
- German Network for Mitochondrial Disorders (mitoNET), Germany, Department of Pediatrics, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Thomas Floss
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environment and Health (GmbH), Neuherberg 85764, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environment and Health (GmbH), Neuherberg 85764, Germany, Technical University Munich, Helmholtz Zentrum München, Neuherberg 85764, Germany, DZNE-German Center for Neurodegenerative Diseases, Munich, Germany, Max Planck Institute of Psychiatry, Munich 80804, Germany, German Center for Vertigo and Balance Disorders, Munich, Germany
| | - Zofia Chrzanowska-Lightowlers
- The Wellcome Trust Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, The Medical School, Newcastle upon Tyne NE2 4HH, UK
| | - Martin Hrabe De Angelis
- German Mouse Clinic, Institute of Experimental Genetics, German Center for Vertigo and Balance Disorders, Munich, Germany, Center of Life and Food Sciences Weihenstephan, Technische Universitat München, Freising 85350, Germany, German Center for Diabetes Research (DZD), Neuherberg 85764, Germany and Technische Universität München, Freising-Weihenstephan 85354, Germany
| | - Thomas Klopstock
- Department of Neurology, Friedrich-Baur-Institute, Ludwig-Maximilians-University, Munich 80336, Germany, German Mouse Clinic, Institute of Experimental Genetics, German Network for Mitochondrial Disorders (mitoNET), Germany, DZNE-German Center for Neurodegenerative Diseases, Munich, Germany, German Center for Vertigo and Balance Disorders, Munich, Germany
| | - Holger Prokisch
- Institute of Human Genetics, Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environment and Health (GmbH), Neuherberg 85764, Germany
| | - Tina Wenz
- Institute for Genetics and Cluster of Excellence: Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Zülpicher Str. 47A, Cologne 50674, Germany, German Network for Mitochondrial Disorders (mitoNET), Germany,
| |
Collapse
|
21
|
Rheb Inhibits Protein Synthesis by Activating the PERK-eIF2α Signaling Cascade. Cell Rep 2015; 10:684-693. [PMID: 25660019 DOI: 10.1016/j.celrep.2015.01.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 12/04/2014] [Accepted: 12/31/2014] [Indexed: 11/20/2022] Open
Abstract
Rheb, a ubiquitous small GTPase, is well known to bind and activate mTOR, which augments protein synthesis. Inhibition of protein synthesis is also physiologically regulated. Thus, with cell stress, the unfolded protein response system leads to phosphorylation of the initiation factor eIF2α and arrest of protein synthesis. We now demonstrate a major role for Rheb in inhibiting protein synthesis by enhancing the phosphorylation of eIF2α by protein kinase-like ER kinase (PERK). Interplay between the stimulatory and inhibitory roles of Rheb may enable cells to modulate protein synthesis in response to varying environmental stresses.
Collapse
|
22
|
Völkers M, Konstandin MH, Doroudgar S, Toko H, Quijada P, Din S, Joyo A, Ornelas L, Samse K, Thuerauf DJ, Gude N, Glembotski CC, Sussman MA. Mechanistic target of rapamycin complex 2 protects the heart from ischemic damage. Circulation 2013; 128:2132-44. [PMID: 24008870 DOI: 10.1161/circulationaha.113.003638] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND The mechanistic target of rapamycin (mTOR) comprises 2 structurally distinct multiprotein complexes, mTOR complexes 1 and 2 (mTORC1 and mTORC2). Deregulation of mTOR signaling occurs during and contributes to the severity of myocardial damage from ischemic heart disease. However, the relative roles of mTORC1 versus mTORC2 in the pathogenesis of ischemic damage are unknown. METHODS AND RESULTS Combined pharmacological and molecular approaches were used to alter the balance of mTORC1 and mTORC2 signaling in cultured cardiac myocytes and in mouse hearts subjected to conditions that mimic ischemic heart disease. The importance of mTOR signaling in cardiac protection was demonstrated by pharmacological inhibition of both mTORC1 and mTORC2 with Torin1, which led to increased cardiomyocyte apoptosis and tissue damage after myocardial infarction. Predominant mTORC1 signaling mediated by suppression of mTORC2 with Rictor similarly increased cardiomyocyte apoptosis and tissue damage after myocardial infarction. In comparison, preferentially shifting toward mTORC2 signaling by inhibition of mTORC1 with PRAS40 led to decreased cardiomyocyte apoptosis and tissue damage after myocardial infarction. CONCLUSIONS These results suggest that selectively increasing mTORC2 while concurrently inhibiting mTORC1 signaling is a novel therapeutic approach for the treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Mirko Völkers
- From SDSU Heart Institute, Department of Biology, San Diego State University, San Diego, CA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Clemens MJ, Elia A, Morley SJ. Requirement for the eIF4E binding proteins for the synergistic down-regulation of protein synthesis by hypertonic conditions and mTOR inhibition. PLoS One 2013; 8:e71138. [PMID: 23940704 PMCID: PMC3733773 DOI: 10.1371/journal.pone.0071138] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 07/02/2013] [Indexed: 11/18/2022] Open
Abstract
The protein kinase mammalian target of rapamycin (mTOR) regulates the phosphorylation and activity of several proteins that have the potential to control translation, including p70S6 kinase and the eIF4E binding proteins 4E-BP1 and 4E-BP2. In spite of this, in exponentially growing cells overall protein synthesis is often resistant to mTOR inhibitors. We report here that sensitivity of wild-type mouse embryonic fibroblasts (MEFs) to mTOR inhibitors can be greatly increased when the cells are subjected to the physiological stress imposed by hypertonic conditions. In contrast, protein synthesis in MEFs with a double knockout of 4E-BP1 and 4E-BP2 remains resistant to mTOR inhibitors under these conditions. Phosphorylation of p70S6 kinase and protein kinase B (Akt) is blocked by the mTOR inhibitor Ku0063794 equally well in both wild-type and 4E-BP knockout cells, under both normal and hypertonic conditions. The response of protein synthesis to hypertonic stress itself does not require the 4E-BPs. These data suggest that under certain stress conditions: (i) translation has a greater requirement for mTOR activity and (ii) there is an absolute requirement for the 4E-BPs for regulation by mTOR. Importantly, dephosphorylation of p70S6 kinase and Akt is not sufficient to affect protein synthesis acutely.
Collapse
Affiliation(s)
- Michael J. Clemens
- Department of Biochemistry and Molecular Biology, School of Life Sciences, University of Sussex, JMS Building, Falmer, Brighton United Kingdom
| | - Androulla Elia
- Division of Biomedical Sciences, St George’s, University of London, Cranmer Terrace, London, United Kingdom
- * E-mail:
| | - Simon J. Morley
- Department of Biochemistry and Molecular Biology, School of Life Sciences, University of Sussex, JMS Building, Falmer, Brighton United Kingdom
| |
Collapse
|
24
|
Metabotropic glutamate receptor I (mGluR1) antagonism impairs cocaine-induced conditioned place preference via inhibition of protein synthesis. Neuropsychopharmacology 2013; 38:1308-21. [PMID: 23348064 PMCID: PMC3656374 DOI: 10.1038/npp.2013.29] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Antagonism of group I metabotropic glutamate receptors (mGluR1 and mGluR5) reduces behavioral effects of drugs of abuse, including cocaine. However, the underlying mechanisms remain poorly understood. Activation of mGluR5 increases protein synthesis at synapses. Although mGluR5-induced excessive protein synthesis has been implicated in the pathology of fragile X syndrome, it remains unknown whether group I mGluR-mediated protein synthesis is involved in any behavioral effects of drugs of abuse. We report that group I mGluR agonist DHPG induced more pronounced initial depression of inhibitory postsynaptic currents (IPSCs) followed by modest long-term depression (I-LTD) in dopamine neurons of rat ventral tegmental area (VTA) through the activation of mGluR1. The early component of DHPG-induced depression of IPSCs was mediated by the cannabinoid CB1 receptors, while DHPG-induced I-LTD was dependent on protein synthesis. Western blotting analysis indicates that mGluR1 was coupled to extracellular signal-regulated kinase (ERK) and mammalian target of rapamycin (mTOR) signaling pathways to increase translation. We also show that cocaine conditioning activated translation machinery in the VTA via an mGluR1-dependent mechanism. Furthermore, intra-VTA microinjections of mGluR1 antagonist JNJ16259685 and protein synthesis inhibitor cycloheximide significantly attenuated or blocked the acquisition of cocaine-induced conditioned place preference (CPP) and activation of translation elongation factors. Taken together, these results suggest that mGluR1 antagonism inhibits de novo protein synthesis; this effect may block the formation of cocaine-cue associations and thus provide a mechanism for the reduction in CPP to cocaine.
Collapse
|
25
|
mTOR: a link from the extracellular milieu to transcriptional regulation of oligodendrocyte development. ASN Neuro 2013; 5:e00108. [PMID: 23421405 PMCID: PMC3601842 DOI: 10.1042/an20120092] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Oligodendrocyte development is controlled by numerous extracellular signals that regulate a series of transcription factors that promote the differentiation of oligodendrocyte progenitor cells to myelinating cells in the central nervous system. A major element of this regulatory system that has only recently been studied is the intracellular signalling from surface receptors to transcription factors to down-regulate inhibitors and up-regulate inducers of oligodendrocyte differentiation and myelination. The current review focuses on one such pathway: the mTOR (mammalian target of rapamycin) pathway, which integrates signals in many cell systems and induces cell responses including cell proliferation and cell differentiation. This review describes the known functions of mTOR as they relate to oligodendrocyte development, and its recently discovered impact on oligodendrocyte differentiation and myelination. A potential model for its role in oligodendrocyte development is proposed.
Collapse
|
26
|
Chang AJ, Sohn R, Lu ZH, Arbeit JM, Lapi SE. Detection of rapalog-mediated therapeutic response in renal cancer xenografts using ⁶⁴Cu-bevacizumab immunoPET. PLoS One 2013; 8:e58949. [PMID: 23516584 PMCID: PMC3597567 DOI: 10.1371/journal.pone.0058949] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Accepted: 02/11/2013] [Indexed: 11/19/2022] Open
Abstract
The importance of neovascularization for primary and metastatic tumor growth fostered numerous clinical trials of angiogenesis inhibitors either alone or in combination with conventional antineoplastic therapies. One challenge with the use of molecularly targeted agents has been the disconnection between size reduction and tumor biologic behavior, either when the drug is efficacious or when tumor resistance emerges. Here, we report the synthesis and characterization of (64)Cu-NOTA-bevacizumab as a PET imaging agent for imaging intratumoral VEGF content in vivo. (64)Cu-NOTA-bevacizumab avidly accumulated in 786-O renal carcinoma xenografts with lower levels in host organs. RAD001 (everolimus) markedly attenuated (64)Cu-NOTA-bevacizumab accumulation within 786-O renal carcinoma xenografts. Tumor tissue and cellular molecular analysis validated PET imaging, demonstrating decreases in total and secreted VEGF content and VEGFR2 activation. Notably, (64)Cu-NOTA-bevacizumab PET imaging was concordant with the growth arrest of RAD001 tumors. These data suggest that immunoPET targeting of angiogenic factors such as VEGF could be a new class of surrogate markers complementing the RECIST criteria in patients receiving molecularly targeted therapies.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal, Humanized/chemistry
- Antibodies, Monoclonal, Humanized/metabolism
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Bevacizumab
- Cell Line, Tumor
- Cell Transformation, Neoplastic
- Copper Radioisotopes
- Everolimus
- Heterocyclic Compounds/chemistry
- Heterocyclic Compounds, 1-Ring
- Humans
- Immunoconjugates
- Kidney Neoplasms/blood supply
- Kidney Neoplasms/diagnostic imaging
- Kidney Neoplasms/pathology
- Kidney Neoplasms/therapy
- Mice
- Neovascularization, Pathologic
- Phosphorylation/drug effects
- Positron-Emission Tomography
- Sirolimus/analogs & derivatives
- Sirolimus/chemistry
- Sirolimus/pharmacology
- Treatment Outcome
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Albert J. Chang
- Department of Radiation Oncology, Washington University, St. Louis, Missouri, United States of America
- Department of Radiology, Washington University, St. Louis, Missouri, United States of America
| | - Rebecca Sohn
- Urology Division, Department of Surgery, Washington University, St. Louis, Missouri, United States of America
| | - Zhi Hong Lu
- Urology Division, Department of Surgery, Washington University, St. Louis, Missouri, United States of America
| | - Jeffrey M. Arbeit
- Urology Division, Department of Surgery, Washington University, St. Louis, Missouri, United States of America
- Siteman Cancer Center, St. Louis, Missouri, United States of America
- * E-mail: (SEL); (JMA)
| | - Suzanne E. Lapi
- Department of Radiology, Washington University, St. Louis, Missouri, United States of America
- Siteman Cancer Center, St. Louis, Missouri, United States of America
- * E-mail: (SEL); (JMA)
| |
Collapse
|
27
|
Caldana C, Li Y, Leisse A, Zhang Y, Bartholomaeus L, Fernie AR, Willmitzer L, Giavalisco P. Systemic analysis of inducible target of rapamycin mutants reveal a general metabolic switch controlling growth in Arabidopsis thaliana. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2013; 73:897-909. [PMID: 23173928 DOI: 10.1111/tpj.12080] [Citation(s) in RCA: 169] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Revised: 11/18/2012] [Accepted: 11/20/2012] [Indexed: 05/19/2023]
Abstract
The target of rapamycin (TOR) pathway is a major regulator of growth in all eukaryotes, integrating energy, nutrient and stress signals into molecular decisions. By using large-scale MS-based metabolite profiling of primary, secondary and lipid compounds in combination with array-based transcript profiling, we show that the TOR protein not only regulates growth but also influences nutrient partitioning and central energy metabolism. The study was performed on plants exhibiting conditional down-regulation of AtTOR expression, revealing strong regulation of genes involved in pathways such as the cell cycle, cell-wall modifications and senescence, together with major changes in transcripts and metabolites of the primary and secondary metabolism. In agreement with these results, our morphological and metabolic analyses disclosed major metabolic changes leading to massive accumulations of storage lipids and starch. The implications of these data in the context of the general role of TOR in eukaryotic systems are discussed in parallel with the plant-specific aspects of TOR function. Finally, we propose a role for harnessing the plant TOR pathway by utilizing it as a potent metabolic switch, offering a possible route for biotechnological optimization of plant energy content and carbon partitioning for the production of bioenergy.
Collapse
Affiliation(s)
- Camila Caldana
- Max Planck Institute of Molecular Plant Physiology, Am Mühlenberg 1, 14476, Potsdam, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Schmitt C, Voegelin M, Marin A, Schmitt M, Schegg F, Hénon P, Guenot D, Tarnus C. Selective aminopeptidase-N (CD13) inhibitors with relevance to cancer chemotherapy. Bioorg Med Chem 2013; 21:2135-44. [PMID: 23428964 DOI: 10.1016/j.bmc.2012.12.038] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Revised: 12/15/2012] [Accepted: 12/21/2012] [Indexed: 11/29/2022]
Abstract
Aminopeptidase-N (APN/CD13) is highly expressed on the surface of numerous types of cancer cells and particularly on the endothelial cells of neoangiogenic vessels during tumourigenesis. This metallo-aminopeptidase has been identified as a potential target for cancer chemotherapy. In this work, we evaluated the efficacy of a novel series of benzosuberone analogues, which were previously reported to be highly potent, selective APN inhibitors with Ki values in the micromolar to sub-nanomolar range. Endothelial cell morphogenesis as well as cell motility were inhibited in vitro in a dose-dependent manner at concentrations that correlated with the potency of the compounds, thus confirming the key role of APN in these established models of angiogenesis. We report toxicity studies in mice showing that these compounds are well tolerated. We report the effects of the compounds, used alone or in combination with rapamycin, on the growth of a select panel of tumours that were subcutaneously xenografted onto Swiss nude mice. Our data indicate that the in vivo efficacy of these new APN inhibitors during the initial phase of tumour growth can be ascribed to their anti-angiogenic activities. However, we also provide evidence that these compounds are effective against established solid tumours. For colonic tumours, the anti-tumour effect depends on the level of APN expression in epithelial cells, and APN expression is associated with down-regulation of the transcription factor HIF-1α. These effects seem to be distinct from those of rapamycin. Our finding that the anti-tumour effect of the inhibitors in the colon requires APN expression strongly suggests that APN plays a crucial function in tumour cells that is distinct from its known role in neovascularisation.
Collapse
Affiliation(s)
- Céline Schmitt
- Université de Haute-Alsace, Ecole Nationale Supérieure de Chimie de Mulhouse, Laboratoire de Chimie Organique et Bioorganique EA4566, 3 rue Alfred Werner, 68093 Mulhouse Cedex, France
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Roles for PI3K/AKT/PTEN Pathway in Cell Signaling of Nonalcoholic Fatty Liver Disease. ISRN ENDOCRINOLOGY 2013; 2013:472432. [PMID: 23431468 PMCID: PMC3570922 DOI: 10.1155/2013/472432] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 01/02/2013] [Indexed: 12/15/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common form of liver pathologies and is associated with obesity and the metabolic syndrome, which represents a range of fatty liver diseases associated with an increased risk of type 2 diabetes. Molecular mechanisms underlying how to make transition from simple fatty liver to nonalcoholic steatohepatitis (NASH) are not well understood. However, accumulating evidence indicates that deregulation of the phosphatidylinositol 3-kinase (PI3K)/AKT pathway in hepatocytes is a common molecular event associated with metabolic dysfunctions including obesity, metabolic syndrome, and the NAFLD. A tumor suppressor PTEN negatively regulates the PI3K/AKT pathways through its lipid phosphatase activity. Molecular studies in the NAFLD support a key role for PTEN in hepatic insulin sensitivity and the development of steatosis, steatohepatitis, and fibrosis. We review recent studies on the features of the PTEN and the PI3K/AKT pathway and discuss the protein functions in the signaling pathways involved in the NAFLD. The molecular mechanisms contributing to the diseases are the subject of considerable investigation, as a better understanding of the pathogenesis will lead to novel therapies for a condition.
Collapse
|
30
|
Millan MJ. An epigenetic framework for neurodevelopmental disorders: from pathogenesis to potential therapy. Neuropharmacology 2012; 68:2-82. [PMID: 23246909 DOI: 10.1016/j.neuropharm.2012.11.015] [Citation(s) in RCA: 151] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2012] [Revised: 11/11/2012] [Accepted: 11/22/2012] [Indexed: 12/12/2022]
Abstract
Neurodevelopmental disorders (NDDs) are characterized by aberrant and delayed early-life development of the brain, leading to deficits in language, cognition, motor behaviour and other functional domains, often accompanied by somatic symptoms. Environmental factors like perinatal infection, malnutrition and trauma can increase the risk of the heterogeneous, multifactorial and polygenic disorders, autism and schizophrenia. Conversely, discrete genetic anomalies are involved in Down, Rett and Fragile X syndromes, tuberous sclerosis and neurofibromatosis, the less familiar Phelan-McDermid, Sotos, Kleefstra, Coffin-Lowry and "ATRX" syndromes, and the disorders of imprinting, Angelman and Prader-Willi syndromes. NDDs have been termed "synaptopathies" in reference to structural and functional disturbance of synaptic plasticity, several involve abnormal Ras-Kinase signalling ("rasopathies"), and many are characterized by disrupted cerebral connectivity and an imbalance between excitatory and inhibitory transmission. However, at a different level of integration, NDDs are accompanied by aberrant "epigenetic" regulation of processes critical for normal and orderly development of the brain. Epigenetics refers to potentially-heritable (by mitosis and/or meiosis) mechanisms controlling gene expression without changes in DNA sequence. In certain NDDs, prototypical epigenetic processes of DNA methylation and covalent histone marking are impacted. Conversely, others involve anomalies in chromatin-modelling, mRNA splicing/editing, mRNA translation, ribosome biogenesis and/or the regulatory actions of small nucleolar RNAs and micro-RNAs. Since epigenetic mechanisms are modifiable, this raises the hope of novel therapy, though questions remain concerning efficacy and safety. The above issues are critically surveyed in this review, which advocates a broad-based epigenetic framework for understanding and ultimately treating a diverse assemblage of NDDs ("epigenopathies") lying at the interface of genetic, developmental and environmental processes. This article is part of the Special Issue entitled 'Neurodevelopmental Disorders'.
Collapse
Affiliation(s)
- Mark J Millan
- Unit for Research and Discovery in Neuroscience, IDR Servier, 125 chemin de ronde, 78290 Croissy sur Seine, Paris, France.
| |
Collapse
|
31
|
Rapamycin inhibits lymphatic endothelial cell tube formation by downregulating vascular endothelial growth factor receptor 3 protein expression. Neoplasia 2012; 14:228-37. [PMID: 22496622 DOI: 10.1593/neo.111570] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Revised: 02/14/2012] [Accepted: 02/15/2012] [Indexed: 02/06/2023]
Abstract
Mammalian target of rapamycin (mTOR) controls lymphangiogenesis. However, the underlying mechanism is not clear. Here we show that rapamycin suppressed insulin-like growth factor 1 (IGF-1)- or fetal bovine serum (FBS)-stimulated lymphatic endothelial cell (LEC) tube formation, an in vitro model of lymphangiogenesis. Expression of a rapamycin-resistant and kinase-active mTOR (S2035T, mTOR-T), but not a rapamycin-resistant and kinase-dead mTOR (S2035T/D2357E, mTOR-TE), conferred resistance to rapamycin inhibition of LEC tube formation, suggesting that rapamycin inhibition of LEC tube formation is mTOR kinase activity dependent. Also, rapamycin inhibited proliferation and motility in the LECs. Furthermore, we found that rapamycin inhibited protein expression of VEGF receptor 3 (VEGFR-3) by inhibiting protein synthesis and promoting protein degradation of VEGFR-3 in the cells. Down-regulation of VEGFR-3 mimicked the effect of rapamycin, inhibiting IGF-1- or FBS-stimulated tube formation, whereas over-expression of VEGFR-3 conferred high resistance to rapamycin inhibition of LEC tube formation. The results indicate that rapamycin inhibits LEC tube formation at least in part by downregulating VEGFR-3 protein expression.
Collapse
|
32
|
Grzmil M, Hemmings BA. Translation Regulation as a Therapeutic Target in Cancer: Figure 1. Cancer Res 2012; 72:3891-900. [DOI: 10.1158/0008-5472.can-12-0026] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
33
|
Abstract
Nutrient sensing and the capacity to respond to starvation is tightly regulated as a means of cell survival. Among the features of the starvation response are induction of both translational repression and autophagy. Despite the fact that intracellular parasite like Toxoplasma gondii within a host cell predicted to be nutrient rich, they encode genes involved in both translational repression and autophagy. We therefore examined the consequence of starvation, a classic trigger of autophagy, on intracellular parasites. As expected, starvation results in the activation of the translational repression system as evidenced by elevation of phosphorylated TgIF2α (TgIF2α-P). Surprisingly, we also observe a rapid and selective fragmentation of the single parasite mitochondrion that leads irreversibly to parasite death. This profound effect was dependent primarily on the limitation of amino acids and involved signalling by the parasite TOR homologue. Notably, the effective blockade of mitochondrial fragmentation by the autophagy inhibitor 3-methyl adenine (3-MA) suggests an autophagic mechanism. In the absence of a documented apoptotic cascade in T. gondii, the data suggest that autophagy is the primary mechanism of programmed cell death in T. gondii and potentially other related parasites.
Collapse
Affiliation(s)
- Debasish Ghosh
- Department of Microbiology, Immunology and Molecular Genetics; University of Kentucky College of Medicine, Lexington KY 40536, USA
| | - Julia L. Walton
- Department of Microbiology, Immunology and Molecular Genetics; University of Kentucky College of Medicine, Lexington KY 40536, USA
| | - Paul D. Roepe
- Departments of Chemistry, Biochemistry and Cellular and Molecular Biology, Georgetown University, Washington DC. 20057, USA
| | - Anthony P. Sinai
- Department of Microbiology, Immunology and Molecular Genetics; University of Kentucky College of Medicine, Lexington KY 40536, USA
| |
Collapse
|
34
|
Abstract
The TOR (target of rapamycin) proteins are found in all eukaryotes. TOR has a protein kinase domain, as well as other domains through which it interacts with partner proteins to form at least two types of multiprotein complex, TORC1 and TORC2 (TOR complexes 1 and 2). Rapamycin, an antibiotic and immunosuppressant, inhibits functions of TORC1. Use of this drug has revealed roles for TORC1 and its mammalian counterpart, mTORC1, in promoting many anabolic processes. mTORC1 signalling is activated by growth factors and nutrients. It is highly active in many cancers and plays a role in tumorigenesis and in other diseases. Much less is known so far about the functions and regulation of (m)TORC2. The goal of this meeting was to bring together researchers studying the roles of mTORC1/2 in normal cell and animal physiology in diverse systems, as well as scientists exploring the therapeutic value of inhibiting mTOR (mammalian TOR) signalling.
Collapse
|