1
|
Gorjão N, Borowski LS, Szczesny RJ, Graczyk D. POLR1D, a shared subunit of RNA polymerase I and III, modulates mTORC1 activity. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119957. [PMID: 40222657 DOI: 10.1016/j.bbamcr.2025.119957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 03/21/2025] [Accepted: 04/08/2025] [Indexed: 04/15/2025]
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) is a crucial nutrient sensor and a major regulator of cell growth and proliferation. While mTORC1 activity is frequently upregulated in cancer, the mechanisms regulating mTORC1 are not fully understood. POLR1D, a shared subunit of RNA polymerases I and III, is often upregulated in colorectal cancer (CRC) and mutated in Treacher-Collins syndrome. POLR1D, together with its binding partner POLR1C, forms a dimer that is believed to initiate the assembly of the multisubunit RNA polymerases I and III. Our data reveal an unexpected link between POLR1D and mTORC1 signalling. We found that the overproduction of POLR1D in human cells stimulates mTORC1 activity. In contrast, the downregulation of POLR1D leads to the repression of the mTORC1 pathway. Additionally, we demonstrate that a pool of POLR1D localises to the cytoplasm and interacts with the mTORC1 regulator RAGA and RAPTOR. Furthermore, POLR1D enhances the interaction between RAPTOR and RAGA and sustains mTORC1 activity under starvation conditions. We have identified a novel role for the RNA polymerase I/III subunit POLR1D in regulating mTORC1 signalling. Our findings suggest the existence of a new node in the already complex mTORC1 signalling network, where POLR1D functions to convey the cell's internal status, namely polymerase assembly, to this kinase.
Collapse
Affiliation(s)
- Neuton Gorjão
- Institute of Biochemistry and Biophysics Polish Academy of Sciences, ul. Pawińskiego 5a, 02-106 Warsaw, Poland
| | - Lukasz S Borowski
- Institute of Biochemistry and Biophysics Polish Academy of Sciences, ul. Pawińskiego 5a, 02-106 Warsaw, Poland; University of Warsaw, Faculty of Biology, Institute of Genetics and Biotechnology, ul. Pawińskiego 5a, 02-106 Warsaw, Poland
| | - Roman J Szczesny
- Institute of Biochemistry and Biophysics Polish Academy of Sciences, ul. Pawińskiego 5a, 02-106 Warsaw, Poland
| | - Damian Graczyk
- Institute of Biochemistry and Biophysics Polish Academy of Sciences, ul. Pawińskiego 5a, 02-106 Warsaw, Poland.
| |
Collapse
|
2
|
Mantoan Ritter L, Annear NMP, Baple EL, Ben-Chaabane LY, Bodi I, Brosson L, Cadwgan JE, Coslett B, Crosby AH, Davies DM, Daykin N, Dedeurwaerdere S, Dühring Fenger C, Dunlop EA, Elmslie FV, Girodengo M, Hambleton S, Jansen AC, Johnson SR, Kearley KC, Kingswood JC, Laaniste L, Lachlan K, Latchford A, Madsen RR, Mansour S, Mihaylov SR, Muhammed L, Oliver C, Pepper T, Rawlins LE, Schim van der Loeff I, Siddiqui A, Takhar P, Tatton-Brown K, Tee AR, Tibarewal P, Tye C, Ultanir SK, Vanhaesebroeck B, Zare B, Pal DK, Bateman JM. mTOR pathway diseases: challenges and opportunities from bench to bedside and the mTOR node. Orphanet J Rare Dis 2025; 20:256. [PMID: 40426219 DOI: 10.1186/s13023-025-03740-1] [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: 11/06/2024] [Accepted: 04/16/2025] [Indexed: 05/29/2025] Open
Abstract
Mechanistic target of rapamycin (mTOR) is a highly conserved serine/threonine kinase that regulates key cellular processes including cell growth, autophagy and metabolism. Hyperactivation of the mTOR pathway causes a group of rare and ultrarare genetic diseases. mTOR pathway diseases have diverse clinical manifestations that are managed by distinct medical disciplines but share a common underlying molecular basis. There is a now a deep understanding of the molecular underpinning that regulates the mTOR pathway but effective treatments for most mTOR pathway diseases are lacking. Translating scientific knowledge into clinical applications to benefit the unmet clinical needs of patients is a major challenge common to many rare diseases. In this article we expound how mTOR pathway diseases provide an opportunity to coordinate basic and translational disease research across the group, together with industry, medical research foundations, charities and patient groups, by pooling expertise and driving progress to benefit patients. We outline the germline and somatic mutations in the mTOR pathway that cause rare diseases and summarise the prevalence, genetic basis, clinical manifestations, pathophysiology and current treatments for each disease in this group. We describe the challenges and opportunities for progress in elucidating the underlying mechanisms, improving diagnosis and prognosis, as well as the development and approval of new therapies for mTOR pathway diseases. We illustrate the crucial role of patient public involvement and engagement in rare disease and mTOR pathway disease research. Finally, we explain how the mTOR Pathway Diseases node, part of the Research Disease Research UK Platform, will address these challenges to improve the understanding, diagnosis and treatment of mTOR pathway diseases.
Collapse
Affiliation(s)
- Laura Mantoan Ritter
- King's College London Institute of Psychiatry Psychology and Neuroscience, London, UK
- King's College Hospital NHS Foundation Trust, London, UK
| | - Nicholas M P Annear
- St George's University Hospitals NHS Foundation Trust, London, UK
- School of Health & Medical Sciences, City St George's, University of London, London, UK
| | | | - Leila Y Ben-Chaabane
- King's College London Institute of Psychiatry Psychology and Neuroscience, London, UK
| | - Istvan Bodi
- King's College Hospital NHS Foundation Trust, London, UK
| | | | | | | | | | | | | | | | | | | | - Frances V Elmslie
- St George's University Hospitals NHS Foundation Trust, London, UK
- School of Health & Medical Sciences, City St George's, University of London, London, UK
| | - Marie Girodengo
- King's College London Institute of Psychiatry Psychology and Neuroscience, London, UK
- The Francis Crick Institute, London, UK
| | - Sophie Hambleton
- Newcastle University Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | | | - Simon R Johnson
- Centre for Respiratory Research, NIHR Nottingham Biomedical Research Centre and Biodiscovery Institute, Translational Medical Sciences, University of Nottingham, Nottingham, UK
| | - Kelly C Kearley
- mTOR Node Advisory Panel (MAP), London, UK
- PTEN UK and Ireland Patient Group, London, UK
| | - John C Kingswood
- St George's University Hospitals NHS Foundation Trust, London, UK
| | | | - Katherine Lachlan
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Andrew Latchford
- Polyposis Registry, St Mark's Hospital, London, UK
- Department of Surgery and Cancer, Imperial College London, London, UK
| | | | - Sahar Mansour
- St George's University Hospitals NHS Foundation Trust, London, UK
- School of Health & Medical Sciences, City St George's, University of London, London, UK
| | | | | | | | - Tom Pepper
- PTEN Research, Cheltenham, Gloucestershire, UK
| | | | - Ina Schim van der Loeff
- Newcastle University Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Ata Siddiqui
- King's College Hospital NHS Foundation Trust, London, UK
| | | | - Katrina Tatton-Brown
- St George's University Hospitals NHS Foundation Trust, London, UK
- School of Health & Medical Sciences, City St George's, University of London, London, UK
| | | | | | - Charlotte Tye
- King's College London Institute of Psychiatry Psychology and Neuroscience, London, UK
| | | | | | | | - Deb K Pal
- King's College London Institute of Psychiatry Psychology and Neuroscience, London, UK
| | - Joseph M Bateman
- King's College London Institute of Psychiatry Psychology and Neuroscience, London, UK.
| |
Collapse
|
3
|
Duan S, Agger K, Messling JE, Nishimura K, Han X, Peña-Rømer I, Shliaha P, Damhofer H, Douglas M, Kohli M, Pal A, Asad Y, Van Dyke A, Reilly R, Köchl R, Tybulewicz VLJ, Hendrickson RC, Raynaud FI, Gallipoli P, Poulogiannis G, Helin K. WNK1 signalling regulates amino acid transport and mTORC1 activity to sustain acute myeloid leukaemia growth. Nat Commun 2025; 16:4920. [PMID: 40425534 DOI: 10.1038/s41467-025-59969-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
The lack of curative therapies for acute myeloid leukaemia (AML) remains an ongoing challenge despite recent advances in the understanding of the molecular basis of the disease. Here we identify the WNK1-OXSR1/STK39 pathway as a previously uncharacterised dependency in AML. We show that genetic depletion and pharmacological inhibition of WNK1 or its downstream phosphorylation targets OXSR1 and STK39 strongly reduce cell proliferation and induce apoptosis in leukaemia cells in vitro and in vivo. Furthermore, we show that the WNK1-OXSR1/STK39 pathway controls mTORC1 signalling via regulating amino acid uptake through a mechanism involving the phosphorylation of amino acid transporters, such as SLC38A2. Our findings underscore an important role of the WNK1-OXSR1/STK39 pathway in regulating amino acid uptake and driving AML progression.
Collapse
Affiliation(s)
- Shunlei Duan
- Division of Cell and Molecular Biology, The Institute of Cancer Research, Londo, UK
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Karl Agger
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Jan-Erik Messling
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Koutarou Nishimura
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
- Cell Biology Program and Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Xuerui Han
- Division of Cell and Molecular Biology, The Institute of Cancer Research, Londo, UK
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Isabel Peña-Rømer
- Division of Cell and Molecular Biology, The Institute of Cancer Research, Londo, UK
| | - Pavel Shliaha
- Microchemistry and Proteomics Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Helene Damhofer
- Division of Cell and Molecular Biology, The Institute of Cancer Research, Londo, UK
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
- Cell Biology Program and Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Max Douglas
- Division of Cell and Molecular Biology, The Institute of Cancer Research, Londo, UK
| | - Manas Kohli
- Division of Cell and Molecular Biology, The Institute of Cancer Research, Londo, UK
| | - Akos Pal
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK
| | - Yasmin Asad
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK
| | - Aaron Van Dyke
- Department of Chemistry & Biochemistry, Fairfield University, Fairfield, CT, USA
| | - Raquel Reilly
- Department of Chemistry & Biochemistry, Fairfield University, Fairfield, CT, USA
| | | | | | - Ronald C Hendrickson
- Microchemistry and Proteomics Core Facility, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Florence I Raynaud
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, UK
| | - Paolo Gallipoli
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - George Poulogiannis
- Division of Cell and Molecular Biology, The Institute of Cancer Research, Londo, UK
| | - Kristian Helin
- Division of Cell and Molecular Biology, The Institute of Cancer Research, Londo, UK.
- Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark.
- Cell Biology Program and Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
4
|
Jiang C, Tan X, Jin J, Wang P. The Molecular Basis of Amino Acids Sensing. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2501889. [PMID: 40411419 DOI: 10.1002/advs.202501889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/29/2025] [Indexed: 05/26/2025]
Abstract
Amino acids are organic compounds that serve as the building blocks of proteins and peptides. Additionally, they function as bioactive molecules that play important roles in metabolic regulation and signal transduction. The ability of cells to sense fluctuations in intracellular and extracellular amino acid levels is vital for effectively regulating protein synthesis and catabolism, maintaining homeostasis, adapting to diverse nutritional environments and influencing cell fate decision. In this review, the recent molecular insights into amino acids sensing are discussed, along with the different sensing mechanisms in distinct organisms.
Collapse
Affiliation(s)
- Cong Jiang
- Shanghai Tenth People's Hospital, School of Medicine, Tongji University Cancer Center, Tongji University, Shanghai, 200092, China
| | - Xiao Tan
- Shanghai Tenth People's Hospital, School of Medicine, Tongji University Cancer Center, Tongji University, Shanghai, 200092, China
| | - Jiali Jin
- Shanghai Tenth People's Hospital, School of Medicine, Tongji University Cancer Center, Tongji University, Shanghai, 200092, China
| | - Ping Wang
- Shanghai Tenth People's Hospital, School of Medicine, Tongji University Cancer Center, Tongji University, Shanghai, 200092, China
| |
Collapse
|
5
|
Nicastro R, Péli-Gulli MP, Caligaris M, Jaquenoud M, Dokládal L, Alba J, Tripodi F, Pillet B, Brunner M, Stumpe M, Muneshige K, Hatakeyama R, Dengjel J, De Virgilio C. TORC1 autonomously controls its spatial partitioning via the Rag GTPase tether Tco89. Cell Rep 2025; 44:115683. [PMID: 40359108 DOI: 10.1016/j.celrep.2025.115683] [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: 10/01/2024] [Revised: 02/17/2025] [Accepted: 04/17/2025] [Indexed: 05/15/2025] Open
Abstract
The eukaryotic target of rapamycin complex 1 (TORC1) kinase is a homeostatic regulator of growth, integrating nutritional cues at the endolysosomal compartment. Amino acids activate mammalian TORC1 (mTORC1) through the Rag GTPases that recruit it to lysosomes via a short domain within the mTORC1 subunit Raptor. Intriguingly, this "Raptor claw" domain is absent in Kog1, the Raptor ortholog in yeast. Instead, as we show here, yeast utilizes the fungal-specific Tco89 to tether TORC1 to active Rag GTPases. This interaction enables TORC1 to precisely calibrate the activity of the S6K1-related effector kinase Sch9 in response to amino acid availability. TORC1 stabilizes Tco89 by phosphorylation, and its inactivation causes swift Tco89 proteolysis, provoking a redistribution of TORC1 from the vacuole to signaling endosomes and its spatial separation from Sch9. Thus, TORC1 not only operates in spatially distinct subcellular pools but also controls its own quantitative distribution between these pools to economize energy resources under fluctuating nutrient conditions.
Collapse
Affiliation(s)
- Raffaele Nicastro
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | | | - Marco Caligaris
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Malika Jaquenoud
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Ladislav Dokládal
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Josephine Alba
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Farida Tripodi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milano, Italy
| | - Benjamin Pillet
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Melanie Brunner
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Michael Stumpe
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Kenji Muneshige
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Aberdeen, UK
| | - Riko Hatakeyama
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Aberdeen, UK
| | - Jörn Dengjel
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | | |
Collapse
|
6
|
Haidurov A, Budanov AV. Sestrins in Carcinogenesis-The Firefighters That Sometimes Stoke the Fire. Cancers (Basel) 2025; 17:1578. [PMID: 40361504 PMCID: PMC12071529 DOI: 10.3390/cancers17091578] [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: 04/02/2025] [Revised: 04/29/2025] [Accepted: 05/05/2025] [Indexed: 05/15/2025] Open
Abstract
Sestrins (SESN1-3) are a family of stress-responsive proteins that regulate cellular metabolism and redox balance, both of which are frequently disrupted in cancer. As direct targets of stress-responsive transcription factors, including tumour suppressor p53, Sestrins function as leucine-dependent inhibitors of mTORC1 and potent antioxidants. Their downregulation is widely observed across multiple cancers and is associated with increased tumour growth and poor prognosis. Despite their consistent tumour-suppressive effects through mTORC1 inhibition and promotion of p53-dependent apoptosis, Sestrins exhibit a limited role in tumour initiation, which appears to be context-dependent. Their antioxidant activity reduces oxidative damage, thereby protecting against genomic instability and other cancer-promoting events. However, in certain contexts, Sestrins may promote tumour survival and progression by stimulating pro-survival pathways, such as AKT signalling through mTORC2 activation. This review examines the molecular mechanisms underlying these dual functions, with a particular focus on mTOR signalling and oxidative stress. We also discuss Sestrin expression patterns and functional outcomes in various cancer types, including lung, liver, colon, skin, prostate, and follicular lymphomas, highlighting their potential as diagnostic markers and therapeutic targets.
Collapse
Affiliation(s)
- Alexander Haidurov
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, D02 R590 Dublin, Ireland
| | - Andrei V. Budanov
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Pearse Street, D02 R590 Dublin, Ireland
| |
Collapse
|
7
|
Li Z, Guan Y, Gao J, Zhu L, Zeng Z, Jing Q, Wan Q, Fan Q, Ren X, Pei H, Zhang D, Rong Y, Rong Z, He J, Zhang Y, Li N, Chen P, Sun L, Xu B, Nie Y, Deng Y. PPDPF-mediated regulation of BCAA metabolism enhances mTORC1 activity and drives cholangiocarcinoma progression. Oncogene 2025; 44:1415-1433. [PMID: 40025229 DOI: 10.1038/s41388-025-03320-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 01/30/2025] [Accepted: 02/19/2025] [Indexed: 03/04/2025]
Abstract
Tumor cells display profound changes in the metabolism of branched-chain amino acids (BCAA). However, how these changes are regulated to facilitate tumorigenesis is not yet completely understood. Here, we identified pancreatic progenitor cell differentiation and proliferation factor (PPDPF) as a BCAA-responsive protein through extensive screening using stable isotope labeling with amino acids in cell culture (SILAC). PPDPF is upregulated in cholangiocarcinoma to enhance the malignant phenotype of cholangiocarcinoma cells by activating the mTORC1 signaling pathway. Metabolic flux analysis and mechanistic studies revealed that PPDPF prevented the interaction between MCCA and MCCB, thus inhibiting leucine catabolism and activating mTORC1 signaling. Moreover, upon amino acid starvation, ariadne RBR E3 ubiquitin protein ligase 2 (ARIH2) and OTU deubiquitinase 4 (OTUD4) cooperatively regulated the stability of the PPDPF protein by modulating its ubiquitination. Additionally, monocytes/macrophage-derived IL-10 increased the BCAA content in cholangiocarcinoma cells and stabilized the PPDPF protein, even under amino acid starvation conditions. Knockout of PPDPF or restriction of leucine intake significantly inhibits the progression of cholangiocarcinoma in a mouse model. Collectively, we discovered a novel role for PPDPF in promoting the progression of cholangiocarcinoma by activating mTORC1 signaling through the inhibition of leucine catabolism. The present study suggests that targeting PPDPF or decreasing dietary leucine intake may provide a new strategy to improve the treatment efficacy of cholangiocarcinoma.
Collapse
Affiliation(s)
- Zhi Li
- Key Laboratory of Molecular Radiation Oncology (Xiangya Hospital, Central South University), Changsha, China
- Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha, China
- Institute of Cancer Research, National Clinical Research Center for Geriatric Disorders (Xiangya), Xiangya Hospital, Central South University, Changsha, China
| | - Yidi Guan
- Key Laboratory of Molecular Radiation Oncology (Xiangya Hospital, Central South University), Changsha, China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jie Gao
- Key Laboratory of Molecular Radiation Oncology (Xiangya Hospital, Central South University), Changsha, China
- Shanghai Key Laboratory of Thoracic Tumor Biotherapy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lan Zhu
- NHC Key Laboratory of Pulmonary Immune-related Diseases, Guizhou Provincial People's Hospital, Guiyang, China
| | - Zimei Zeng
- Shanghai Key Laboratory of Thoracic Tumor Biotherapy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qianyu Jing
- NHC Key Laboratory of Pulmonary Immune-related Diseases, Guizhou Provincial People's Hospital, Guiyang, China
| | - Quan Wan
- NHC Key Laboratory of Pulmonary Immune-related Diseases, Guizhou Provincial People's Hospital, Guiyang, China
| | - Qi Fan
- Shanghai Key Laboratory of Thoracic Tumor Biotherapy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinxin Ren
- Cancer Center, Department of Pathology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Haiping Pei
- Key Laboratory of Molecular Radiation Oncology (Xiangya Hospital, Central South University), Changsha, China
| | - Dexiang Zhang
- Department of General Surgery, Zhongshan Xuhui Hospital Affiliated to Fudan University, Shanghai, China
| | - Yefei Rong
- The Department of Emergency Surgery, the Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhuoxian Rong
- Key Laboratory of Molecular Radiation Oncology (Xiangya Hospital, Central South University), Changsha, China
| | - Junju He
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yuefang Zhang
- Songjiang Research Institute, Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Nan Li
- Department of Hepatic Surgery I (Ward I), Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Pan Chen
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Lunquan Sun
- Key Laboratory of Molecular Radiation Oncology (Xiangya Hospital, Central South University), Changsha, China.
- Hunan International Science and Technology Collaboration Base of Precision Medicine for Cancer, Changsha, China.
- Institute of Cancer Research, National Clinical Research Center for Geriatric Disorders (Xiangya), Xiangya Hospital, Central South University, Changsha, China.
| | - Bin Xu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China.
| | - Yingjie Nie
- Department of Research, the University of HongKong-Shenzhen Hospital, Shenzhen, China.
| | - Yuezhen Deng
- Shanghai Key Laboratory of Thoracic Tumor Biotherapy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Yangjiang Key Laboratory of Respiratory Disease, People's Hospital of Yangjiang, 529500, Yangjiang, Guangdong, China.
| |
Collapse
|
8
|
Zhao J, Shen Q, Yong X, Li X, Tian X, Sun S, Xu Z, Zhang X, Zhang L, Yang H, Shao Z, Xu H, Jiang Y, Zhang Y, Yan W. Cryo-EM reveals cholesterol binding in the lysosomal GPCR-like protein LYCHOS. Nat Struct Mol Biol 2025; 32:896-904. [PMID: 39824976 DOI: 10.1038/s41594-024-01470-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 12/06/2024] [Indexed: 01/20/2025]
Abstract
Cholesterol plays a pivotal role in modulating the activity of mechanistic target of rapamycin complex 1 (mTOR1), thereby regulating cell growth and metabolic homeostasis. LYCHOS, a lysosome-localized G-protein-coupled receptor-like protein, emerges as a cholesterol sensor and is capable of transducing the cholesterol signal to affect the mTORC1 function. However, the precise mechanism by which LYCHOS recognizes cholesterol remains unknown. Here, using cryo-electron microscopy, we determined the three-dimensional structural architecture of LYCHOS in complex with cholesterol molecules, revealing a unique arrangement of two sequential structural domains. Through a comprehensive analysis of this structure, we elucidated the specific structural features of these two domains and their collaborative role in the process of cholesterol recognition by LYCHOS.
Collapse
Affiliation(s)
- Jie Zhao
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China
| | - Qingya Shen
- Department of Pathology of Sir Run Shaw Hospital, Department of Pharmacology, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, China
| | - Xihao Yong
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China
| | - Xin Li
- NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, Metabolomics and Proteomics Technology Platform, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaowen Tian
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China
| | - Suyue Sun
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China
| | - Zheng Xu
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China
| | - Xiaoyu Zhang
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China
| | - Lu Zhang
- NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, Metabolomics and Proteomics Technology Platform, West China Hospital, Sichuan University, Chengdu, China
| | - Hao Yang
- NHC Key Lab of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, Metabolomics and Proteomics Technology Platform, West China Hospital, Sichuan University, Chengdu, China
| | - Zhenhua Shao
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China.
| | - Haoxing Xu
- New Cornerstone Science Laboratory & Liangzhu Laboratory, the Second Affiliated Hospital & School of Basic Medical Sciences, Zhejiang University, Hangzhou, China.
| | - Yiyang Jiang
- Department of Hepatobiliary Surgery, Innovative Institute of Tumor Immunity and Medicine (ITIM), Anhui Province Key Laboratory of Tumor Immune Microenvironment and Immunotherapy, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| | - Yan Zhang
- Department of Pathology of Sir Run Shaw Hospital, Department of Pharmacology, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China.
- MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, China.
| | - Wei Yan
- Division of Nephrology and Kidney Research Institute, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
9
|
Hassan FA, Slone C, McDonald RJ, Dueber JC, Ashraf AM, Windon MJ, Fackelmayer OJ, Lee CY, Bocklage TJ, Allison DB. Folliculin ( FLCN) in Thyroid Tumors: Incidence, Significance, and Role as a Driver Gene and Secondary Alteration. Curr Oncol 2025; 32:224. [PMID: 40277780 PMCID: PMC12026003 DOI: 10.3390/curroncol32040224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/01/2025] [Accepted: 04/05/2025] [Indexed: 04/26/2025] Open
Abstract
Thyroid carcinomas are driven by diverse molecular alterations, but the tumor suppressor gene folliculin (FLCN), best known for its role in Birt-Hogg-Dubé (BHD) syndrome, has received limited attention in thyroid tumors. Here, we describe two thyroid tumors with pathogenic FLCN alterations-one germline and one somatic-and analyze the broader prevalence and significance of FLCN in thyroid carcinomas using multiple large sequencing datasets, including ORIEN-AVATAR. Patient 1, with a germline FLCN mutation and a history of BHD syndrome, presented with a well-circumscribed oncocytic adenoma. Molecular testing confirmed biallelic FLCN inactivation, but no additional mutations or aggressive features were observed, and the patient remained disease-free post-thyroidectomy. Patient 2 harbored a somatic FLCN mutation in an oncocytic poorly differentiated thyroid carcinoma, which exhibited extensive angioinvasion, high proliferative activity, and concurrent TP53 and RB1 mutations. The tumor progressed with metastatic disease despite multimodal treatment. Thyroid carcinomas revealed FLCN alterations in 1.1% of cases. Pathogenic mutations were rare but associated with oncocytic morphology, while homozygous deletions occurred more frequently in genomically unstable tumors, including anaplastic thyroid carcinoma. These findings suggest FLCN mutations may act as early oncogenic drivers in oncocytic thyroid neoplasms, while deletions represent secondary events in aggressive tumor evolution. The lack of FLCN coverage in standard thyroid molecular panels likely underestimates its clinical relevance. Including FLCN in genetic testing could improve tumor detection and characterization, particularly in BHD patients who may benefit from routine thyroid screening. Further studies are needed to clarify FLCN's role in thyroid cancer pathogenesis.
Collapse
Affiliation(s)
- Faisal A. Hassan
- Department of Pathology & Laboratory Medicine, University of Kentucky College of Medicine, Lexington, KY 40506, USA
| | - Camryn Slone
- Kentucky College of Medicine, University of Kentucky, Lexington, KY 40506, USA
| | - Robert J. McDonald
- Department of Pathology & Laboratory Medicine, University of Kentucky College of Medicine, Lexington, KY 40506, USA
| | - Julie C. Dueber
- Department of Pathology & Laboratory Medicine, University of Kentucky College of Medicine, Lexington, KY 40506, USA
| | - Adeel M. Ashraf
- Department of Pathology & Laboratory Medicine, University of Kentucky College of Medicine, Lexington, KY 40506, USA
| | - Melina J. Windon
- Department of Otolaryngology—Head and Neck Surgery, University of Kentucky College of Medicine, Lexington, KY 40506, USA
| | - Oliver J. Fackelmayer
- Department of Surgery, Division of Endocrine Surgery, University of Kentucky College of Medicine, Lexington, KY 40506, USA
| | - Cortney Y. Lee
- Department of Surgery, Division of Endocrine Surgery, University of Kentucky College of Medicine, Lexington, KY 40506, USA
| | - Therese J. Bocklage
- Department of Pathology & Laboratory Medicine, University of Kentucky College of Medicine, Lexington, KY 40506, USA
- Markey Cancer Center, Lexington, KY 40536, USA
| | - Derek B. Allison
- Department of Pathology & Laboratory Medicine, University of Kentucky College of Medicine, Lexington, KY 40506, USA
- Markey Cancer Center, Lexington, KY 40536, USA
| |
Collapse
|
10
|
Solanki S, Taranto J, Rebernick R, Castillo C, Ponnusamy V, Sykes MM, Leiser SF, Lee JH, Schmidt T, Shah YM. Low Protein Diet Exacerbates Experimental Mouse Models of Colitis through Epithelial Autonomous and Nonautonomous Mechanisms. J Nutr 2025:S0022-3166(25)00186-5. [PMID: 40216295 DOI: 10.1016/j.tjnut.2025.03.031] [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: 10/07/2024] [Revised: 01/30/2025] [Accepted: 03/04/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Patients with inflammatory bowel diseases (IBDs) are at risk of protein malnutrition due to increased protein loss or reduced dietary intake. The consequences of protein malnutrition on intestinal epithelial metabolism and disease progression remain poorly understood. OBJECTIVES Given the critical role of the mechanistic target of rapamycin complex 1 (mTORC1) as an amino acid sensor and a key regulator of intestinal epithelial metabolism and homeostasis, along with the well-established influence of diet on the gut microbiota and IBD, we focused on accessing the role of dietary protein in modulating intestinal epithelial mTORC1, determine the contributions of specific amino acids such as leucine and arginine, and examine the interplay between protein malnutrition and gut microbiota driving IBD. METHODS C57BL/6 mice were assigned to a control (20% protein, n = 6), a low protein (4% protein, n = 7), or diets selectively deficient in leucine, arginine, and other essential amino acids (n = 5-6). Colitis was induced by administering 2.5% dextran sulfate sodium in drinking water for 6 d. Intestinal epithelial mTORC1 activity was assessed by immunoblotting. Gut microbiota composition was characterized using 16S sequencing, and the microbiota's role in colitis was evaluated through broad-spectrum antibiotic treatment. Disease severity was quantified by monitoring weight loss, colon shortening, histopathological damage, and inflammatory cytokine expression. RESULTS Protein restriction increased the severity of dextran sulfate sodium-induced colitis compared to the control diet (∗∗∗P < 0.001). Mice fed arginine-restricted diets exhibited increased colitis (∗P < 0.05). Protein restriction induced significant alterations in gut microbiota composition, and antibiotic-mediated microbiota depletion partially ameliorated colitis severity, revealing a microbiota-dependent mechanism underlying disease exacerbation. CONCLUSIONS Our study demonstrates a complex interplay between dietary protein, epithelial mTORC1 signaling, and gut microbiota in modulating IBD pathogenesis and highlights the potential for targeted dietary strategies, including amino acid supplementation, to improve disease management in patients with IBD.
Collapse
Affiliation(s)
- Sumeet Solanki
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Joseph Taranto
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Ryan Rebernick
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Cristina Castillo
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Varun Ponnusamy
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Madeline M Sykes
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Scott F Leiser
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Jun Hee Lee
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Thomas Schmidt
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, United States
| | - Yatrik M Shah
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, Michigan, United States; Division of Gastroenterology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, United States; Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, Michigan, United States.
| |
Collapse
|
11
|
Ahn E, Kim SI, Park S, Kim S, Kim H, Lee H, Kim H, Song EJ, Ahn T, Song YS. Innovative qPCR Algorithm Using Platelet-Derived RNA for High-Specificity and Cost-Effective Ovarian Cancer Detection. Cancers (Basel) 2025; 17:1251. [PMID: 40227838 PMCID: PMC11988175 DOI: 10.3390/cancers17071251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 03/31/2025] [Accepted: 03/31/2025] [Indexed: 04/15/2025] Open
Abstract
Background/Objectives: Ovarian cancer (OC) remains one of the most lethal gynecologic malignancies, largely due to the challenges of early detection. While next-generation sequencing (NGS) has been explored for screening, its high cost limits large-scale implementation. To develop a more accessible diagnostic solution, we designed a qPCR-based algorithm optimized for early OC detection, with a focus on high-grade serous ovarian cancer (HGSOC), the most aggressive subtype. Methods: Peripheral blood samples from 19 ovarian cancer patients, 37 benign tumor patients, and 34 asymptomatic controls were analyzed using RNA sequencing to identify splice junction-based biomarkers with minimal expression in benign samples but elevated in OC. Results: A final panel of 10 markers was validated via qPCR, demonstrating strong agreement with sequencing data (R2 = 0.44-0.98). The classification algorithm achieved 94.1% sensitivity and 94.4% specificity (AUC = 0.933). Conclusions: By leveraging platelet RNA profiling, this approach offers high specificity, accessibility, and potential for early OC detection. Future studies will focus on expanding histologic diversity and refining biomarker panels to further enhance diagnostic performance.
Collapse
Affiliation(s)
- Eunyong Ahn
- Foretell My Health, Inc., 558 Handong-ro Buk-gu, Pohang 37554, Republic of Korea
| | - Se Ik Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Sungmin Park
- Foretell My Health, Inc., 558 Handong-ro Buk-gu, Pohang 37554, Republic of Korea
| | - Sarah Kim
- Foretell My Health, Inc., 558 Handong-ro Buk-gu, Pohang 37554, Republic of Korea
| | - Hyunjung Kim
- Foretell My Health, Inc., 558 Handong-ro Buk-gu, Pohang 37554, Republic of Korea
| | - Hyejin Lee
- Foretell My Health, Inc., 558 Handong-ro Buk-gu, Pohang 37554, Republic of Korea
| | - Heeyeon Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Eun Ji Song
- Foretell My Health, Inc., 558 Handong-ro Buk-gu, Pohang 37554, Republic of Korea
| | - TaeJin Ahn
- Foretell My Health, Inc., 558 Handong-ro Buk-gu, Pohang 37554, Republic of Korea
- Department of Advanced Convergence, Handong Global University, Pohang 37554, Republic of Korea
| | - Yong-Sang Song
- Department of Obstetrics and Gynecology, Myongji Hospital, Goyang 10475, Republic of Korea
| |
Collapse
|
12
|
Sutherland AE. The role of serendipity in our investigation of embryo implantation. Dev Biol 2025; 520:135-140. [PMID: 39826766 PMCID: PMC11830518 DOI: 10.1016/j.ydbio.2025.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 01/07/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025]
Abstract
Serendipity plays a huge role in science, and having a prepared mind that can seize upon a chance observation or occurrence can drive a project forward. This happened in my lab with a project centered on the regulation of trophoblast cell behavior at implantation. We discovered that amino acids regulate the onset of trophoblast motility through the activation of the kinase complex mTORC1, and that this acts as a checkpoint to trophoblast differentiation. This finding not only broadened our understanding of the mechanisms underlying embryo implantation, but also provided new ways of thinking about the regulation of diapause, a state of suspended embryonic development that occurs in many species. I should say that we re-discovered the fact that amino acids regulate the onset of trophoblast motility, as reading the literature showed us that others had made this same observation some 30 years previously and we were fortuitously able to build upon those findings. This project confirmed to me how valuable it is to read the literature widely, both historical papers and those in fields outside one's area of research, and to go to seminars on topics outside one's area.
Collapse
Affiliation(s)
- Ann E Sutherland
- Department of Cell Biology, University of Virginia Health System, PO Box 800732, Charlottesville, VA, 22908-0732, USA.
| |
Collapse
|
13
|
Liu J, Huang R, Tang F, Ma Y, Kwan P. A missense variant in DEPDC5 resulted in abnormal morphology and increased seizure susceptibility and mortality through regulating mTOR signaling. Neurobiol Dis 2025; 207:106842. [PMID: 39954744 DOI: 10.1016/j.nbd.2025.106842] [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: 06/28/2024] [Revised: 02/12/2025] [Accepted: 02/12/2025] [Indexed: 02/17/2025] Open
Abstract
Dishevelled, Egl-10 and Pleckstrin domain-containing 5 (DEPDC5), a key inhibitor of the mammalian/mechanistic target of rapamycin (mTOR) pathway, is frequently associated with epilepsy. However, the functional consequences of most DEPDC5 variants rely on in silico predictions and have not been experimentally confirmed.This study aimed to determine the functional consequences of a DEPDC5 variant identified in patients with epilepsy across multiple generations in a Chinese family. We identified a missense heterozygous variant (c. 2055C > A; p. Phe685Leu) in DEPDC5 in Chinese family affected by epilepsy across three generations. This variant has not been previously reported in the Chinese population. Primary neuron cultures transfected with the mutant plasmid exhibited altered subcellular localization. To explore the mechanisms of epilepsy linked to this variant, we created nervous system-specific conditional human DEPDC5 knock-in mouse using Cre-recombination under the Nestin promotor (hDEPDC5WT mice, hDEPDC5F685L mice). Compared to wildtype (WT) and hDEPDC5WT mice, hDEPDC5F685L mice exhibited histological signs of mTOR hyperactivation, enlarged neuronal soma, abnormal neurons, and heightened susceptibility to seizures and mortality. Administering rapamycin to hDEPDC5F685L mice starting two weeks after birth normalized neuronal size and mTOR activity, decreased seizure susceptibility and mortality, and showed no effects in the WT or hDEPDC5WT mice. Collectively, these results indicate that the DEPDC5 variant causes abnormal morphology and increased seizure vulnerability through modulation of mTOR signaling.
Collapse
Affiliation(s)
- Jie Liu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Rui Huang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Fenglin Tang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China
| | - Yuanlin Ma
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China; Chongqing Key Laboratory of Cerebrovascular Disease Research, Chongqing, 402160, China; Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing, 402160, China; Key Laboratory of Major Brain Disease and Aging Research (Ministry of Education), Chongqing Medical University, Chongqing, China.
| | - Patrick Kwan
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, Chongqing, China; Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia; Epilepsy Unit, Brain Program, Alfred Hospital, Melbourne, Victoria, Australia.
| |
Collapse
|
14
|
Wang Y, Ping Y, Zhou R, Wang G, Zhang Y, Yang X, Zhao M, Liu D, Kulkarni M, Lamb H, Niu Q, Hardwick JM, Teng X. The Whi2-Psr1-Psr2 complex selectively regulates TORC1 and autophagy under low leucine conditions but not nitrogen depletion. Autophagy 2025:1-17. [PMID: 40103213 DOI: 10.1080/15548627.2025.2481014] [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: 10/07/2024] [Revised: 03/10/2025] [Accepted: 03/14/2025] [Indexed: 03/20/2025] Open
Abstract
Amino acids and ammonia serve as sources of nitrogen for cell growth and were previously thought to have similar effects on yeast. Consistent with this idea, depletion of either of these two nitrogen sources inhibits the target of rapamycin complex 1 (TORC1), leading to induction of macroautophagy/autophagy and inhibition of cell growth. In this study, we show that Whi2 and the haloacid dehalogenase (HAD)-type phosphatases Psr1 and Psr2 distinguish between these two nitrogen sources in Saccharomyces cerevisiae, as the Whi2-Psr1-Psr2 complex inhibits TORC1 in response to low leucine but not in the absence of nitrogen. In contrast, a parallel pathway controlled by Npr2 and Npr3, components of the Seh1-associated complex inhibiting TORC1 (SEACIT), suppress TORC1 under both low leucine- and nitrogen-depletion conditions. Co-immunoprecipitations with mutants of Whi2, Psr1, Psr2 and fragments of Tor1 support the model that Whi2 recruits Psr1 and Psr2 to TORC1. In accordance, the interaction between Whi2 and Tor1 appears to increase under low leucine but decreases under nitrogen-depletion conditions. Although the targets of Psr1 and Psr2 phosphatases are not known, mutation of their active sites abolishes their inhibitory effects on TORC1. Consistent with the conservation of HAD phosphatases across species, human HAD phosphatases CTDSP1 (CTD small phosphatase 1), CTDSP2, and CTDSPL can functionally replace Psr1 and Psr2 in yeast, restoring TORC1 inhibition and autophagy activation in response to low leucine conditions.
Collapse
Affiliation(s)
- Yitao Wang
- International College of Pharmaceutical Innovation, Soochow University, Suzhou, Jiangsu, China
| | - Yang Ping
- International College of Pharmaceutical Innovation, Soochow University, Suzhou, Jiangsu, China
| | - Rui Zhou
- International College of Pharmaceutical Innovation, Soochow University, Suzhou, Jiangsu, China
| | - Guiqin Wang
- International College of Pharmaceutical Innovation, Soochow University, Suzhou, Jiangsu, China
| | - Yu Zhang
- International College of Pharmaceutical Innovation, Soochow University, Suzhou, Jiangsu, China
| | - Xueyu Yang
- International College of Pharmaceutical Innovation, Soochow University, Suzhou, Jiangsu, China
| | - Mingjun Zhao
- International College of Pharmaceutical Innovation, Soochow University, Suzhou, Jiangsu, China
| | - Dongsheng Liu
- International College of Pharmaceutical Innovation, Soochow University, Suzhou, Jiangsu, China
| | - Madhura Kulkarni
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Heather Lamb
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Qingwei Niu
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - J Marie Hardwick
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Xinchen Teng
- International College of Pharmaceutical Innovation, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
15
|
Adams-Brown SE, Reid KZ. The Central FacilitaTOR: Coordinating Transcription and Translation in Eukaryotes. Int J Mol Sci 2025; 26:2845. [PMID: 40243440 PMCID: PMC11989106 DOI: 10.3390/ijms26072845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/11/2025] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
One of the biggest challenges to eukaryotic gene expression is coordinating transcription in the nucleus and protein synthesis in the cytoplasm. However, little is known about how these major steps in gene expression are connected. The Target of Rapamycin (TOR) signaling pathway is crucial in connecting these critical phases of gene expression. Highly conserved among eukaryotic cells, TOR regulates growth, metabolism, and cellular equilibrium in response to changes in nutrients, energy levels, and stress conditions. This review examines the extensive role of TOR in gene expression regulation. We highlight how TOR is involved in phosphorylation, remodeling chromatin structure, and managing the factors that facilitate transcription and translation. Furthermore, the critical functions of TOR extend to processing RNA, assembling RNA-protein complexes, and managing their export from the nucleus, demonstrating its wide-reaching impact throughout the cell. Our discussion emphasizes the integral roles of TOR in bridging the processes of transcription and translation and explores how it orchestrates these complex cellular processes.
Collapse
Affiliation(s)
| | - Ke Zhang Reid
- Department of Biology, Wake Forest University, Winston-Salem, NC 27109, USA
| |
Collapse
|
16
|
Ryspayeva D, Seyhan AA, MacDonald WJ, Purcell C, Roady TJ, Ghandali M, Verovkina N, El-Deiry WS, Taylor MS, Graff SL. Signaling pathway dysregulation in breast cancer. Oncotarget 2025; 16:168-201. [PMID: 40080721 PMCID: PMC11906143 DOI: 10.18632/oncotarget.28701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
This article provides a comprehensive analysis of the signaling pathways implicated in breast cancer (BC), the most prevalent malignancy among women and a leading cause of cancer-related mortality globally. Special emphasis is placed on the structural dynamics of protein complexes that are integral to the regulation of these signaling cascades. Dysregulation of cellular signaling is a fundamental aspect of BC pathophysiology, with both upstream and downstream signaling cascade activation contributing to cellular process aberrations that not only drive tumor growth, but also contribute to resistance against current treatments. The review explores alterations within these pathways across different BC subtypes and highlights potential therapeutic strategies targeting these pathways. Additionally, the influence of specific mutations on therapeutic decision-making is examined, underscoring their relevance to particular BC subtypes. The article also discusses both approved therapeutic modalities and ongoing clinical trials targeting disrupted signaling pathways. However, further investigation is necessary to fully elucidate the underlying mechanisms and optimize personalized treatment approaches.
Collapse
Affiliation(s)
- Dinara Ryspayeva
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
| | - Attila A. Seyhan
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
- Pathobiology Graduate Program, Brown University, RI 02903, USA
| | - William J. MacDonald
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
| | - Connor Purcell
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
| | - Tyler J. Roady
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
- Pathobiology Graduate Program, Brown University, RI 02903, USA
| | - Maryam Ghandali
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
| | - Nataliia Verovkina
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
| | - Wafik S. El-Deiry
- Laboratory of Translational Oncology and Experimental Cancer Therapeutics, Warren Alpert Medical School, Brown University, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
- Pathobiology Graduate Program, Brown University, RI 02903, USA
- Department of Medicine, Hematology/Oncology Division, Lifespan Health System and Brown University, RI 02903, USA
| | - Martin S. Taylor
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, RI 02903, USA
- Joint Program in Cancer Biology, Lifespan Health System and Brown University, RI 02903, USA
- Legorreta Cancer Center at Brown University, RI 02903, USA
- Pathobiology Graduate Program, Brown University, RI 02903, USA
- Brown Center on the Biology of Aging, Brown University, RI 02903, USA
| | - Stephanie L. Graff
- Legorreta Cancer Center at Brown University, RI 02903, USA
- Department of Medicine, Hematology/Oncology Division, Lifespan Health System and Brown University, RI 02903, USA
| |
Collapse
|
17
|
Liao J, Zhang P, Yin J, Zhang X. New insights into the effects of dietary amino acid composition on meat quality in pigs: A review. Meat Sci 2025; 221:109721. [PMID: 39642438 DOI: 10.1016/j.meatsci.2024.109721] [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: 07/30/2024] [Revised: 11/11/2024] [Accepted: 12/01/2024] [Indexed: 12/09/2024]
Abstract
Pork is an affordable protein source with higher nutrient density. In recent years, meat quality in pigs is getting increasing attention, which has a direct impact on the economic value of pork. Dietary amino acids play a key role in pig production, not only regulating pig growth and health, but also contributing significantly to meat quality. In this review, we discuss the effect of skeletal muscle composition on meat quality. Importantly, we summarize the levels of essential amino acids (EAAs), such as lysine, methionine, threonine, tryptophan and branched-chain amino acids (BCAAs), in diets for finishing pigs to improve meat quality. The beneficial effects of flavor amino acids on meat quality, including flavor production, muscle fiber-type composition and intramuscular fat deposition, are further systematically summarized. We also focus on the impact of dietary amino acid levels on environmental benefits, although research in this area is still limited. Considering that the previously established EAA requirements are based on the principle of maximizing growth rate and feed conversion, this review will provide new insights into the effects of dietary amino acids on aspects of meat quality and highlight the current gaps to promote future research.
Collapse
Affiliation(s)
- Jialong Liao
- Frontier Science Center of Molecular Design Breeding, Ministry of Education, Beijing 100193, China; State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Pengguang Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Jingdong Yin
- Frontier Science Center of Molecular Design Breeding, Ministry of Education, Beijing 100193, China; State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Xin Zhang
- Frontier Science Center of Molecular Design Breeding, Ministry of Education, Beijing 100193, China; State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
18
|
Wang X, Liu T, Sheng Y, Qiu C, Zhang Y, Liu Y, Wu C. Exploration and verification of circulating diagnostic biomarkers in osteoarthritis based on machine learning. Front Genet 2025; 16:1513675. [PMID: 40034747 PMCID: PMC11872907 DOI: 10.3389/fgene.2025.1513675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 01/23/2025] [Indexed: 03/05/2025] Open
Abstract
Background Osteoarthritis (OA) is a prevalent chronic joint condition. This study sought to explore potential diagnostic biomarkers for OA and assess their relevance in clinical samples. Methods We searched the GEO database for peripheral blood leukocytes expression profiles of OA patients as a training set to conduct differentially expressed gene (DEG) analysis. Two machine learning algorithms, least absolute shrinkage and selection operator (LASSO) logistic regression and support vector machine-recursive feature elimination (SVM-RFE), were employed to identify candidate biomarkers for OA diagnosis. The performance was assessed using receiver operating characteristic (ROC) curves, and the areas under the curve (AUCs) with 95% confidence interval (CI) were calculated. Furthermore, we gathered clinical peripheral blood samples from healthy donors and OA patients (validation set) to validate our findings. Small interfering RNA and CCK8 proliferation assay were used for experimental verification. Results A total of 31 DEGs were discovered, and the machine learning screening found five DEGs that were considered to be candidate biomarkers. Notably, BIRC2 had a very good discriminatory effect among the five candidate biomarkers, with an AUC of 0.814 (95% CI: 0.697-0.915). In our validation set, results showed that the levels of BIRC2 and SEH1L were remarkably higher in healthy donors than OA patients, consistent with the results of the training set. SEH1L owned the largest AUC of 0.964 (95% CI: 0.855-1.000). BIRC2 also displayed a larger AUC of 0.836 (95% CI: 0.618-1.000) in the training set. Knockdown of these two genes could significantly suppress human chondrocyte proliferation. Conclusion Two novel biomarkers, SEH1L and BIRC2, were indicated to have the capacity to differentiate healthy people from OA patients at the peripheral level. Experiments have shown that knockdown of these two genes could inhibit human chondrocyte proliferation, as verified by cell proliferation assays.
Collapse
Affiliation(s)
- Xinyu Wang
- Department of Molecular Orthopaedics, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
- Department of Anaesthesia, National Center for Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Tianyi Liu
- Department of Molecular Orthopaedics, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yueyang Sheng
- Department of Molecular Orthopaedics, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Cheng Qiu
- Department of Orthopedic Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yanzhuo Zhang
- Department of Molecular Orthopaedics, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| | - Yanqun Liu
- Department of Molecular Orthopaedics, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
- Trauma Reconstructive and Plastic Clinical Research Center, Yanbian University Hospital, Jilin, China
| | - Chengai Wu
- Department of Molecular Orthopaedics, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
19
|
Meraz IM, Majidi M, Song R, Meng F, Gao L, Wang Q, Wang J, Shpall EJ, Roth JA. NPRL2 gene therapy induces effective antitumor immunity in KRAS/STK11 mutant anti-PD1 resistant metastatic non-small cell lung cancer (NSCLC) in a humanized mouse model. eLife 2025; 13:RP98258. [PMID: 39932765 PMCID: PMC11813225 DOI: 10.7554/elife.98258] [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] [Indexed: 02/13/2025] Open
Abstract
Expression of NPRL2/TUSC4, a tumor-suppressor gene, is reduced in many cancers including NSCLC. Restoration of NPRL2 induces DNA damage, apoptosis, and cell-cycle arrest. We investigated NPRL2 antitumor immune responses in aPD1R/KRAS/STK11mt NSCLC in humanized-mice. Humanized-mice were generated by transplanting fresh human cord blood-derived CD34 stem cells into sub-lethally irradiated NSG mice. Lung-metastases were developed from KRAS/STK11mt/aPD1R A549 cells and treated with NPRL2 w/wo pembrolizumab. NPRL2-treatment reduced lung metastases significantly, whereas pembrolizumab was ineffective. Antitumor effect was greater in humanized than non-humanized-mice. NPRL2 + pembrolizumab was not synergistic in KRAS/STK11mt/aPD1R tumors but was synergistic in KRASwt/aPD1S H1299. NPRL2 also showed a significant antitumor effect on KRASmt/aPD1R LLC2 syngeneic-tumors. The antitumor effect was correlated with increased infiltration of human cytotoxic-T, HLA-DR+DC, CD11c+DC, and downregulation of myeloid and regulatory-T cells in TME. Antitumor effect was abolished upon in-vivo depletion of CD8-T, macrophages, and CD4-T cells whereas remained unaffected upon NK-cell depletion. A distinctive protein-expression profile was found after NPRL2 treatment. IFNγ, CD8b, and TBX21 associated with T-cell functions were significantly increased, whereas FOXP3, TGFB1/B2, and IL-10RA were strongly inhibited by NPRL2. A list of T-cell co-inhibitory molecules was also downregulated. Restoration of NPRL2 exhibited significantly slower tumor growth in humanized-mice, which was associated with increased presence of human cytotoxic-T, and DC and decreased percentage of Treg, MDSC, and TAM in TME. NPRL2-stable cells showed a substantial increase in colony-formation inhibition and heightened sensitivity to carboplatin. Stable-expression of NPRL2 resulted in the downregulation of MAPK and AKT-mTOR signaling. Taken-together, NPRL2 gene-therapy induces antitumor activity on KRAS/STK11mt/aPD1R tumors through DC-mediated antigen-presentation and cytotoxic immune-cell activation.
Collapse
Affiliation(s)
- Ismail M Meraz
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, HoustonHoustonUnited States
| | - Mourad Majidi
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, HoustonHoustonUnited States
| | - Renduo Song
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, HoustonHoustonUnited States
| | - Feng Meng
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, HoustonHoustonUnited States
| | - Lihui Gao
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, HoustonHoustonUnited States
| | - Qi Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, HoustonHoustonUnited States
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, HoustonHoustonUnited States
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation, The University of Texas MD Anderson Cancer Center, HoustonHoustonUnited States
| | - Jack A Roth
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, HoustonHoustonUnited States
| |
Collapse
|
20
|
Ke Y, Peng X, Song C, Fang X, Wang Y, Zhang C. Animal amino acid sensor - A review. Anim Biosci 2025; 38:198-208. [PMID: 39210799 PMCID: PMC11725744 DOI: 10.5713/ab.24.0366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/20/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
Cell growth and metabolism necessitate the involvement of amino acids, which are sensed and integrated by the mammalian target of rapamycin complex 1 (mTORC1). However, the molecular mechanisms underlying amino acid sensing remain poorly understood. Research indicates that amino acids are detected by specific sensors, with the signals being relayed to mTORC1 indirectly. This paper reviews the structures and biological functions of the amino acid sensors identified thus far. Additionally, it evaluates the potential role these sensors play in the developmental changes of the livestock production.
Collapse
Affiliation(s)
- Yongding Ke
- School of Life Science, Jiangsu Normal University, Jiangsu 221116,
China
| | - Xinyu Peng
- School of Life Science, Jiangsu Normal University, Jiangsu 221116,
China
| | - Chengchuang Song
- School of Life Science, Jiangsu Normal University, Jiangsu 221116,
China
| | - Xingtang Fang
- School of Life Science, Jiangsu Normal University, Jiangsu 221116,
China
| | - Yanhong Wang
- School of Life Science, Jiangsu Normal University, Jiangsu 221116,
China
| | - Chunlei Zhang
- School of Life Science, Jiangsu Normal University, Jiangsu 221116,
China
| |
Collapse
|
21
|
Zhang F, Xiong X, Li Z, Wang H, Wang W, Zhao Y, Sun Y. RHEB neddylation by the UBE2F-SAG axis enhances mTORC1 activity and aggravates liver tumorigenesis. EMBO J 2025; 44:1185-1219. [PMID: 39762645 PMCID: PMC11832924 DOI: 10.1038/s44318-024-00353-5] [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: 06/02/2024] [Revised: 11/27/2024] [Accepted: 12/04/2024] [Indexed: 02/19/2025] Open
Abstract
Small GTPase RHEB is a well-known mTORC1 activator, whereas neddylation modifies cullins and non-cullin substrates to regulate their activity, subcellular localization and stability. Whether and how RHEB is subjected to neddylation modification remains unknown. Here, we report that RHEB is a substrate of NEDD8-conjugating E2 enzyme UBE2F. In cell culture, UBE2F depletion inactivates mTORC1, inhibiting cell cycle progression, cell growth and inducing autophagy. Mechanistically, UBE2F cooperates with E3 ligase SAG in neddylation of RHEB at K169 to enhance its lysosome localization and GTP-binding affinity. Furthermore, liver-specific Ube2f knockout attenuates steatosis and tumorigenesis induced by Pten loss in an mTORC1-dependent manner, suggesting a causal role of UBE2F in liver tumorigenesis. Finally, UBE2F expression levels and mTORC1 activity correlate with patient survival in hepatocellular carcinoma. Collectively, our study identifies RHEB as neddylation substrate of the UBE2F-SAG axis, and highlights the UBE2F-SAG axis as a potential target for the treatment of non-alcoholic fatty liver disease and hepatocellular carcinoma.
Collapse
Affiliation(s)
- Fengwu Zhang
- Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, 310009, Hangzhou, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, 310029, Hangzhou, China
| | - Xiufang Xiong
- Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, 310009, Hangzhou, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, 310029, Hangzhou, China
| | - Zhijian Li
- Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, 310009, Hangzhou, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, 310029, Hangzhou, China
| | - Haibo Wang
- Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, 310009, Hangzhou, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, 310029, Hangzhou, China
| | - Weilin Wang
- Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, China
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, 310009, Hangzhou, China
| | - Yongchao Zhao
- Institute of Translational Medicine, Zhejiang University School of Medicine, 310029, Hangzhou, China.
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Cancer Center, Zhejiang University, 310058, Hangzhou, China.
| | - Yi Sun
- Cancer Institute, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009, Hangzhou, China.
- Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, 310009, Hangzhou, China.
- Institute of Translational Medicine, Zhejiang University School of Medicine, 310029, Hangzhou, China.
- Cancer Center, Zhejiang University, 310058, Hangzhou, China.
- Leading Innovative and Entrepreneur Team Introduction Program of Zhejiang, Hangzhou, China.
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, 310053, Hangzhou, China.
- Institute of Fundamental and Transdisciplinary Research Zhejiang University, Hangzhou, China.
| |
Collapse
|
22
|
Dentel B, Angeles-Perez L, Flores AY, Lei K, Ren C, Sanchez AP, Tsai PT. Neuronal cell type specific roles for Nprl2 in neurodevelopmental disorder-relevant behaviors. Neurobiol Dis 2025; 205:106790. [PMID: 39765274 DOI: 10.1016/j.nbd.2025.106790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 01/03/2025] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
Loss of function in the subunits of the GTPase-activating protein (GAP) activity toward Rags-1 (GATOR1) complex, an amino-acid sensitive negative regulator of the mechanistic target of rapamycin complex 1 (mTORC1), is implicated in both genetic familial epilepsies and Neurodevelopmental Disorders (NDDs) (Baldassari et al., 2018). Previous studies have found seizure phenotypes and increased activity resulting from conditional deletion of GATOR1 function from forebrain excitatory neurons (Yuskaitis et al., 2018; Dentel et al., 2022); however, studies focused on understanding mechanisms contributing to NDD-relevant behaviors are lacking, especially studies understanding the contributions of GATOR1's critical GAP catalytic subunit, nitrogen permease regulator like-2 (Nprl2). Given the clinical phenotypes observed in patients with Nprl2 mutations, in this study, we sought to investigate the neuronal cell type contributions of Nprl2 to NDD behaviors. We conditionally deleted Nprl2 broadly in most neurons (Synapsin1cre), in inhibitory neurons only (Vgatcre), and in Purkinje cells within the cerebellum (L7cre). Broad neuronal deletion of Nprl2 resulted in seizures, social and learning deficits, and hyperactivity. In contrast, deleting Nprl2 from inhibitory neurons led to increased motor learning, hyperactive behavior, in addition to social and learning deficits. Lastly, Purkinje cell (PC) loss of Nprl2 also led to learning and social deficits but did not affect locomotor activity. These phenotypes enhance understanding of the spectrum of disease found in human populations with GATOR1 loss of function and highlight the significance of distinct cellular populations to NDD-related behaviors. SIGNIFICANCE STATEMENT: We aim to elucidate the neuronal-specific contributions of nitrogen permease regulator like-2 (Nprl2) to its neurodevelopmental disorder (NDD)-relevant phenotypes. We conditionally deleted Nprl2 broadly in neurons (Syn1cre), in inhibitory neurons (Vgatcre), and in cerebellar Purkinje cells (L7cre). We identify seizures only in the Syn1cre conditional mutant (cKO); hyperactivity, learning difficulties, social deficits, and impulsivity in the Syn1cre and Vgatcre cKOs; and social deficits, and fear learning deficits in L7cre cKOs. To our knowledge, we are the first to describe the behavioral contributions of Nprl2's function across multiple cell types. Our findings highlight both critical roles for Nprl2 in learning and behavior and also distinct contributions of select neuronal populations to these NDD-relevant behaviors.
Collapse
Affiliation(s)
- Brianne Dentel
- The University of Texas Southwestern Medical Center, Department of Neurology, Dallas, TX, United States of America
| | - Lidiette Angeles-Perez
- The University of Texas Southwestern Medical Center, Department of Neurology, Dallas, TX, United States of America
| | - Abigail Y Flores
- The University of Texas Southwestern Medical Center, Department of Neurology, Dallas, TX, United States of America
| | - Katherine Lei
- The University of Texas Southwestern Medical Center, Department of Neurology, Dallas, TX, United States of America
| | - Chongyu Ren
- The University of Texas Southwestern Medical Center, Department of Neurology, Dallas, TX, United States of America
| | - Andrea Pineda Sanchez
- The University of Texas Southwestern Medical Center, Department of Neurology, Dallas, TX, United States of America
| | - Peter T Tsai
- The University of Texas Southwestern Medical Center, Department of Neurology, Dallas, TX, United States of America; The University of Texas Southwestern Medical Center, Department of Psychiatry, Dallas, TX, United States of America; The University of Texas Southwestern Medical Center, Department of Pediatrics, Dallas, TX, United States of America; The University of Texas Southwestern Medical Center, Department of Neuroscience; O'Donnell Brain Institute, Dallas, TX, United States of America.
| |
Collapse
|
23
|
Zhang W, Wu H, Liao Y, Zhu C, Zou Z. Caspase family in autoimmune diseases. Autoimmun Rev 2025; 24:103714. [PMID: 39638102 DOI: 10.1016/j.autrev.2024.103714] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/28/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024]
Abstract
Programmed cell death (PCD) plays a crucial role in maintaining tissue homeostasis, with its primary forms including apoptosis, pyroptosis, and necroptosis. The caspase family is central to these processes, and its complex functions across different cell death pathways and other non-cell death roles have been closely linked to the pathogenesis of autoimmune diseases. This article provides a comprehensive review of the role of the caspase family in autoimmune diseases such as rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), type 1 diabetes (T1D), and multiple sclerosis (MS). It particularly emphasizes the intricate functions of caspases within various cell death pathways and their potential as therapeutic targets, thereby offering innovative insights and a thorough discussion in this field. In terms of therapy, strategies targeting caspases hold significant promise. We emphasize the importance of a holistic understanding of caspases in the overall concept of cell death, exploring their unique functions and interrelationships across multiple cell death pathways, including apoptosis, pyroptosis, necroptosis, and PANoptosis. This approach transcends the limitations of previous studies that focused on singular cell death pathways. Additionally, caspases play a key role in non-cell death functions, such as immune cell activation, cytokine processing, inflammation regulation, and tissue repair, thereby opening new avenues for the treatment of autoimmune diseases. Regulating caspase activity holds the potential to restore immune balance in autoimmune diseases. Potential therapeutic approaches include small molecule inhibitors (both reversible and irreversible), biological agents (such as monoclonal antibodies), and gene therapies. However, achieving specific modulation of caspases to avoid interference with normal physiological functions remains a major challenge. Future research must delve deeper into the regulatory mechanisms of caspases and their associated complexes linked to PANoptosis to facilitate precision medicine. In summary, this article offers a comprehensive and in-depth analysis, providing a novel perspective on the complex roles of caspases in autoimmune diseases, with the potential to catalyze breakthroughs in understanding disease mechanisms and developing therapeutic strategies.
Collapse
Affiliation(s)
- Wangzheqi Zhang
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China; School of Anesthesiology, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Huang Wu
- Basic Medical University, Naval Medical University, Shanghai 200433, China
| | - Yan Liao
- School of Anesthesiology, Naval Medical University, 168 Changhai Road, Shanghai 200433, China
| | - Chenglong Zhu
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China; School of Anesthesiology, Naval Medical University, 168 Changhai Road, Shanghai 200433, China.
| | - Zui Zou
- Faculty of Anesthesiology, Changhai Hospital, Naval Medical University, Shanghai 200433, China; School of Anesthesiology, Naval Medical University, 168 Changhai Road, Shanghai 200433, China.
| |
Collapse
|
24
|
He L, Cho S, Blenis J. mTORC1, the maestro of cell metabolism and growth. Genes Dev 2025; 39:109-131. [PMID: 39572234 PMCID: PMC11789495 DOI: 10.1101/gad.352084.124] [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] [Indexed: 01/11/2025]
Abstract
The mechanistic target of rapamycin (mTOR) pathway senses and integrates various environmental and intracellular cues to regulate cell growth and proliferation. As a key conductor of the balance between anabolic and catabolic processes, mTOR complex 1 (mTORC1) orchestrates the symphonic regulation of glycolysis, nucleic acid and lipid metabolism, protein translation and degradation, and gene expression. Dysregulation of the mTOR pathway is linked to numerous human diseases, including cancer, neurodegenerative disorders, obesity, diabetes, and aging. This review provides an in-depth understanding of how nutrients and growth signals are coordinated to influence mTOR signaling and the extensive metabolic rewiring under its command. Additionally, we discuss the use of mTORC1 inhibitors in various aging-associated metabolic diseases and the current and future potential for targeting mTOR in clinical settings. By deciphering the complex landscape of mTORC1 signaling, this review aims to inform novel therapeutic strategies and provide a road map for future research endeavors in this dynamic and rapidly evolving field.
Collapse
Affiliation(s)
- Long He
- Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10021, USA;
- Department of Pharmacology, Weill Cornell Medicine, New York, New York 10021, USA
| | - Sungyun Cho
- Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10021, USA
- Department of Pharmacology, Weill Cornell Medicine, New York, New York 10021, USA
| | - John Blenis
- Meyer Cancer Center, Weill Cornell Medicine, New York, New York 10021, USA;
- Department of Pharmacology, Weill Cornell Medicine, New York, New York 10021, USA
| |
Collapse
|
25
|
Chen J, Ou Y, Liu Y. Reply to: Amino acids and KLHL22 do not activate mTORC1 via DEPDC5 degradation. Nature 2025; 637:E15-E17. [PMID: 39780003 DOI: 10.1038/s41586-024-07975-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Affiliation(s)
- Jie Chen
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Yuhui Ou
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Ying Liu
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, China.
| |
Collapse
|
26
|
Gu X. Determination of Nutrient Ligand-Sensor Binding Affinity. Methods Mol Biol 2025; 2882:163-178. [PMID: 39992509 DOI: 10.1007/978-1-0716-4284-9_8] [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] [Indexed: 02/25/2025]
Abstract
Cells contain dedicated mechanisms to sense nutrient levels in the environment to regulate their growth by balancing anabolism and catabolism [1, 2]. The mechanistic Target of Rapamycin Complex 1 (mTORC1), a multi-protein kinase complex, serves as an essential growth regulator that integrates various upstream inputs including growth factors and nutrients like amino acids [1, 2] Nutrient sensors upstream of mTORC1 directly bind cognate nutrient ligands to convey their availability and thereby regulate mTORC1 signaling [1, 2]. A reliable method is needed to quantitatively determine the binding affinity (Kd) of the nutrient sensor to its ligand. In parallel, quantitative metabolomic analysis can reveal metabolite levels in fed and starved cells; which represent the physiological range of the nutrient of interest. Whether or not the binding affinity is within the physiological range serves as an indicator to determine the physiological relevance of the sensing mechanism. This chapter describes a generalizable protocol that allows reproducible determination of nutrient ligand-nutrient sensor binding affinity. Here, the S-adenosylmethionine (nutrient ligand)-SAMTOR (nutrient sensor) pair is used as an example [3]. Nutrient sensor purification, radioactive nutrient ligand incubation, and eventual scintillation counting are included, along with a description of the mathematical equation that is used to calculate the binding affinity.
Collapse
Affiliation(s)
- Xin Gu
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA.
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
27
|
Zhao D, Xu R, Zhou Y, Wu J, Zhang X, Lin H, Wang J, Ding Z, Zou Y. ORP5 promotes cardiac hypertrophy by regulating the activation of mTORC1 on lysosome. J Adv Res 2024:S2090-1232(24)00591-5. [PMID: 39667666 DOI: 10.1016/j.jare.2024.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/07/2024] [Accepted: 12/09/2024] [Indexed: 12/14/2024] Open
Abstract
INTRODUCTION Oxysterol binding protein (OSBP)-related protein 5 (ORP5) mainly functions as a lipid transfer protein at membrane contact sites (MCS). ORP5 facilitates cell proliferation through the activation of mTORC1 signaling. While the pro-hypertrophic effects of mTORC1 are well-documented, the specific role of ORP5 in the context of pathological cardiac hypertrophy remains inadequately understood. METHODS To investigate the role of ORP5 in pathological cardiac hypertrophy, AAV9-treated mice and neonatal rat ventricular myocytes (NRVMs) were utilized. Cardiac function, morphology, and mTORC1 signaling alterations induced by pro-hypertrophic stimuli were assessed in both myocardium and NRVMs. Additionally, a range of molecular techniques were employed to elucidate the regulatory mechanisms of ORP5 on mTORC1 in hypertrophied hearts. RESULTS Increased expression of ORP5 was observed in the hearts of patients with hypertrophic cardiomyopathy (HCM), in mice subjected to transverse aortic constriction (TAC), and in NRVMs treated with angiotensin II (AngII). We found that ORP5 binds to mTOR in cardiomyocytes. Upon exposure to TAC surgery, ORP5-deficient hearts exhibited enhanced cardiac function, reduced cardiomyocyte hypertrophy, and diminished collagen deposition than wild type. Conversely, overexpression of ORP5 significantly aggravated hypertrophic responses in both hearts and NRVMs. Notably, the promotion of cardiac hypertrophy induced by ORP5 overexpression was reversed by rapamycin, an inhibitor of mTORC1. Mechanistically, our study elucidated that the ORD domain of ORP5 interacts with mTORC1, facilitating its translocation to the outer membrane of the lysosome for subsequent activation. This activation triggers the downstream signaling pathways involving S6K1 and 4E-BP1, which initiate protein synthesis, thereby promoting pathological cardiac hypertrophy. CONCLUSIONS Our findings provide the inaugural evidence that ORP5 facilitates pathological ventricular hypertrophy through the translocation of mTORC1 to the lysosome for subsequent activation. Consequently, ORP5 has the potential to serve as a diagnostic biomarker or therapeutic target for pathological cardiac hypertrophy in the future.
Collapse
Affiliation(s)
- Di Zhao
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, China; NHC Key Laboratory of Ischemic Heart Diseases, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China; National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Ran Xu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, China; NHC Key Laboratory of Ischemic Heart Diseases, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China; National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Yufei Zhou
- Department of Cardiology, State Key Laboratory of Trans-vascular Implantation Devices, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiaying Wu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, China; NHC Key Laboratory of Ischemic Heart Diseases, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China; National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Xiaoxue Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, China; NHC Key Laboratory of Ischemic Heart Diseases, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China; National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Hong Lin
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, China; NHC Key Laboratory of Ischemic Heart Diseases, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China; National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Jienan Wang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, China; NHC Key Laboratory of Ischemic Heart Diseases, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China; National Clinical Research Center for Interventional Medicine, Shanghai, China
| | - Zhiwen Ding
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, China; NHC Key Laboratory of Ischemic Heart Diseases, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China; National Clinical Research Center for Interventional Medicine, Shanghai, China.
| | - Yunzeng Zou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, China; State Key Laboratory of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, China; NHC Key Laboratory of Ischemic Heart Diseases, China; Key Laboratory of Viral Heart Diseases, Chinese Academy of Medical Sciences, China; National Clinical Research Center for Interventional Medicine, Shanghai, China; Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
28
|
Tucker SK, Eberhart JK. The convergence of mTOR signaling and ethanol teratogenesis. Reprod Toxicol 2024; 130:108720. [PMID: 39306261 DOI: 10.1016/j.reprotox.2024.108720] [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: 07/01/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 10/04/2024]
Abstract
Ethanol is one of the most common teratogens and causes of human developmental disabilities. Fetal alcohol spectrum disorders (FASD), which describes the wide range of deficits due to prenatal ethanol exposure, are estimated to affect between 1.1 % and 5.0 % of births in the United States. Ethanol dysregulates numerous cellular mechanisms such as programmed cell death (apoptosis), protein synthesis, autophagy, and various aspects of cell signaling, all of which contribute to FASD. The mechanistic target of rapamycin (mTOR) regulates these cellular mechanisms via sensing of nutrients like amino acids and glucose, DNA damage, and growth factor signaling. Despite an extensive literature on ethanol teratogenesis and mTOR signaling, there has been less attention paid to their interaction. Here, we discuss the impact of ethanol teratogenesis on mTORC1's ability to coordinate growth factor and amino acid sensing with protein synthesis, autophagy, and apoptosis. Notably, the effect of ethanol exposure on mTOR signaling depends on the timing and dose of ethanol as well as the system studied. Overall, the overlap between the functions of mTORC1 and the phenotypes observed in FASD suggest a mechanistic interaction. However, more work is required to fully understand the impact of ethanol teratogenesis on mTOR signaling.
Collapse
Affiliation(s)
- Scott K Tucker
- Department of Molecular Biosciences, Waggoner Center for Alcohol and Addiction Research and Institute for Neuroscience, University of Texas, Austin, TX, USA
| | - Johann K Eberhart
- Department of Molecular Biosciences, Waggoner Center for Alcohol and Addiction Research and Institute for Neuroscience, University of Texas, Austin, TX, USA.
| |
Collapse
|
29
|
Ye W, Sun L, Fu C, Dong H, Zhou T. A Novel Lysosome-Related Gene Signature Predicts Lung Cancer Prognosis: A Bioinformatics-Driven Study. Health Sci Rep 2024; 7:e70236. [PMID: 39633837 PMCID: PMC11615650 DOI: 10.1002/hsr2.70236] [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: 03/24/2024] [Revised: 11/05/2024] [Accepted: 11/13/2024] [Indexed: 12/07/2024] Open
Abstract
Background and Aims The biological function of lysosomes has been increasingly appreciated in cancer. However, the relationship between lysosome and lung adenocarcinoma (LUAD) was not well understood. In this study, a lysosome-related signature was developed for LUAD risk stratification and prognosis prediction. Methods Download RNA-seq data of LUAD and clinical information of corresponding samples from the UCSC-Xena platform. GSE31210 databases is used as a validation cohort. The lysosome-related genes was obtained from molecular signature database. The differentially expressed genes (DEGs) as well as lysosome-associated prognosis signatures were identified by using univariate, multivariate cox, and Lasso regression. A nomogram was constructed and evaluated using ROC and DCA. Results A total of 109 lysosome-related DEGs were identified and 30 prognostic related DEGs were subsequently screened. Cluster analysis further divides the TCGA cohort into clusters 1 and 2. Patients in cluster 2 had a worse prognosis (p = 0.016), lower LYSOSOME score. Enrichment analysis showed that 21 significantly enriched gene sets in the cluster 2 were activated. And 10 pathways, such as E2F_TARGETS, G2M_CHECKPOINT were upregulated. Multivariate Cox regression analysis identified 17 best prognostic genes as risk signature. An independent prognostic factor, the risk signature, was identified. A prognostic nomogram including risk signature, age, TNM stage, and gender was constructed, and ROC and DCA curves proved its excellent performance. We examined CTSZ and AP3S2 protein expression in 48 stage 3-4 NSCLC samples. Low AP3S2 expression was associated with better prognosis (median overall survival: 37.87 vs. 8.53 months, p = 0.0211). Increased CTSZ expression also indicated better prognosis (median overall survival: 6.77 vs. 30.50 months, p = 0.0306). Conclusion We identified molecular subtypes and lysosomal-based prognostic signatures for LUAD patients, as well as 17 genes that serve as a biomarker for evaluating the prognosis of LUAD patients.
Collapse
Affiliation(s)
- Wei Ye
- Department of OncologyChangzhou Tumor HospitalChangzhouChina
| | - Lin Sun
- Department of OncologyChangzhou Tumor HospitalChangzhouChina
| | - Cong Fu
- Department of OncologyChangzhou Tumor HospitalChangzhouChina
| | - Huajie Dong
- Department of OncologyChangzhou Tumor HospitalChangzhouChina
| | - Tong Zhou
- Department of OncologyChangzhou Tumor HospitalChangzhouChina
| |
Collapse
|
30
|
Zwakenberg S, Westland D, van Es RM, Rehmann H, Anink J, Ciapaite J, Bosma M, Stelloo E, Liv N, Sobrevals Alcaraz P, Verhoeven-Duif NM, Jans JJM, Vos HR, Aronica E, Zwartkruis FJT. mTORC1 restricts TFE3 activity by auto-regulating its presence on lysosomes. Mol Cell 2024; 84:4368-4384.e6. [PMID: 39486419 DOI: 10.1016/j.molcel.2024.10.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 07/10/2024] [Accepted: 10/07/2024] [Indexed: 11/04/2024]
Abstract
To stimulate cell growth, the protein kinase complex mTORC1 requires intracellular amino acids for activation. Amino-acid sufficiency is relayed to mTORC1 by Rag GTPases on lysosomes, where growth factor signaling enhances mTORC1 activity via the GTPase Rheb. In the absence of amino acids, GATOR1 inactivates the Rags, resulting in lysosomal detachment and inactivation of mTORC1. We demonstrate that in human cells, the release of mTORC1 from lysosomes depends on its kinase activity. In accordance with a negative feedback mechanism, activated mTOR mutants display low lysosome occupancy, causing hypo-phosphorylation and nuclear localization of the lysosomal substrate TFE3. Surprisingly, mTORC1 activated by Rheb does not increase the cytoplasmic/lysosomal ratio of mTORC1, indicating the existence of mTORC1 pools with distinct substrate specificity. Dysregulation of either pool results in aberrant TFE3 activity and may explain nuclear accumulation of TFE3 in epileptogenic malformations in focal cortical dysplasia type II (FCD II) and tuberous sclerosis (TSC).
Collapse
Affiliation(s)
- Susan Zwakenberg
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Denise Westland
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Robert M van Es
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Holger Rehmann
- Department of Energy and Life Science, Flensburg University of Applied Sciences, Flensburg, Germany
| | - Jasper Anink
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Jolita Ciapaite
- Department of Genetics, Section Metabolic Diagnostics, University Medical Center Utrecht, 3584 EA Utrecht, the Netherlands
| | - Marjolein Bosma
- Department of Genetics, Section Metabolic Diagnostics, University Medical Center Utrecht, 3584 EA Utrecht, the Netherlands
| | - Ellen Stelloo
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Nalan Liv
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Paula Sobrevals Alcaraz
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Nanda M Verhoeven-Duif
- Department of Genetics, Section Metabolic Diagnostics, University Medical Center Utrecht, 3584 EA Utrecht, the Netherlands
| | - Judith J M Jans
- Department of Genetics, Section Metabolic Diagnostics, University Medical Center Utrecht, 3584 EA Utrecht, the Netherlands
| | - Harmjan R Vos
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Eleonora Aronica
- Department of (Neuro)Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, the Netherlands; Stichting Epilepsie Instellingen Nederland (SEIN), Heemstede, the Netherlands
| | - Fried J T Zwartkruis
- Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands.
| |
Collapse
|
31
|
Acharya A, Demetriades C. mTORC1 activity licenses its own release from the lysosomal surface. Mol Cell 2024; 84:4385-4400.e7. [PMID: 39486418 DOI: 10.1016/j.molcel.2024.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 08/17/2024] [Accepted: 10/07/2024] [Indexed: 11/04/2024]
Abstract
Nutrient signaling converges on mTORC1, which, in turn, orchestrates a physiological cellular response. A key determinant of mTORC1 activity is its shuttling between the lysosomal surface and the cytoplasm, with nutrients promoting its recruitment to lysosomes by the Rag GTPases. Active mTORC1 regulates various cellular functions by phosphorylating distinct substrates at different subcellular locations. Importantly, how mTORC1 that is activated on lysosomes is released to meet its non-lysosomal targets and whether mTORC1 activity itself impacts its localization remain unclear. Here, we show that, in human cells, mTORC1 inhibition prevents its release from lysosomes, even under starvation conditions, which is accompanied by elevated and sustained phosphorylation of its lysosomal substrate TFEB. Mechanistically, "inactive" mTORC1 causes persistent Rag activation, underlining its release as another process actively mediated via the Rags. In sum, we describe a mechanism by which mTORC1 controls its own localization, likely to prevent futile cycling on and off lysosomes.
Collapse
Affiliation(s)
- Aishwarya Acharya
- Max Planck Institute for Biology of Ageing (MPI-AGE), 50931 Cologne, Germany; Cologne Graduate School of Ageing Research (CGA), 50931 Cologne, Germany
| | - Constantinos Demetriades
- Max Planck Institute for Biology of Ageing (MPI-AGE), 50931 Cologne, Germany; Cologne Graduate School of Ageing Research (CGA), 50931 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany.
| |
Collapse
|
32
|
Chen Y, Xu W, Jin H, Zhang M, Liu S, Liu Y, Zhang H. Nutritional Glutamine-Modified Iron-Delivery System with Enhanced Endocytosis for Ferroptosis Therapy of Pancreatic Tumors. ACS NANO 2024; 18:31846-31868. [PMID: 39512234 DOI: 10.1021/acsnano.4c08083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
Heterogeneous reprogrammed nutrient metabolic networks formed by oncogenes exhibit the potential for exploring novel druggable targets and developing innovative anticancer therapeutics. Herein, based on the heterogeneous metabolic characteristics of glutamine (Gln) addiction in pancreatic cancer cells, an iron-delivery system (IDS) with enhanced endocytosis is designed for efficient ferroptosis therapy. The IDS is characterized by Gln modification and can be recognized as a source of Gln nutrients for efficient endocytic uptake by pancreatic tumor cells. Because the IDS is flexible to combine with amino acid-like components, the IDS with enhanced endocytosis is further produced by loading the Gln transporter inhibitor of V9302. V9302 is capable of suppressing molecular Gln uptake via transporter ASCT2, which generates Gln deprivation to direct metabolic reprogramming of cancer cells and enhances cellular uptake of Gln-modified IDS via RAS-stimulated macropinocytosis. The enhanced endocytosis and high iron content of IDS facilitate ferroptosis in mice pancreatic tumor models; thus, an amino acid-like ferroptosis inducer of l-buthionine sulfoximine (BSO) is further combined. The enhanced endocytosis resulting from the synergism of Gln and V9302 enables the efficient delivery of iron and BSO for ferroptosis tumor therapy. This work provides an alternative approach for enhancing intracellular drug delivery of the tumors with heterogeneous nutrient metabolism by virtue of combining nutrient-modified nanodrugs with the corresponding nutrient transporter inhibitors.
Collapse
Affiliation(s)
- Yang Chen
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Wenzhe Xu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Hao Jin
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Mengsi Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Shuwei Liu
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130021, P. R. China
| | - Yi Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
| | - Hao Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P. R. China
- Joint Laboratory of Opto-Functional Theranostics in Medicine and Chemistry, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun 130021, P. R. China
| |
Collapse
|
33
|
Smiles WJ, Ovens AJ, Kemp BE, Galic S, Petersen J, Oakhill JS. New developments in AMPK and mTORC1 cross-talk. Essays Biochem 2024; 68:321-336. [PMID: 38994736 PMCID: PMC12055038 DOI: 10.1042/ebc20240007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/13/2024]
Abstract
Metabolic homeostasis and the ability to link energy supply to demand are essential requirements for all living cells to grow and proliferate. Key to metabolic homeostasis in all eukaryotes are AMPK and mTORC1, two kinases that sense nutrient levels and function as counteracting regulators of catabolism (AMPK) and anabolism (mTORC1) to control cell survival, growth and proliferation. Discoveries beginning in the early 2000s revealed that AMPK and mTORC1 communicate, or cross-talk, through direct and indirect phosphorylation events to regulate the activities of each other and their shared protein substrate ULK1, the master initiator of autophagy, thereby allowing cellular metabolism to rapidly adapt to energy and nutritional state. More recent reports describe divergent mechanisms of AMPK/mTORC1 cross-talk and the elaborate means by which AMPK and mTORC1 are activated at the lysosome. Here, we provide a comprehensive overview of current understanding in this exciting area and comment on new evidence showing mTORC1 feedback extends to the level of the AMPK isoform, which is particularly pertinent for some cancers where specific AMPK isoforms are implicated in disease pathogenesis.
Collapse
Affiliation(s)
- William J Smiles
- Metabolic Signalling Laboratory, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Research Program for Receptor Biochemistry and Tumour Metabolism, Department of Paediatrics, University Hospital of the Paracelsus Medical University, Salzburg, Austria
| | - Ashley J Ovens
- Protein Engineering in Immunity and Metabolism, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Bruce E Kemp
- Protein Chemistry and Metabolism, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Department of Medicine, University of Melbourne, Parkville, VIC 3010, Australia
- Mary Mackillop Institute for Health Research, Australian Catholic University, Fitzroy, Vic 3065, Vic. Australia
| | - Sandra Galic
- Department of Medicine, University of Melbourne, Parkville, VIC 3010, Australia
- Metabolic Physiology, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Janni Petersen
- Flinders Health and Medical Research Institute, Flinders Centre for Innovation in Cancer, Flinders University, Adelaide, SA 5042, Australia
- Nutrition and Metabolism, South Australia Health and Medical Research Institute, Adelaide, SA 5000, Australia
| | - Jonathan S Oakhill
- Metabolic Signalling Laboratory, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
- Department of Medicine, University of Melbourne, Parkville, VIC 3010, Australia
| |
Collapse
|
34
|
Gao J, Lin M, Qing J, Li H, Zeng X, Yuan W, Li T, He S. HSP70 promotes amino acid-dependent mTORC1 signaling by mediating CHIP-induced NPRL2 ubiquitination and degradation. FASEB J 2024; 38:e70147. [PMID: 39495541 DOI: 10.1096/fj.202401352r] [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: 06/15/2024] [Revised: 09/21/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024]
Abstract
Mechanistic target of rapamycin complex 1 (mTORC1) is a master regulator of cell growth and its dysregulation leads to a variety of human diseases. Although NPRL2, an essential component of the GATOR1 complex, is reported to effectively suppress amino acid-induced mTORC1 activation, the regulation of NPRL2 protein stability is unclear. In this study, we show that chaperon-associated ubiquitin ligase CHIP interacts with NPRL2 and promotes its polyubiquitination and proteasomal degradation. Moreover, HSP70 mediates CHIP-induced ubiquitination and degradation of NPRL2. Consistently, overexpression of HSP70 enhances whereas HSP70 depletion inhibits amino acid-induced mTORC1 activation. Accordingly, knockdown of HSP70 promotes basal autophagic flux, and inhibits cell growth and proliferation. Taken together, these results demonstrated that HSP70 is a novel activator of mTORC1 through mediating CHIP-induced ubiquitination and degradation of NPRL2.
Collapse
Affiliation(s)
- Jianfang Gao
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, School of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Mingjun Lin
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, School of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Jina Qing
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, School of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Hongxia Li
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, School of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Xiao Zeng
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, School of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Wuzhou Yuan
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, School of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Tingting Li
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, School of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| | - Shanping He
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, School of Life Sciences, Hunan Normal University, Changsha, Hunan, China
| |
Collapse
|
35
|
Chen R, Yang C, Yang F, Yang A, Xiao H, Peng B, Chen C, Geng B, Xia Y. Targeting the mTOR-Autophagy Axis: Unveiling Therapeutic Potentials in Osteoporosis. Biomolecules 2024; 14:1452. [PMID: 39595628 PMCID: PMC11591800 DOI: 10.3390/biom14111452] [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: 10/12/2024] [Revised: 11/02/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Osteoporosis (OP) is a widespread age-related disorder marked by decreased bone density and increased fracture risk, presenting a significant public health challenge. Central to the development and progression of OP is the dysregulation of the mechanistic target of the rapamycin (mTOR)-signaling pathway, which plays a critical role in cellular processes including autophagy, growth, and proliferation. The mTOR-autophagy axis is emerging as a promising therapeutic target due to its regulatory capacity in bone metabolism and homeostasis. This review aims to (1) elucidate the role of mTOR signaling in bone metabolism and its dysregulation in OP, (2) explore the interplay between mTOR and autophagy in the context of bone cell activity, and (3) assess the therapeutic potential of targeting the mTOR pathway with modulators as innovative strategies for OP treatment. By examining the interactions among autophagy, mTOR, and OP, including insights from various types of OP and the impact on different bone cells, this review underscores the complexity of mTOR's role in bone health. Despite advances, significant gaps remain in understanding the detailed mechanisms of mTOR's effects on autophagy and bone cell function, highlighting the need for comprehensive clinical trials to establish the efficacy and safety of mTOR inhibitors in OP management. Future research directions include clarifying mTOR's molecular interactions with bone metabolism and investigating the combined benefits of mTOR modulation with other therapeutic approaches. Addressing these challenges is crucial for developing more effective treatments and improving outcomes for individuals with OP, thereby unveiling the therapeutic potentials of targeting the mTOR-autophagy axis in this prevalent disease.
Collapse
Affiliation(s)
- Rongjin Chen
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou 730030, China; (R.C.); (C.Y.); (F.Y.); (A.Y.); (H.X.); (B.P.); (C.C.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
- Department of Orthopedics, Tianshui Hand and Foot Surgery Hospital, Tianshui 741000, China
| | - Chenhui Yang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou 730030, China; (R.C.); (C.Y.); (F.Y.); (A.Y.); (H.X.); (B.P.); (C.C.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
- Department of Orthopedics, Tianshui Hand and Foot Surgery Hospital, Tianshui 741000, China
| | - Fei Yang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou 730030, China; (R.C.); (C.Y.); (F.Y.); (A.Y.); (H.X.); (B.P.); (C.C.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Ao Yang
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou 730030, China; (R.C.); (C.Y.); (F.Y.); (A.Y.); (H.X.); (B.P.); (C.C.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Hefang Xiao
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou 730030, China; (R.C.); (C.Y.); (F.Y.); (A.Y.); (H.X.); (B.P.); (C.C.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Bo Peng
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou 730030, China; (R.C.); (C.Y.); (F.Y.); (A.Y.); (H.X.); (B.P.); (C.C.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Changshun Chen
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou 730030, China; (R.C.); (C.Y.); (F.Y.); (A.Y.); (H.X.); (B.P.); (C.C.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Bin Geng
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou 730030, China; (R.C.); (C.Y.); (F.Y.); (A.Y.); (H.X.); (B.P.); (C.C.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Yayi Xia
- Department of Orthopedics, The Second Hospital of Lanzhou University, Lanzhou 730030, China; (R.C.); (C.Y.); (F.Y.); (A.Y.); (H.X.); (B.P.); (C.C.); (B.G.)
- Orthopedic Clinical Medical Research Center and Intelligent Orthopedic Industry Technology Center of Gansu Province, Lanzhou 730030, China
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| |
Collapse
|
36
|
Jiang C, Tan X, Liu N, Yan P, Hou T, Wei W. Nutrient sensing of mTORC1 signaling in cancer and aging. Semin Cancer Biol 2024; 106-107:1-12. [PMID: 39153724 DOI: 10.1016/j.semcancer.2024.08.001] [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: 06/27/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 08/19/2024]
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) is indispensable for preserving cellular and organismal homeostasis by balancing the anabolic and catabolic processes in response to various environmental cues, such as nutrients, growth factors, energy status, oxygen levels, and stress. Dysregulation of mTORC1 signaling is associated with the progression of many types of human disorders including cancer, age-related diseases, neurodegenerative disorders, and metabolic diseases. The way mTORC1 senses various upstream signals and converts them into specific downstream responses remains a crucial question with significant impacts for our perception of the related physiological and pathological process. In this review, we discuss the recent molecular and functional insights into the nutrient sensing of the mTORC1 signaling pathway, along with the emerging role of deregulating nutrient-mTORC1 signaling in cancer and age-related disorders.
Collapse
Affiliation(s)
- Cong Jiang
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China.
| | - Xiao Tan
- Tongji University Cancer Center, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Ning Liu
- International Research Center for Food and Health, College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, China
| | - Peiqiang Yan
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Tao Hou
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Wenyi Wei
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
37
|
Song Y, Wang Y, Zhang H, Saddique MAB, Luo X, Ren M. The TOR signalling pathway in fungal phytopathogens: A target for plant disease control. MOLECULAR PLANT PATHOLOGY 2024; 25:e70024. [PMID: 39508186 PMCID: PMC11541241 DOI: 10.1111/mpp.70024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/10/2024] [Accepted: 10/18/2024] [Indexed: 11/08/2024]
Abstract
Plant diseases caused by fungal phytopathogens have led to significant economic losses in agriculture worldwide. The management of fungal diseases is mainly dependent on the application of fungicides, which are not suitable for sustainable agriculture, human health, and environmental safety. Thus, it is necessary to develop novel targets and green strategies to mitigate the losses caused by these pathogens. The target of rapamycin (TOR) complexes and key components of the TOR signalling pathway are evolutionally conserved in pathogens and closely related to the vegetative growth and pathogenicity. As indicated in recent systems, chemical, genetic, and genomic studies on the TOR signalling pathway, phytopathogens with TOR dysfunctions show severe growth defects and nonpathogenicity, which makes the TOR signalling pathway to be developed into an ideal candidate target for controlling plant disease. In this review, we comprehensively discuss the current knowledge on components of the TOR signalling pathway in microorganisms and the diverse roles of various plant TOR in response to plant pathogens. Furthermore, we analyse a range of disease management strategies that rely on the TOR signalling pathway, including genetic modification technologies and chemical controls. In the future, disease control strategies based on the TOR signalling network are expected to become a highly effective weapon for crop protection.
Collapse
Affiliation(s)
- Yun Song
- College of Agriculture and BiologyLiaocheng UniversityLiaochengChina
| | - Yaru Wang
- College of Agriculture and BiologyLiaocheng UniversityLiaochengChina
| | - Huafang Zhang
- Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences; Chengdu Agricultural Science and Technology CenterChengduChina
| | - Muhammad Abu Bakar Saddique
- Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences; Chengdu Agricultural Science and Technology CenterChengduChina
| | - Xiumei Luo
- Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences; Chengdu Agricultural Science and Technology CenterChengduChina
| | - Maozhi Ren
- Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences; Chengdu Agricultural Science and Technology CenterChengduChina
| |
Collapse
|
38
|
Fernandes SA, Angelidaki DD, Nüchel J, Pan J, Gollwitzer P, Elkis Y, Artoni F, Wilhelm S, Kovacevic-Sarmiento M, Demetriades C. Spatial and functional separation of mTORC1 signalling in response to different amino acid sources. Nat Cell Biol 2024; 26:1918-1933. [PMID: 39385049 PMCID: PMC11567901 DOI: 10.1038/s41556-024-01523-7] [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: 02/01/2024] [Accepted: 09/09/2024] [Indexed: 10/11/2024]
Abstract
Amino acid (AA) availability is a robust determinant of cell growth through controlling mechanistic/mammalian target of rapamycin complex 1 (mTORC1) activity. According to the predominant model in the field, AA sufficiency drives the recruitment and activation of mTORC1 on the lysosomal surface by the heterodimeric Rag GTPases, from where it coordinates the majority of cellular processes. Importantly, however, the teleonomy of the proposed lysosomal regulation of mTORC1 and where mTORC1 acts on its effector proteins remain enigmatic. Here, by using multiple pharmacological and genetic means to perturb the lysosomal AA-sensing and protein recycling machineries, we describe the spatial separation of mTORC1 regulation and downstream functions in mammalian cells, with lysosomal and non-lysosomal mTORC1 phosphorylating distinct substrates in response to different AA sources. Moreover, we reveal that a fraction of mTOR localizes at lysosomes owing to basal lysosomal proteolysis that locally supplies new AAs, even in cells grown in the presence of extracellular nutrients, whereas cytoplasmic mTORC1 is regulated by exogenous AAs. Overall, our study substantially expands our knowledge about the topology of mTORC1 regulation by AAs and hints at the existence of distinct, Rag- and lysosome-independent mechanisms that control its activity at other subcellular locations. Given the importance of mTORC1 signalling and AA sensing for human ageing and disease, our findings will probably pave the way towards the identification of function-specific mTORC1 regulators and thus highlight more effective targets for drug discovery against conditions with dysregulated mTORC1 activity in the future.
Collapse
Affiliation(s)
- Stephanie A Fernandes
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Cologne Graduate School of Ageing Research, Cologne, Germany
| | | | - Julian Nüchel
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Jiyoung Pan
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Cologne Graduate School of Ageing Research, Cologne, Germany
| | | | - Yoav Elkis
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Filippo Artoni
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Cologne Graduate School of Ageing Research, Cologne, Germany
| | - Sabine Wilhelm
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | | | - Constantinos Demetriades
- Max Planck Institute for Biology of Ageing, Cologne, Germany.
- Cologne Graduate School of Ageing Research, Cologne, Germany.
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany.
| |
Collapse
|
39
|
Bayly-Jones C, Lupton CJ, Keen AC, Dong S, Mastos C, Luo W, Qian C, Jones GD, Venugopal H, Chang YG, Clarke RJ, Halls ML, Ellisdon AM. LYCHOS is a human hybrid of a plant-like PIN transporter and a GPCR. Nature 2024; 634:1238-1244. [PMID: 39358511 PMCID: PMC11525196 DOI: 10.1038/s41586-024-08012-9] [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: 09/18/2023] [Accepted: 09/04/2024] [Indexed: 10/04/2024]
Abstract
Lysosomes have crucial roles in regulating eukaryotic metabolism and cell growth by acting as signalling platforms to sense and respond to changes in nutrient and energy availability1. LYCHOS (GPR155) is a lysosomal transmembrane protein that functions as a cholesterol sensor, facilitating the cholesterol-dependent activation of the master protein kinase mechanistic target of rapamycin complex 1 (mTORC1)2. However, the structural basis of LYCHOS assembly and activity remains unclear. Here we determine several high-resolution cryo-electron microscopy structures of human LYCHOS, revealing a homodimeric transmembrane assembly of a transporter-like domain fused to a G-protein-coupled receptor (GPCR) domain. The class B2-like GPCR domain is captured in the apo state and packs against the surface of the transporter-like domain, providing an unusual example of a GPCR as a domain in a larger transmembrane assembly. Cholesterol sensing is mediated by a conserved cholesterol-binding motif, positioned between the GPCR and transporter domains. We reveal that the LYCHOS transporter-like domain is an orthologue of the plant PIN-FORMED (PIN) auxin transporter family, and has greater structural similarity to plant auxin transporters than to known human transporters. Activity assays support a model in which the LYCHOS transporter and GPCR domains coordinate to sense cholesterol and regulate mTORC1 activation.
Collapse
Affiliation(s)
- Charles Bayly-Jones
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- School of Chemistry, University of Sydney, Camperdown, New South Wales, Australia
| | - Christopher J Lupton
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Alastair C Keen
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Shuqi Dong
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Chantel Mastos
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Wentong Luo
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Chunyi Qian
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Gareth D Jones
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Hari Venugopal
- Ramaciotti Centre for Cryo-Electron Microscopy, Monash University, Clayton, Victoria, Australia
| | - Yong-Gang Chang
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Ronald J Clarke
- School of Chemistry, University of Sydney, Camperdown, New South Wales, Australia
- University of Sydney Nano Institute, Camperdown, New South Wales, Australia
| | - Michelle L Halls
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia.
| | - Andrew M Ellisdon
- Cancer Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
40
|
Swaroop V, Ozkan E, Herrmann L, Thurman A, Kopasz-Gemmen O, Kunamneni A, Inoki K. mTORC1 signaling and diabetic kidney disease. Diabetol Int 2024; 15:707-718. [PMID: 39469564 PMCID: PMC11512951 DOI: 10.1007/s13340-024-00738-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/26/2024] [Indexed: 10/30/2024]
Abstract
Diabetic kidney disease (DKD) represents the most lethal complication in both type 1 and type 2 diabetes. The disease progresses without obvious symptoms and is often refractory when apparent symptoms have emerged. Although the molecular mechanisms underlying the onset/progression of DKD have been extensively studied, only a few effective therapies are currently available. Pathogenesis of DKD involves multifaced events caused by diabetes, which include alterations of metabolisms, signals, and hemodynamics. While the considerable efficacy of sodium/glucose cotransporter-2 (SGLT2) inhibitors or angiotensin II receptor blockers (ARBs) for DKD has been recognized, the ever-increasing number of patients with diabetes and DKD warrants additional practical therapeutic approaches that prevent DKD from diabetes. One plausible but promising target is the mechanistic target of the rapamycin complex 1 (mTORC1) signaling pathway, which senses cellular nutrients to control various anabolic and catabolic processes. This review introduces the current understanding of the mTOR signaling pathway and its roles in the development of DKD and other chronic kidney diseases (CKDs), and discusses potential therapeutic approaches targeting this pathway for the future treatment of DKD.
Collapse
Affiliation(s)
- Vinamra Swaroop
- Life Sciences Institute, University of Michigan, Ann Arbor, USA
| | - Eden Ozkan
- Life Sciences Institute, University of Michigan, Ann Arbor, USA
| | - Lydia Herrmann
- Life Sciences Institute, University of Michigan, Ann Arbor, USA
| | - Aaron Thurman
- Life Sciences Institute, University of Michigan, Ann Arbor, USA
| | | | | | - Ken Inoki
- Life Sciences Institute, University of Michigan, Ann Arbor, USA
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, USA
- Department of Internal Medicine, Division of Nephrology, University of Michigan Medical School, Ann Arbor, USA
| |
Collapse
|
41
|
Feng Z, Cao K, Sun H, Liu X. SEH1L siliencing induces ferroptosis and suppresses hepatocellular carcinoma progression via ATF3/HMOX1/GPX4 axis. Apoptosis 2024; 29:1723-1737. [PMID: 39095556 PMCID: PMC11416379 DOI: 10.1007/s10495-024-02009-5] [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] [Accepted: 07/17/2024] [Indexed: 08/04/2024]
Abstract
SEH1 like nucleoporin (SEH1L) is an important component of nuclear pore complex (NPC), which is crucial in the regulation of cell division. However, the interrelation between SEH1L expression and tumor progression is less studied. In this research, we performed a systematic bioinformatic analysis about SEH1L using TCGA, Timer 2.0, Cbioportal, UCLAN and CellMiner™ databases in pan-cancer. Besides, we further validated the bioinformatic results through in vitro and in vivo experiments in HCC, including transcriptome sequencing, real-time quantitative PCR (RT-qPCR), western blotting (WB), immunohistochemistry (IHC), cell proliferation assays, clone formation, EdU, transwell, flow cytometry and subcutaneous tumor model. Our results suggested that SEH1L was significantly up-regulated and related to poor prognosis in most cancers, and may serve as a potential biomarker. SEH1L could promote HCC progression in vitro and in vivo. Besides, the next generation sequencing suggested that 684 genes was significantly up-regulated and 678 genes was down-regulated after the knock down of SEH1L. SEH1L siliencing could activate ATF3/HMOX1/GPX4 axis, decrease mitochondrial membrane potential and GSH, but increase ROS and MDA, and these effects could be reversed by the knock down of ATF3. This study indicated that SEH1L siliencing could induce ferroptosis and suppresses hepatocellular carcinoma (HCC) progression via ATF3/HMOX1/GPX4 axis.
Collapse
Affiliation(s)
- Ziyang Feng
- Postdoctoral Station of Medical Aspects of Specific Environments, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, P.R. China
| | - Ke Cao
- Department of Oncology, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, P.R. China
| | - Haojia Sun
- Department of Oncology, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, P.R. China
| | - Xuewen Liu
- Department of Oncology, The Third Xiangya Hospital, Central South University, Changsha, 410013, Hunan, P.R. China.
| |
Collapse
|
42
|
Zhang Q, Chen C, Ma Y, Yan X, Lai N, Wang H, Gao B, Gu AM, Han Q, Zhang Q, La L, Sun X. PGAM5 interacts with and maintains BNIP3 to license cancer-associated muscle wasting. Autophagy 2024; 20:2205-2220. [PMID: 38919131 PMCID: PMC11423673 DOI: 10.1080/15548627.2024.2360340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 05/14/2024] [Accepted: 05/22/2024] [Indexed: 06/27/2024] Open
Abstract
Regressing the accelerated degradation of skeletal muscle protein is a significant goal for cancer cachexia management. Here, we show that genetic deletion of Pgam5 ameliorates skeletal muscle atrophy in various tumor-bearing mice. pgam5 ablation represses excessive myoblast mitophagy and effectively suppresses mitochondria meltdown and muscle wastage. Next, we define BNIP3 as a mitophagy receptor constitutively associating with PGAM5. bnip3 deletion restricts body weight loss and enhances the gastrocnemius mass index in the age- and tumor size-matched experiments. The NH2-terminal region of PGAM5 binds to the PEST motif-containing region of BNIP3 to dampen the ubiquitination and degradation of BNIP3 to maintain continuous mitophagy. Finally, we identify S100A9 as a pro-cachectic chemokine via activating AGER/RAGE. AGER deficiency or S100A9 inhibition restrains skeletal muscle loss by weakening the interaction between PGAM5 and BNIP3. In conclusion, the AGER-PGAM5-BNIP3 axis is a novel but common pathway in cancer-associated muscle wasting that can be targetable. Abbreviation: AGER/RAGE: advanced glycation end-product specific receptor; BA1: bafilomycin A1; BNIP3: BCL2 interacting protein 3; BNIP3L: BCL2 interacting protein 3 like; Ckm-Cre: creatinine kinase, muscle-specific Cre; CM: conditioned medium; CON/CTRL: control; CRC: colorectal cancer; FUNDC1: FUN14 domain containing 1; MAP1LC3A/LC3A: microtubule associated protein 1 light chain 3 alpha; PGAM5: PGAM family member 5, mitochondrial serine/threonine protein phosphatase; S100A9: S100 calcium binding protein A9; SQSTM1/p62: sequestosome 1; TOMM20: translocase of outer mitochondrial membrane 20; TIMM23: translocase of inner mitochondrial membrane 23; TSKO: tissue-specific knockout; VDAC1: voltage dependent anion channel 1.
Collapse
Affiliation(s)
- Qingyuan Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Chunhui Chen
- Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ye Ma
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Xinyi Yan
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Nianhong Lai
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Hao Wang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Baogui Gao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Anna Meilin Gu
- Department of Biology, University of Washington, Seattle, WA, USA
| | - Qinrui Han
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Qingling Zhang
- Department of Pathology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Lei La
- Department of Pharmacy, Clinical Pharmacy Center, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xuegang Sun
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
43
|
Xavier A, Dikic I. Feeding cancer to death - a triad of aromatic acids reduces tumor growth. Cell Death Differ 2024; 31:1239-1241. [PMID: 39266718 PMCID: PMC11445509 DOI: 10.1038/s41418-024-01372-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/05/2024] [Accepted: 08/27/2024] [Indexed: 09/14/2024] Open
Affiliation(s)
- Audrey Xavier
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt am Main, Germany
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Ivan Dikic
- Institute of Biochemistry II, Faculty of Medicine, Goethe University Frankfurt, Frankfurt am Main, Germany.
- Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Frankfurt am Main, Germany.
- Max Planck Institute of Biophysics, Frankfurt am Main, Germany.
| |
Collapse
|
44
|
Cormerais Y, Lapp SC, Kalafut KC, Cissé MY, Shin J, Stefadu B, Personnaz J, Schrotter S, D’Amore A, Martin ER, Salussolia CL, Sahin M, Menon S, Byles V, Manning BD. AKT-mediated phosphorylation of TSC2 controls stimulus- and tissue-specific mTORC1 signaling and organ growth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.23.614519. [PMID: 39386441 PMCID: PMC11463511 DOI: 10.1101/2024.09.23.614519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Mechanistic target of rapamycin (mTOR) complex 1 (mTORC1) integrates diverse intracellular and extracellular growth signals to regulate cell and tissue growth. How the molecular mechanisms regulating mTORC1 signaling established through biochemical and cell biological studies function under physiological states in specific mammalian tissues are unknown. Here, we characterize a genetic mouse model lacking the 5 phosphorylation sites on the tuberous sclerosis complex 2 (TSC2) protein through which the growth factor-stimulated protein kinase AKT can active mTORC1 signaling in cell culture models. These phospho-mutant mice (TSC2-5A) are developmentally normal but exhibit reduced body weight and the weight of specific organs, such as brain and skeletal muscle, associated with cell intrinsic decreases in growth factor-stimulated mTORC1 signaling. The TSC2-5A mouse model demonstrates that TSC2 phosphorylation is a primary mechanism of mTORC1 activation in some, but not all, tissues and provides a genetic tool to facilitate studies on the physiological regulation of mTORC1.
Collapse
Affiliation(s)
- Yann Cormerais
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Samuel C. Lapp
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
- These authors contributed equally
| | - Krystle C. Kalafut
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
- These authors contributed equally
| | - Madi Y. Cissé
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
- These authors contributed equally
| | - Jong Shin
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
- These authors contributed equally
| | - Benjamin Stefadu
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Jean Personnaz
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Present address: IRSD, Université de Toulouse, INSERM, INRAE, ENVT, Univ Toulouse III - Paul Sabatier (UPS), Toulouse, France
| | - Sandra Schrotter
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Present address: Cell Signaling Technologies, Inc, Beverly, MA, 01915, USA
| | - Angelica D’Amore
- Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Emma R. Martin
- Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Catherine L. Salussolia
- Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Mustafa Sahin
- Kirby Neurobiology Center, Department of Neurology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Suchithra Menon
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Present address: Novartis Institutes for BioMedical Research, Cambridge, MA, 02139, USA
| | - Vanessa Byles
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Brendan D. Manning
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| |
Collapse
|
45
|
Chen S, Abou-Khalil BW, Afawi Z, Ali QZ, Amadori E, Anderson A, Anderson J, Andrade DM, Annesi G, Arslan M, Auce P, Bahlo M, Baker MD, Balagura G, Balestrini S, Banks E, Barba C, Barboza K, Bartolomei F, Bass N, Baum LW, Baumgartner TH, Baykan B, Bebek N, Becker F, Bennett CA, Beydoun A, Bianchini C, Bisulli F, Blackwood D, Blatt I, Borggräfe I, Bosselmann C, Braatz V, Brand H, Brockmann K, Buono RJ, Busch RM, Caglayan SH, Canafoglia L, Canavati C, Castellotti B, Cavalleri GL, Cerrato F, Chassoux F, Cherian C, Cherny SS, Cheung CL, Chou IJ, Chung SK, Churchhouse C, Ciullo V, Clark PO, Cole AJ, Cosico M, Cossette P, Cotsapas C, Cusick C, Daly MJ, Davis LK, Jonghe PD, Delanty N, Dennig D, Depondt C, Derambure P, Devinsky O, Di Vito L, Dickerson F, Dlugos DJ, Doccini V, Doherty CP, El-Naggar H, Ellis CA, Epstein L, Evans M, Faucon A, Feng YCA, Ferguson L, Ferraro TN, Da Silva IF, Ferri L, Feucht M, Fields MC, Fitzgerald M, Fonferko-Shadrach B, Fortunato F, Franceschetti S, French JA, Freri E, Fu JM, Gabriel S, Gagliardi M, Gambardella A, Gauthier L, Giangregorio T, Gili T, Glauser TA, Goldberg E, Goldman A, Goldstein DB, et alChen S, Abou-Khalil BW, Afawi Z, Ali QZ, Amadori E, Anderson A, Anderson J, Andrade DM, Annesi G, Arslan M, Auce P, Bahlo M, Baker MD, Balagura G, Balestrini S, Banks E, Barba C, Barboza K, Bartolomei F, Bass N, Baum LW, Baumgartner TH, Baykan B, Bebek N, Becker F, Bennett CA, Beydoun A, Bianchini C, Bisulli F, Blackwood D, Blatt I, Borggräfe I, Bosselmann C, Braatz V, Brand H, Brockmann K, Buono RJ, Busch RM, Caglayan SH, Canafoglia L, Canavati C, Castellotti B, Cavalleri GL, Cerrato F, Chassoux F, Cherian C, Cherny SS, Cheung CL, Chou IJ, Chung SK, Churchhouse C, Ciullo V, Clark PO, Cole AJ, Cosico M, Cossette P, Cotsapas C, Cusick C, Daly MJ, Davis LK, Jonghe PD, Delanty N, Dennig D, Depondt C, Derambure P, Devinsky O, Di Vito L, Dickerson F, Dlugos DJ, Doccini V, Doherty CP, El-Naggar H, Ellis CA, Epstein L, Evans M, Faucon A, Feng YCA, Ferguson L, Ferraro TN, Da Silva IF, Ferri L, Feucht M, Fields MC, Fitzgerald M, Fonferko-Shadrach B, Fortunato F, Franceschetti S, French JA, Freri E, Fu JM, Gabriel S, Gagliardi M, Gambardella A, Gauthier L, Giangregorio T, Gili T, Glauser TA, Goldberg E, Goldman A, Goldstein DB, Granata T, Grant R, Greenberg DA, Guerrini R, Gundogdu-Eken A, Gupta N, Haas K, Hakonarson H, Haryanyan G, Häusler M, Hegde M, Heinzen EL, Helbig I, Hengsbach C, Heyne H, Hirose S, Hirsch E, Ho CJ, Hoeper O, Howrigan DP, Hucks D, Hung PC, Iacomino M, Inoue Y, Inuzuka LM, Ishii A, Jehi L, Johnson MR, Johnstone M, Kälviäinen R, Kanaan M, Kara B, Kariuki SM, Kegele J, Kesim Y, Khoueiry-Zgheib N, Khoury J, King C, Klein KM, Kluger G, Knake S, Kok F, Korczyn AD, Korinthenberg R, Koupparis A, Kousiappa I, Krause R, Krenn M, Krestel H, Krey I, Kunz WS, Kurlemann G, Kuzniecky RI, Kwan P, La Vega-Talbott M, Labate A, Lacey A, Lal D, Laššuthová P, Lauxmann S, Lawthom C, Leech SL, Lehesjoki AE, Lemke JR, Lerche H, Lesca G, Leu C, Lewin N, Lewis-Smith D, Li GHY, Liao C, Licchetta L, Lin CH, Lin KL, Linnankivi T, Lo W, Lowenstein DH, Lowther C, Lubbers L, Lui CHT, Macedo-Souza LI, Madeleyn R, Madia F, Magri S, Maillard L, Marcuse L, Marques P, Marson AG, Matthews AG, May P, Mayer T, McArdle W, McCarroll SM, McGoldrick P, McGraw CM, McIntosh A, McQuillan A, Meador KJ, Mei D, Michel V, Millichap JJ, Minardi R, Montomoli M, Mostacci B, Muccioli L, Muhle H, Müller-Schlüter K, Najm IM, Nasreddine W, Neaves S, Neubauer BA, Newton CRJC, Noebels JL, Northstone K, Novod S, O'Brien TJ, Owusu-Agyei S, Özkara Ç, Palotie A, Papacostas SS, Parrini E, Pato C, Pato M, Pendziwiat M, Pennell PB, Petrovski S, Pickrell WO, Pinsky R, Pinto D, Pippucci T, Piras F, Piras F, Poduri A, Pondrelli F, Posthuma D, Powell RHW, Privitera M, Rademacher A, Ragona F, Ramirez-Hamouz B, Rau S, Raynes HR, Rees MI, Regan BM, Reif A, Reinthaler E, Rheims S, Ring SM, Riva A, Rojas E, Rosenow F, Ryvlin P, Saarela A, Sadleir LG, Salman B, Salmon A, Salpietro V, Sammarra I, Scala M, Schachter S, Schaller A, Schankin CJ, Scheffer IE, Schneider N, Schubert-Bast S, Schulze-Bonhage A, Scudieri P, Sedláčková L, Shain C, Sham PC, Shiedley BR, Siena SA, Sills GJ, Sisodiya SM, Smoller JW, Solomonson M, Spalletta G, Sparks KR, Sperling MR, Stamberger H, Steinhoff BJ, Stephani U, Štěrbová K, Stewart WC, Stipa C, Striano P, Strzelczyk A, Surges R, Suzuki T, Talarico M, Talkowski ME, Taneja RS, Tanteles GA, Timonen O, Timpson NJ, Tinuper P, Todaro M, Topaloglu P, Tsai MH, Tumiene B, Turkdogan D, Uğur-İşeri S, Utkus A, Vaidiswaran P, Valton L, van Baalen A, Vari MS, Vetro A, Vlčková M, von Brauchitsch S, von Spiczak S, Wagner RG, Watts N, Weber YG, Weckhuysen S, Widdess-Walsh P, Wiebe S, Wolf SM, Wolff M, Wolking S, Wong I, von Wrede R, Wu D, Yamakawa K, Yapıcı Z, Yis U, Yolken R, Yücesan E, Zagaglia S, Zahnert F, Zara F, Zimprich F, Zizovic M, Zsurka G, Neale BM, Berkovic SF. Exome sequencing of 20,979 individuals with epilepsy reveals shared and distinct ultra-rare genetic risk across disorder subtypes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.02.22.23286310. [PMID: 36865150 PMCID: PMC9980234 DOI: 10.1101/2023.02.22.23286310] [Show More Authors] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Identifying genetic risk factors for highly heterogeneous disorders like epilepsy remains challenging. Here, we present the largest whole-exome sequencing study of epilepsy to date, with >54,000 human exomes, comprising 20,979 deeply phenotyped patients from multiple genetic ancestry groups with diverse epilepsy subtypes and 33,444 controls, to investigate rare variants that confer disease risk. These analyses implicate seven individual genes, three gene sets, and four copy number variants at exome-wide significance. Genes encoding ion channels show strong association with multiple epilepsy subtypes, including epileptic encephalopathies, generalized and focal epilepsies, while most other gene discoveries are subtype-specific, highlighting distinct genetic contributions to different epilepsies. Combining results from rare single nucleotide/short indel-, copy number-, and common variants, we offer an expanded view of the genetic architecture of epilepsy, with growing evidence of convergence among different genetic risk loci on the same genes. Top candidate genes are enriched for roles in synaptic transmission and neuronal excitability, particularly postnatally and in the neocortex. We also identify shared rare variant risk between epilepsy and other neurodevelopmental disorders. Our data can be accessed via an interactive browser, hopefully facilitating diagnostic efforts and accelerating the development of follow-up studies.
Collapse
Affiliation(s)
- Siwei Chen
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Bassel W Abou-Khalil
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Zaid Afawi
- Tel-Aviv University Sackler Faculty of Medicine, Ramat Aviv 69978, Israel
| | | | | | - Alison Anderson
- Department of Medicine, University of Melbourne, Royal Melbourne Hospital, Parkville 3050, Australia
- Department of Neuroscience, The School of Translational Medicine, Alfred Health, Monash University, Melbourne 3004, Australia
| | - Joe Anderson
- Neurology Department, Aneurin Bevan University Health Board, Newport, Wales, UK
| | | | - Grazia Annesi
- Department of Medical and Surgical Sciences, Neuroscience Research Center, Magna Graecia University, Catanzaro, Italy
| | - Mutluay Arslan
- Department of Child Neurology, Gülhane Education and Research Hospital, Health Sciences University, Ankara, Turkey
| | - Pauls Auce
- St George's University Hospital NHS Foundation Trust, London, UK
| | - Melanie Bahlo
- Population Health and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville 3052, Australia
- Department of Biology, University of Melbourne, Parkville 3010, Australia
| | - Mark D Baker
- Swansea University Medical School, Swansea University, Swansea, Wales, UK
| | - Ganna Balagura
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | - Simona Balestrini
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
- Chalfont Centre for Epilepsy, Chalfont-St-Peter, Buckinghamshire SL9 0RJ, UK
| | - Eric Banks
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Carmen Barba
- Neuroscience Department, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Karen Barboza
- University Health Network, University of Toronto, Toronto, ON, Canada
| | - Fabrice Bartolomei
- Clinical Neurophysiology and Epileptology Department, Timone Hospital, Marseille, France
| | - Nick Bass
- Division of Psychiatry, University College London
| | - Larry W Baum
- Department of Psychiatry, The University of Hong Kong, Pokulam, Hong Kong
| | - Tobias H Baumgartner
- Department of Epileptology, University of Bonn Medical Centre, Bonn 53127, Germany
| | - Betül Baykan
- Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Nerses Bebek
- Department of Child Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Felicitas Becker
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen 72076, Germany
- Department of Neurology, University of Ulm, Ulm 89081, Germany
| | - Caitlin A Bennett
- Epilepsy Research Centre, University of Melbourne, Austin Health, Heidelberg 3084, Australia
| | - Ahmad Beydoun
- Department of Neurology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Claudia Bianchini
- Neuroscience Department, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Francesca Bisulli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- IRCCS Istituto delle Scienze Neurologiche di Bologna, (Reference Center for Rare and Complex Epilepsies - EpiCARE), Bologna, Italy
| | - Douglas Blackwood
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Ilan Blatt
- Tel-Aviv University Sackler Faculty of Medicine, Ramat Aviv 69978, Israel
- Department of Neurology, Sheba Medical Center, Ramat Gan, Israel
| | - Ingo Borggräfe
- Department of Pediatric Neurology, Dr von Hauner Children's Hospital, Ludwig Maximilians University, Munchen, Germany
| | - Christian Bosselmann
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen 72076, Germany
| | - Vera Braatz
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
- Chalfont Centre for Epilepsy, Chalfont-St-Peter, Buckinghamshire SL9 0RJ, UK
| | - Harrison Brand
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Knut Brockmann
- Children's Hospital, Dept. of Pediatric Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Russell J Buono
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104, USA
- Department of Biomedical Sciences, Cooper Medical School of Rowan University Camden, NJ 08103, USA
- Department of Neurology, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA
| | - Robyn M Busch
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Cleveland Clinic Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Neurology, Neurological Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - S Hande Caglayan
- Department of Molecular Biology and Genetics, Bogaziçi University, Istanbul, Turkey
| | - Laura Canafoglia
- Integrated Diagnostics for Epilepsy, Fondazione IRCCS Istituto Neurologico C. Besta, Milan, Italy
| | | | - Barbara Castellotti
- Unit of Medical Genetics and Neurogenetics, Department of Diagnostic and Technology, Fondazione IRCCS Istituto Neurologico Carlo Besta Milano, Italy
| | - Gianpiero L Cavalleri
- School of Pharmacy and Biomolecular Sciences, The Royal College of Surgeons in Ireland, Dublin, Ireland
- The FutureNeuro Research Centre, Dublin, Ireland
| | - Felecia Cerrato
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Francine Chassoux
- Epilepsy Unit, Department of Neurosurgery, Centre Hospitalier Sainte-Anne, and University Paris Descartes, Paris, France
| | - Christina Cherian
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Stacey S Cherny
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ching-Lung Cheung
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Pokfulam, Hong Kong
| | - I-Jun Chou
- Department of Pediatric Neurology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Seo-Kyung Chung
- Swansea University Medical School, Swansea University, Swansea, Wales, UK
- Kids Research, Children's Hospital at Westmead Clinical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Brain & Mind Centre, Faculty of Medicine & Health, University of Sydney, Sydney, New South Wales, Australia
| | - Claire Churchhouse
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Valentina Ciullo
- Neuropsychiatry Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Peggy O Clark
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Andrew J Cole
- Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Mahgenn Cosico
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104, USA
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children's Hospital of Philadelphia, Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Patrick Cossette
- Department of Neurosciences, Université de Montréal, Montréal, CA 26758, Canada
| | | | - Caroline Cusick
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Mark J Daly
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Institute for Molecular Medicine Finland, FIMM, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Lea K Davis
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Peter De Jonghe
- Applied & Translational Neurogenomics Group, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Neurology, Antwerp University Hospital, Edegem 2650, Belgium
- Translational Neurosciences, Faculty of Medicine and Health Science, University of Antwerp, Antwerp, Belgium
| | - Norman Delanty
- School of Pharmacy and Biomolecular Sciences, The Royal College of Surgeons in Ireland, Dublin, Ireland
- The FutureNeuro Research Centre, Dublin, Ireland
- Department of Neurology, Beaumont Hospital, Dublin D09 FT51, Ireland
| | | | - Chantal Depondt
- Department of Neurology, CUB Erasme Hospital, Hôpital Universitaire de Bruxelles (H.U.B.), Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Philippe Derambure
- Department of Clinical Neurophysiology, Lille University Medical Center, EA 1046, University of Lille
| | - Orrin Devinsky
- Department of Neurology, New York University/Langone Health, New York NY, USA
| | - Lidia Di Vito
- IRCCS Istituto delle Scienze Neurologiche di Bologna, (Reference Center for Rare and Complex Epilepsies - EpiCARE), Bologna, Italy
| | - Faith Dickerson
- Sheppard Pratt, 6501 North Charles Street, Baltimore, Maryland, USA
| | - Dennis J Dlugos
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Viola Doccini
- Neuroscience Department, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Colin P Doherty
- The FutureNeuro Research Centre, Dublin, Ireland
- Neurology Department, St. James's Hospital, Dublin D03 VX82, Ireland
| | - Hany El-Naggar
- The FutureNeuro Research Centre, Dublin, Ireland
- Department of Neurology, Beaumont Hospital, Dublin D09 FT51, Ireland
| | - Colin A Ellis
- Department of Neurology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104 USA
| | - Leon Epstein
- Division of Neurology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL USA
| | - Meghan Evans
- Department of Paediatrics and Child Health, University of Otago, Wellington, New Zealand
| | - Annika Faucon
- Human Genetics Training Program, Vanderbilt University, Nashville, TN, USA
| | - Yen-Chen Anne Feng
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Psychiatric & Neurodevelopmental Genetics Unit, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Division of Biostatistics, Institute of Epidemiology and Preventive Medicine, College of Public Health, National Taiwan University, Taipei 100, Taiwan
| | - Lisa Ferguson
- Cleveland Clinic Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Thomas N Ferraro
- Department of Biomedical Sciences, Cooper Medical School of Rowan University Camden, NJ 08103, USA
- Department of Pharmacology and Psychiatry, University of Pennsylvania Perlman School of Medicine, Philadelphia, PA 19104, USA
| | - Izabela Ferreira Da Silva
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette L-4362, Luxembourg
| | - Lorenzo Ferri
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- IRCCS Istituto delle Scienze Neurologiche di Bologna, (Reference Center for Rare and Complex Epilepsies - EpiCARE), Bologna, Italy
| | - Martha Feucht
- Department of Pediatrics and Neonatology, Medical University of Vienna, Vienna 1090, Austria
| | - Madeline C Fields
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mark Fitzgerald
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104, USA
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children's Hospital of Philadelphia, Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104, USA
- Department of Neurology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104 USA
| | | | - Francesco Fortunato
- Institute of Neurology, Department of Medical and Surgical Sciences, University "Magna Graecia", Catanzaro, Italy
| | | | - Jacqueline A French
- Department of Neurology, New York University/Langone Health, New York NY, USA
| | - Elena Freri
- Department of Pediatric Neuroscience, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Jack M Fu
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Stacey Gabriel
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Monica Gagliardi
- Department of Medical and Surgical Sciences, Neuroscience Research Center, Magna Graecia University, Catanzaro, Italy
| | - Antonio Gambardella
- Institute of Neurology, Department of Medical and Surgical Sciences, University "Magna Graecia", Catanzaro, Italy
| | - Laura Gauthier
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Tania Giangregorio
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Medical Genetics Unit, Bologna, Italy
| | - Tommaso Gili
- Neuropsychiatry Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
- IMT School for Advanced Studies Lucca, Lucca, Italy
| | - Tracy A Glauser
- Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Ethan Goldberg
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104, USA
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children's Hospital of Philadelphia, Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104, USA
| | | | - David B Goldstein
- Institute for Genomic Medicine, Columbia University Medical Center, New York, NY 10032, USA
| | - Tiziana Granata
- Department of Pediatric Neuroscience, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Riley Grant
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - David A Greenberg
- Department of Pediatrics, Nationwide Children's Hospital, Columbia, Ohio, USA
| | - Renzo Guerrini
- Neuroscience Department, Meyer Children's Hospital IRCCS, Florence, Italy
- Department of NEUROFARBA, University of Florence, Florence, Italy
| | - Aslı Gundogdu-Eken
- Department of Molecular Biology and Genetics, Bogaziçi University, Istanbul, Turkey
| | - Namrata Gupta
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kevin Haas
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hakon Hakonarson
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Garen Haryanyan
- Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Martin Häusler
- Division of Neuropediatrics and Social Pediatrics, Department of Pediatrics, University Hospital, RWTH Aachen, Aachen, Germany
| | - Manu Hegde
- Department of Neurology, University of California, San Francisco, CA 94143, USA
| | - Erin L Heinzen
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Ingo Helbig
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104, USA
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children's Hospital of Philadelphia, Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104, USA
- Department of Neurology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, 19104 USA
- Department of Neuropediatrics, University Medical Center Schleswig-Holstein, Christian-Albrechts-University, Kiel, Germany
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
- Department of Biomedical and Health Informatics (DBHi), Children's Hospital of Philadelphia, Philadelphia, PA, 19104 USA
| | - Christian Hengsbach
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen 72076, Germany
| | - Henrike Heyne
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Hasso Plattner Institute, Digital Engineering Faculty, University of Potsdam, Germany
| | - Shinichi Hirose
- General Medical Research Center, School of Medicine, Fukuoka University, Japan
| | - Edouard Hirsch
- Department of Neurology, University Hospital of Strasbourg, Strasbourg, France
| | - Chen-Jui Ho
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Olivia Hoeper
- Epilepsy Research Centre, University of Melbourne, Austin Health, Heidelberg 3084, Australia
| | - Daniel P Howrigan
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Donald Hucks
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Po-Chen Hung
- Department of Pediatric Neurology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | | | - Yushi Inoue
- National Epilepsy Center, Shizuoka Institute of Epilepsy and Neurological Disorder, Shizuoka, Japan
| | - Luciana Midori Inuzuka
- Epilepsy Clinic, Hospital Sirio-Libanes, Sao Paulo, Brazil
- Department of Neurology, University of Sao Paulo School of Medicine, Brazil
| | - Atsushi Ishii
- Department of Pediatrics, Fukuoka Sanno Hospital, Japan
| | - Lara Jehi
- Cleveland Clinic Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Neurology, Neurological Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Michael R Johnson
- Division of Brain Sciences, Imperial College London, London SW7 2AZ, UK
| | - Mandy Johnstone
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Reetta Kälviäinen
- Kuopio Epilepsy Center, Neurocenter, Kuopio University Hospital, Kuopio 70210, Finland
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio 70210, Finland
| | - Moien Kanaan
- Hereditary Research Lab, Bethlehem University, Bethlehem, Palestine
| | - Bulent Kara
- Department of Child Neurology, Medical School, Kocaeli University, Kocaeli, Turkey
| | - Symon M Kariuki
- Neuroscience Unit, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Department of Public Health, Pwani University, Kilifi, Kenya
- Department of Psychiatry, University of Oxford, Oxford, UK
| | - Josua Kegele
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen 72076, Germany
| | - Yeşim Kesim
- Department of Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Nathalie Khoueiry-Zgheib
- Department of Pharmacology and Toxicology, American University of Beirut Faculty of Medicine, Beirut, Lebanon
| | - Jean Khoury
- Cleveland Clinic Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Neurology, Neurological Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Chontelle King
- Department of Paediatrics and Child Health, University of Otago, Wellington, New Zealand
| | - Karl Martin Klein
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
- Epilepsy Center Frankfurt Rhine-Main, Center of Neurology and Neurosurgery, Goethe University Frankfurt, Frankfurt, Germany
- LOEWE Center for Personalized Translational Epilepsy Research (CePTER), Goethe University Frankfurt, Germany
| | - Gerhard Kluger
- Neuropediatric Clinic and Clinic for Neurorehabilitation, Epilepsy Center for Children and Adolescents, Vogtareuth, Germany
- Research Institute Rehabilitation / Transition, / Palliation, PMU Salzburg, Austria
| | - Susanne Knake
- LOEWE Center for Personalized Translational Epilepsy Research (CePTER), Goethe University Frankfurt, Germany
- Epilepsy Center Hessen-Marburg, Department of Neurology, Philipps University Marburg, Marburg, Germany
| | - Fernando Kok
- Department of Neurology, University of Sao Paulo School of Medicine, Brazil
- Mendelics Genomic Analysis, São Paulo, Brazil
| | - Amos D Korczyn
- Tel-Aviv University Sackler Faculty of Medicine, Ramat Aviv 69978, Israel
| | - Rudolf Korinthenberg
- Department of Neuropediatrics and Muscular Disorders, University Medical Center, University of Freiburg, Freiburg, Germany
| | | | | | - Roland Krause
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette L-4362, Luxembourg
| | - Martin Krenn
- Department of Neurology, Medical University of Vienna, Vienna 1090, Austria
| | - Heinz Krestel
- Yale School of Medicine, New Haven, CT 06510, USA
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern 3010, Switzerland
| | - Ilona Krey
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
| | - Wolfram S Kunz
- Department of Epileptology, University of Bonn Medical Centre, Bonn 53127, Germany
- Institute of Experimental Epileptology and Cognition Research, Medical Faculty, University of Bonn, Bonn, Germany
| | - Gerhard Kurlemann
- Bonifatius Hospital Lingen, Neuropediatrics Wilhelmstrasse 13, 49808 Lingen, Germany
| | - Ruben I Kuzniecky
- Department of Neurology, Hofstra-Northwell Medical School, New York, NY, USA
| | - Patrick Kwan
- Department of Medicine, University of Melbourne, Royal Melbourne Hospital, Parkville 3050, Australia
- Department of Neuroscience, The School of Translational Medicine, Alfred Health, Monash University, Melbourne 3004, Australia
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, Hong Kong, China
| | - Maite La Vega-Talbott
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Angelo Labate
- Neurophysiopatology and Movement Disorders Clinic, University of Messina, Messina, Italy
| | - Austin Lacey
- School of Pharmacy and Biomolecular Sciences, The Royal College of Surgeons in Ireland, Dublin, Ireland
- The FutureNeuro Research Centre, Dublin, Ireland
| | - Dennis Lal
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Cleveland Clinic Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Petra Laššuthová
- Department of Paediatric Neurology, 2nd Faculty of Medicine, Charles University and Motol Hospital, Prague, Czech Republic
| | - Stephan Lauxmann
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen 72076, Germany
| | - Charlotte Lawthom
- Neurology Department, Aneurin Bevan University Health Board, Newport, Wales, UK
- Swansea University Medical School, Swansea University, Swansea, Wales, UK
| | - Stephanie L Leech
- Epilepsy Research Centre, University of Melbourne, Austin Health, Heidelberg 3084, Australia
| | - Anna-Elina Lehesjoki
- Folkhälsan Research Center, Helsinki 00290, Finland
- Medicum, University of Helsinki, Helsinki 00290, Finland
| | - Johannes R Lemke
- Institute of Human Genetics, University of Leipzig Medical Center, Leipzig, Germany
| | - Holger Lerche
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen 72076, Germany
| | - Gaetan Lesca
- Department of Medical Genetics, Hospices Civils de Lyon and University of Lyon, Lyon, France
| | - Costin Leu
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Naomi Lewin
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104, USA
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children's Hospital of Philadelphia, Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - David Lewis-Smith
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104, USA
- Department of Biomedical and Health Informatics (DBHi), Children's Hospital of Philadelphia, Philadelphia, PA, 19104 USA
- Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, UK
- Department of Clinical Neurosciences, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Gloria Hoi-Yee Li
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hung Hum, Hong Kong
| | - Calwing Liao
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Laura Licchetta
- IRCCS Istituto delle Scienze Neurologiche di Bologna, (Reference Center for Rare and Complex Epilepsies - EpiCARE), Bologna, Italy
| | - Chih-Hsiang Lin
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Kuang-Lin Lin
- Department of Pediatric Neurology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Tarja Linnankivi
- Child Neurology, New Childreńs Hospital, Helsinki, Finland
- Pediatric Research Center, University of Helsinki, Helsinki, Finland
- Helsinki University Hospital, Helsinki, Finland
| | - Warren Lo
- Department of Pediatrics and Neurology, Nationwide Children's Hospital, Columbus, OH, USA
| | - Daniel H Lowenstein
- Department of Neurology, University of California, San Francisco, CA 94143, USA
| | - Chelsea Lowther
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Laura Lubbers
- Citizens United for Research in Epilepsy, Chicago, Illinois, USA
| | - Colin H T Lui
- Department of Medicine, Tseung Kwan O Hospital, Hong Kong
| | - Lucia Inês Macedo-Souza
- Department of Biology, Institute of Biological Sciences and Center for Study on Human Genome, University of São Paulo, São Paulo, Brazil
| | - Rene Madeleyn
- Department of Pediatrics, Filderklinik, Filderstadt, Germany
| | | | - Stefania Magri
- Unit of Genetics of Neurodegenerative and Metabolic Diseases, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Louis Maillard
- Neurology Department, University Hospital of Nancy, UMR 7039, CNRS, Lorraine University, Nancy, France
| | - Lara Marcuse
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Paula Marques
- University Health Network, University of Toronto, Toronto, ON, Canada
| | - Anthony G Marson
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool L69 3GL, UK
| | | | - Patrick May
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette L-4362, Luxembourg
| | - Thomas Mayer
- Epilepsy Center Kleinwachau, Radeberg 01454, Germany
| | - Wendy McArdle
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 2BN, UK
| | - Steven M McCarroll
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Patricia McGoldrick
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Neurology, Boston Children's Health Physicians, Maria Fareri Children's Hospital at Westchester Medical Center, New York Medical College, New York, NY 10595, USA
| | | | - Andrew McIntosh
- Division of Psychiatry, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | | | | | - Davide Mei
- Neuroscience Department, Meyer Children's Hospital IRCCS, Florence, Italy
| | | | | | - Raffaella Minardi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, (Reference Center for Rare and Complex Epilepsies - EpiCARE), Bologna, Italy
| | - Martino Montomoli
- Neuroscience Department, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Barbara Mostacci
- IRCCS Istituto delle Scienze Neurologiche di Bologna, (Reference Center for Rare and Complex Epilepsies - EpiCARE), Bologna, Italy
| | - Lorenzo Muccioli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Hiltrud Muhle
- Department of Neuropediatrics, University Medical Center Schleswig-Holstein, Christian-Albrechts-University, Kiel, Germany
| | - Karen Müller-Schlüter
- Epilepsy Center for Children, University Hospital Ruppin-Brandenburg, Brandenburg Medical School, 16816 Neuruppin, Germany
| | - Imad M Najm
- Cleveland Clinic Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Neurology, Neurological Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Wassim Nasreddine
- Department of Neurology, American University of Beirut Medical Center, Beirut, Lebanon
| | - Samuel Neaves
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 2BN, UK
- MRC Integrative Epidemiology Unit at University of Bristol, Bristol BS8 2BN, UK
| | | | - Charles R J C Newton
- Neuroscience Unit, KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
- Department of Public Health, Pwani University, Kilifi, Kenya
- Department of Psychiatry, University of Oxford, Oxford, UK
- Department of Psychiatry, University of Cape Town, South Africa
| | | | - Kate Northstone
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 2BN, UK
| | - Sam Novod
- Data Sciences Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Terence J O'Brien
- Department of Medicine, University of Melbourne, Royal Melbourne Hospital, Parkville 3050, Australia
- Department of Neuroscience, The School of Translational Medicine, Alfred Health, Monash University, Melbourne 3004, Australia
| | - Seth Owusu-Agyei
- Kintampo Health Research Centre, Ghana Health Service, Kintampo, Ghana
- University of Health and Allied Science in Ho, Ghana
| | - Çiğdem Özkara
- Istanbul University-Cerrahpaşa, Cerrahpaşa Medical Faculty, Department of Neurology, Istanbul, Turkey
| | - Aarno Palotie
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Neurology, Massachusetts General Hospital, Boston, MA, USA
- Psychiatric & Neurodevelopmental Genetics Unit, Department of Psychiatry, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki 0014, Finland
| | | | - Elena Parrini
- Neuroscience Department, Meyer Children's Hospital IRCCS, Florence, Italy
- Department of NEUROFARBA, University of Florence, Florence, Italy
| | - Carlos Pato
- Departments of Psychiatry, Rutgers University, Robert Wood Johnson Medical School and New Jersey Medical School, New Brunswick, NJ, USA
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Michele Pato
- Departments of Psychiatry, Rutgers University, Robert Wood Johnson Medical School and New Jersey Medical School, New Brunswick, NJ, USA
- Department of Psychiatry and Behavioral Sciences, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Manuela Pendziwiat
- Department of Neuropediatrics, University Medical Center Schleswig-Holstein, Christian-Albrechts-University, Kiel, Germany
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | | | - Slavé Petrovski
- Department of Medicine, University of Melbourne, Royal Melbourne Hospital, Parkville 3050, Australia
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge CB2 0AA, UK
| | - William O Pickrell
- Swansea University Medical School, Swansea University, Swansea, Wales, UK
- Department of Neurology, Morriston Hospital, Swansea Bay University Bay Health Board, Swansea, Wales, UK
| | - Rebecca Pinsky
- Epilepsy Genetics Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| | - Dalila Pinto
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Tommaso Pippucci
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Medical Genetics Unit, Bologna, Italy
| | - Fabrizio Piras
- Neuropsychiatry Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Federica Piras
- Neuropsychiatry Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Annapurna Poduri
- Epilepsy Genetics Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| | - Federica Pondrelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Danielle Posthuma
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, VU Amsterdam, Amsterdam, the Netherlands
| | - Robert H W Powell
- Swansea University Medical School, Swansea University, Swansea, Wales, UK
- Department of Neurology, Morriston Hospital, Swansea Bay University Bay Health Board, Swansea, Wales, UK
| | - Michael Privitera
- Department of Neurology, Gardner Neuroscience Institute, University of Cincinnati Medical Center, Cincinnati, OH 45220, USA
| | - Annika Rademacher
- Department of Neuropediatrics, University Medical Center Schleswig-Holstein, Christian-Albrechts-University, Kiel, Germany
| | - Francesca Ragona
- Department of Pediatric Neuroscience, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Byron Ramirez-Hamouz
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sarah Rau
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen 72076, Germany
| | - Hillary R Raynes
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Mark I Rees
- Swansea University Medical School, Swansea University, Swansea, Wales, UK
- Brain & Mind Centre, Faculty of Medicine & Health, University of Sydney, Sydney, New South Wales, Australia
| | - Brigid M Regan
- Epilepsy Research Centre, University of Melbourne, Austin Health, Heidelberg 3084, Australia
| | - Andreas Reif
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt
- Department of Psychiatry, Psychotherapy and Psychosomatics, University Hospital Würzburg
| | - Eva Reinthaler
- Department of Neurology, Medical University of Vienna, Vienna 1090, Austria
| | - Sylvain Rheims
- Department of Functional Neurology and Epileptology, Hospices Civils de Lyon and University of Lyon, France
- Lyon's Neuroscience Research Center, INSERM U1028 / CNRS UMR 5292, Lyon, France
| | - Susan M Ring
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 2BN, UK
- MRC Integrative Epidemiology Unit at University of Bristol, Bristol BS8 2BN, UK
| | - Antonella Riva
- IRCCS Istituto Giannina Gaslini, Genova, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | - Enrique Rojas
- Division of Neurology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL USA
| | - Felix Rosenow
- Epilepsy Center Frankfurt Rhine-Main, Center of Neurology and Neurosurgery, Goethe University Frankfurt, Frankfurt, Germany
- LOEWE Center for Personalized Translational Epilepsy Research (CePTER), Goethe University Frankfurt, Germany
- Epilepsy Center Hessen-Marburg, Department of Neurology, Philipps University Marburg, Marburg, Germany
| | - Philippe Ryvlin
- Department of Clinical Neurosciences, Centre Hospitalo-Universitaire Vaudois, Lausanne, Switzerland
| | - Anni Saarela
- Kuopio Epilepsy Center, Neurocenter, Kuopio University Hospital, Kuopio 70210, Finland
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio 70210, Finland
| | - Lynette G Sadleir
- Department of Paediatrics and Child Health, University of Otago, Wellington, New Zealand
| | - Barış Salman
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Andrea Salmon
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | - Ilaria Sammarra
- Department of Medical and Surgical Sciences, Neuroscience Research Center, Magna Graecia University, Catanzaro, Italy
| | - Marcello Scala
- IRCCS Istituto Giannina Gaslini, Genova, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | - Steven Schachter
- Departments of Neurology, Beth Israel Deaconess Medical Center, Massachusetts General Hospital, and Harvard Medical School, Boston, MA 02215, USA
| | - André Schaller
- Institute of Human Genetics, Bern University Hospital, Bern, Switzerland
| | - Christoph J Schankin
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern 3010, Switzerland
- Department of Neurology, Ludwig Maximilians University, Munich, Germany
| | - Ingrid E Scheffer
- Epilepsy Research Centre, University of Melbourne, Austin Health, Heidelberg 3084, Australia
- Florey and Murdoch Children's Research Institutes, Parkville, Victoria 3052, Australia
- Department of Paediatrics, The University of Melbourne, Royal Children's Hospital, Parkville, Victoria 3052, Australia
| | - Natascha Schneider
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
- Chalfont Centre for Epilepsy, Chalfont-St-Peter, Buckinghamshire SL9 0RJ, UK
| | - Susanne Schubert-Bast
- Epilepsy Center Frankfurt Rhine-Main, Center of Neurology and Neurosurgery, Goethe University Frankfurt, Frankfurt, Germany
- LOEWE Center for Personalized Translational Epilepsy Research (CePTER), Goethe University Frankfurt, Germany
- Department of Neuropediatrics, Children's Hospital, Goethe University Frankfurt, Frankfurt, Germany
| | | | - Paolo Scudieri
- IRCCS Istituto Giannina Gaslini, Genova, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | - Lucie Sedláčková
- Department of Paediatric Neurology, 2nd Faculty of Medicine, Charles University and Motol Hospital, Prague, Czech Republic
| | - Catherine Shain
- Epilepsy Genetics Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| | - Pak C Sham
- Department of Psychiatry, The University of Hong Kong, Pokulam, Hong Kong
| | - Beth R Shiedley
- Epilepsy Genetics Program, Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Boston, MA, USA
| | - S Anthony Siena
- Medical School, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Graeme J Sills
- School of Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Sanjay M Sisodiya
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
- Chalfont Centre for Epilepsy, Chalfont-St-Peter, Buckinghamshire SL9 0RJ, UK
| | - Jordan W Smoller
- Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Psychiatry, Harvard Medical School, Boston, MA, USA
| | - Matthew Solomonson
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Gianfranco Spalletta
- Neuropsychiatry Laboratory, IRCCS Santa Lucia Foundation, Rome, Italy
- Division of Neuropsychiatry, Menninger Department of Psychiatry and Behavioral Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Kathryn R Sparks
- Division of Neurology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL USA
| | - Michael R Sperling
- Department of Neurology and Comprehensive Epilepsy Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Hannah Stamberger
- Applied & Translational Neurogenomics Group, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Neurology, Antwerp University Hospital, Edegem 2650, Belgium
- Translational Neurosciences, Faculty of Medicine and Health Science, University of Antwerp, Antwerp, Belgium
| | | | - Ulrich Stephani
- Department of Neuropediatrics, University Medical Center Schleswig-Holstein, Christian-Albrechts-University, Kiel, Germany
| | - Katalin Štěrbová
- Department of Paediatric Neurology, 2nd Faculty of Medicine, Charles University and Motol Hospital, Prague, Czech Republic
| | - William C Stewart
- Department of Pediatrics, Nationwide Children's Hospital, Columbia, Ohio, USA
| | - Carlotta Stipa
- IRCCS Istituto delle Scienze Neurologiche di Bologna, (Reference Center for Rare and Complex Epilepsies - EpiCARE), Bologna, Italy
| | - Pasquale Striano
- IRCCS Istituto Giannina Gaslini, Genova, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | - Adam Strzelczyk
- Epilepsy Center Frankfurt Rhine-Main, Center of Neurology and Neurosurgery, Goethe University Frankfurt, Frankfurt, Germany
- LOEWE Center for Personalized Translational Epilepsy Research (CePTER), Goethe University Frankfurt, Germany
- Epilepsy Center Hessen-Marburg, Department of Neurology, Philipps University Marburg, Marburg, Germany
| | - Rainer Surges
- Department of Epileptology, University of Bonn Medical Centre, Bonn 53127, Germany
| | - Toshimitsu Suzuki
- Department of Neurodevelopmental Disorder Genetics, Institute of Brain Science, Nagoya City University Graduate School of Medical Science, Nagoya, Aichi, Japan
- Laboratory for Neurogenetics, RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Mariagrazia Talarico
- Department of Medical and Surgical Sciences, Neuroscience Research Center, Magna Graecia University, Catanzaro, Italy
| | - Michael E Talkowski
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Randip S Taneja
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Oskari Timonen
- Institute of Clinical Medicine, University of Eastern Finland, Kuopio 70210, Finland
| | - Nicholas John Timpson
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol BS8 2BN, UK
- MRC Integrative Epidemiology Unit at University of Bristol, Bristol BS8 2BN, UK
| | - Paolo Tinuper
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- IRCCS Istituto delle Scienze Neurologiche di Bologna, (Reference Center for Rare and Complex Epilepsies - EpiCARE), Bologna, Italy
| | - Marian Todaro
- Department of Medicine, University of Melbourne, Royal Melbourne Hospital, Parkville 3050, Australia
- Department of Neuroscience, The School of Translational Medicine, Alfred Health, Monash University, Melbourne 3004, Australia
| | - Pınar Topaloglu
- Department of Child Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Meng-Han Tsai
- Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Birute Tumiene
- Centre for Medical Genetics, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
- Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Dilsad Turkdogan
- Department of Child Neurology, Medical School, Marmara University, Istanbul, Turkey
| | - Sibel Uğur-İşeri
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Algirdas Utkus
- Centre for Medical Genetics, Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania
- Institute of Biomedical Sciences, Faculty of Medicine, Vilnius University, Vilnius, Lithuania
| | - Priya Vaidiswaran
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104, USA
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children's Hospital of Philadelphia, Philadelphia, 3401 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Luc Valton
- Department of Neurology, UMR 5549, CNRS, Toulouse University Hospital, University of Toulouse, Toulouse, France
| | - Andreas van Baalen
- Department of Neuropediatrics, University Medical Center Schleswig-Holstein, Christian-Albrechts-University, Kiel, Germany
| | | | - Annalisa Vetro
- Neuroscience Department, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Markéta Vlčková
- Department of Paediatric Neurology, 2nd Faculty of Medicine, Charles University and Motol Hospital, Prague, Czech Republic
| | - Sophie von Brauchitsch
- Epilepsy Center Frankfurt Rhine-Main, Center of Neurology and Neurosurgery, Goethe University Frankfurt, Frankfurt, Germany
- LOEWE Center for Personalized Translational Epilepsy Research (CePTER), Goethe University Frankfurt, Germany
- Epilepsy Center Hessen-Marburg, Department of Neurology, Philipps University Marburg, Marburg, Germany
| | - Sarah von Spiczak
- Department of Neuropediatrics, University Medical Center Schleswig-Holstein, Christian-Albrechts-University, Kiel, Germany
- DRK-Northern German Epilepsy Centre for Children and Adolescents, 24223 Schwentinental-Raisdorf, Germany
| | - Ryan G Wagner
- MRC/Wits Rural Public Health & Health Transitions Research Unit (Agincourt), School of Public Health, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Epidemiology and Global Health, Umeå University, Umeå, Sweden
- Department of Clinical Sciences, Neurosciences, Umeå University, Umeå, Sweden
| | - Nick Watts
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Yvonne G Weber
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen 72076, Germany
- Department of Neurology and Epileptology, University of Aachen, Aachen 52074, Germany
| | - Sarah Weckhuysen
- Applied & Translational Neurogenomics Group, VIB Center for Molecular Neurology, VIB, Antwerp, Belgium
- Department of Neurology, Antwerp University Hospital, Edegem 2650, Belgium
- Translational Neurosciences, Faculty of Medicine and Health Science, University of Antwerp, Antwerp, Belgium
| | - Peter Widdess-Walsh
- School of Pharmacy and Biomolecular Sciences, The Royal College of Surgeons in Ireland, Dublin, Ireland
- The FutureNeuro Research Centre, Dublin, Ireland
- Department of Neurology, Beaumont Hospital, Dublin D09 FT51, Ireland
| | - Samuel Wiebe
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Community Health Sciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- O'Brien Institute for Public Health, University of Calgary, Calgary, Alberta, Canada
- Clinical Research Unit, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Steven M Wolf
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Neurology, Boston Children's Health Physicians, Maria Fareri Children's Hospital at Westchester Medical Center, New York Medical College, New York, NY 10595, USA
| | - Markus Wolff
- Department of Pediatric Neurology, Vivantes Hospital Neukölln, 12351 Berlin, Germany
| | - Stefan Wolking
- Department of Neurology and Epileptology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen 72076, Germany
- Department of Neurology and Epileptology, University of Aachen, Aachen 52074, Germany
| | - Isaac Wong
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Randi von Wrede
- Department of Epileptology, University of Bonn Medical Centre, Bonn 53127, Germany
| | - David Wu
- Human Genetics Training Program, Vanderbilt University, Nashville, TN, USA
| | - Kazuhiro Yamakawa
- Department of Neurodevelopmental Disorder Genetics, Institute of Brain Science, Nagoya City University Graduate School of Medical Science, Nagoya, Aichi, Japan
- Laboratory for Neurogenetics, RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Zuhal Yapıcı
- Department of Child Neurology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Uluc Yis
- Department of Child Neurology, Medical School, Dokuz Eylul University, Izmir, Turkey
| | - Robert Yolken
- Stanley Division of Developmental Neurovirology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Emrah Yücesan
- Bezmialem Vakif University, Institute of Life Sciences and Biotechnology, Istanbul, Turkey
| | - Sara Zagaglia
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London WC1N 3BG, UK
- Chalfont Centre for Epilepsy, Chalfont-St-Peter, Buckinghamshire SL9 0RJ, UK
| | - Felix Zahnert
- LOEWE Center for Personalized Translational Epilepsy Research (CePTER), Goethe University Frankfurt, Germany
- Epilepsy Center Hessen-Marburg, Department of Neurology, Philipps University Marburg, Marburg, Germany
| | - Federico Zara
- IRCCS Istituto Giannina Gaslini, Genova, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | - Fritz Zimprich
- Department of Neurology, Medical University of Vienna, Vienna 1090, Austria
| | - Milena Zizovic
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette L-4362, Luxembourg
| | - Gábor Zsurka
- Department of Epileptology, University of Bonn Medical Centre, Bonn 53127, Germany
- Institute of Experimental Epileptology and Cognition Research, Medical Faculty, University of Bonn, Bonn, Germany
| | - Benjamin M Neale
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Samuel F Berkovic
- Epilepsy Research Centre, University of Melbourne, Austin Health, Heidelberg 3084, Australia
| |
Collapse
|
46
|
Chen F, Peng S, Li C, Yang F, Yi Y, Chen X, Xu H, Cheng B, Xu Y, Xie X. Nitidine chloride inhibits mTORC1 signaling through ATF4-mediated Sestrin2 induction and targets IGF2R for lysosomal degradation. Life Sci 2024; 353:122918. [PMID: 39034027 DOI: 10.1016/j.lfs.2024.122918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/26/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024]
Abstract
AIMS Nitidine chloride (NC), a natural phytochemical alkaloid derived from Zanthoxylum nitidum (Roxb.) DC, exhibits multiple bioactivities, including antitumor, anti-inflammatory, and other therapeutic effects. However, the primary targets of NC and the mechanism of action (MOA) have not been explicitly defined. METHODS We explored the effects of NC on mTORC1 signaling by immunoblotting and fluorescence microscopy in wild-type and gene knockout cell lines generated by the CRISPR/Cas9 gene editing technique. We identified IGF2R as a direct target of NC via the drug affinity-responsive target stability (DARTS) method. We investigated the antitumor effects of NC using a mouse melanoma B16 tumor xenograft model. KEY FINDINGS NC inhibits mTORC1 activity by targeting amino acid-sensing signaling through activating transcription factor 4 (ATF4)-mediated Sestrin2 induction. NC directly binds to IGF2R and promotes its lysosomal degradation. Moreover, NC displayed potent cytotoxicity against various cancer cells and inhibited B16 tumor xenografts. SIGNIFICANCE NC inhibits mTORC1 signaling through nutrient sensing and directly targets IGF2R for lysosomal degradation, providing mechanistic insights into the MOA of NC.
Collapse
Affiliation(s)
- Fengzhi Chen
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Shujun Peng
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Canrong Li
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Fan Yang
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Yuguo Yi
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Xinyu Chen
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Haolun Xu
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Baicheng Cheng
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China
| | - Yumin Xu
- Department of Infectious Diseases & Department of Hospital Infection Management, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoduo Xie
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Sun Yat-sen University, Shenzhen, China.
| |
Collapse
|
47
|
Reifenberg P, Zimmer A. Branched-chain amino acids: physico-chemical properties, industrial synthesis and role in signaling, metabolism and energy production. Amino Acids 2024; 56:51. [PMID: 39198298 PMCID: PMC11358235 DOI: 10.1007/s00726-024-03417-2] [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: 06/13/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024]
Abstract
Branched-chain amino acids (BCAAs)-leucine (Leu), isoleucine (Ile), and valine (Val)-are essential nutrients with significant roles in protein synthesis, metabolic regulation, and energy production. This review paper offers a detailed examination of the physico-chemical properties of BCAAs, their industrial synthesis, and their critical functions in various biological processes. The unique isomerism of BCAAs is presented, focusing on analytical challenges in their separation and quantification as well as their solubility characteristics, which are crucial for formulation and purification applications. The industrial synthesis of BCAAs, particularly using bacterial strains like Corynebacterium glutamicum, is explored, alongside methods such as genetic engineering aimed at enhancing production, detailing the enzymatic processes and specific precursors. The dietary uptake, distribution, and catabolism of BCAAs are reviewed as fundamental components of their physiological functions. Ultimately, their multifaceted impact on signaling pathways, immune function, and disease progression is discussed, providing insights into their profound influence on muscle protein synthesis and metabolic health. This comprehensive analysis serves as a resource for understanding both the basic and complex roles of BCAAs in biological systems and their industrial application.
Collapse
Affiliation(s)
- Philipp Reifenberg
- Merck Life Science KGaA, Upstream R&D, Frankfurter Strasse 250, 64293, Darmstadt, Germany
- Institute for Organic Chemistry and Biochemistry, Technische Universität Darmstadt, Alarich‑Weiss‑Strasse 4, 64287, Darmstadt, Germany
| | - Aline Zimmer
- Merck Life Science KGaA, Upstream R&D, Frankfurter Strasse 250, 64293, Darmstadt, Germany.
| |
Collapse
|
48
|
Valenstein ML, Lalgudi PV, Gu X, Kedir JF, Taylor MS, Chivukula RR, Sabatini DM. Rag-Ragulator is the central organizer of the physical architecture of the mTORC1 nutrient-sensing pathway. Proc Natl Acad Sci U S A 2024; 121:e2322755121. [PMID: 39163330 PMCID: PMC11363303 DOI: 10.1073/pnas.2322755121] [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: 12/24/2023] [Accepted: 07/12/2024] [Indexed: 08/22/2024] Open
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) pathway regulates cell growth and metabolism in response to many environmental cues, including nutrients. Amino acids signal to mTORC1 by modulating the guanine nucleotide loading states of the heterodimeric Rag GTPases, which bind and recruit mTORC1 to the lysosomal surface, its site of activation. The Rag GTPases are tethered to the lysosome by the Ragulator complex and regulated by the GATOR1, GATOR2, and KICSTOR multiprotein complexes that localize to the lysosomal surface through an unknown mechanism(s). Here, we show that mTORC1 is completely insensitive to amino acids in cells lacking the Rag GTPases or the Ragulator component p18. Moreover, not only are the Rag GTPases and Ragulator required for amino acids to regulate mTORC1, they are also essential for the lysosomal recruitment of the GATOR1, GATOR2, and KICSTOR complexes, which stably associate and traffic to the lysosome as the "GATOR" supercomplex. The nucleotide state of RagA/B controls the lysosomal association of GATOR, in a fashion competitively antagonized by the N terminus of the amino acid transporter SLC38A9. Targeting of Ragulator to the surface of mitochondria is sufficient to relocalize the Rags and GATOR to this organelle, but not to enable the nutrient-regulated recruitment of mTORC1 to mitochondria. Thus, our results reveal that the Rag-Ragulator complex is the central organizer of the physical architecture of the mTORC1 nutrient-sensing pathway and underscore that mTORC1 activation requires signal transduction on the lysosomal surface.
Collapse
Affiliation(s)
- Max L. Valenstein
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114
- Whitehead Institute for Biomedical Research, Cambridge, MA02142
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA02139
- Harvard Medical School, Boston, MA02115
| | - Pranav V. Lalgudi
- Whitehead Institute for Biomedical Research, Cambridge, MA02142
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Xin Gu
- Whitehead Institute for Biomedical Research, Cambridge, MA02142
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA02139
- Harvard Medical School, Boston, MA02115
| | - Jibril F. Kedir
- Whitehead Institute for Biomedical Research, Cambridge, MA02142
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA02139
- Harvard Medical School, Boston, MA02115
| | - Martin S. Taylor
- Harvard Medical School, Boston, MA02115
- Department of Pathology, Massachusetts General Hospital, Boston, MA02114
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI02903
- Brown Center on the Biology of Aging, Brown University, Providence, RI02903
- Legorreta Cancer Center, Brown University, Providence, RI02903
| | - Raghu R. Chivukula
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114
- Harvard Medical School, Boston, MA02115
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA02114
- Department of Surgery, Massachusetts General Hospital, Boston, MA02114
- Broad Institute of Harvard and the Massachusetts Institute of Technology, Cambridge, MA02142
| | - David M. Sabatini
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague166 10, Czech Republic
| |
Collapse
|
49
|
Liu X, Guo B, Li Q, Nie J. mTOR in metabolic homeostasis and disease. Exp Cell Res 2024; 441:114173. [PMID: 39047807 DOI: 10.1016/j.yexcr.2024.114173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 07/20/2024] [Accepted: 07/21/2024] [Indexed: 07/27/2024]
Abstract
The ability to maintain cellular metabolic homeostasis is critical to life, in which mTOR plays an important role. This kinase integrates upstream nutrient signals and performs essential functions in physiology and metabolism by increasing metabolism and suppressing autophagy. Thus, dysregulation of mTOR activity leads to diseases, especially metabolic diseases such as cancer, type 2 diabetes and neurological disorders. Therefore, inhibition of overactivated mTOR becomes a rational approach to treat a variety of metabolic diseases. In this review, we discuss how mTOR responds to upstream signals and how mTOR regulates metabolic processes, including protein, nucleic acid, and lipid metabolism. Furthermore, we discuss the possible causes and consequences of dysregulated mTOR signaling activity, and summarize relevant applications, such as inhibition of mTOR activity to treat these diseases. This review will advance our comprehensive knowledge of the association between mTOR and metabolic homeostasis, which has significant ramifications for human health.
Collapse
Affiliation(s)
- Xuejia Liu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Bin Guo
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Qiye Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Jing Nie
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China.
| |
Collapse
|
50
|
Lehmer M, Zoncu R. mTORC1 Signaling Inhibition Modulates Mitochondrial Function in Frataxin Deficiency. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.06.606942. [PMID: 39211218 PMCID: PMC11360942 DOI: 10.1101/2024.08.06.606942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Lysosomes regulate mitochondrial function through multiple mechanisms including the master regulator, mechanistic Target of Rapamycin Complex 1 (mTORC1) protein kinase, which is activated at the lysosomal membrane by nutrient, growth factor and energy signals. mTORC1 promotes mitochondrial protein composition changes, respiratory capacity, and dynamics, though the full range of mitochondrial-regulating functions of this protein kinase remain undetermined. We find that acute chemical modulation of mTORC1 signaling decreased mitochondrial oxygen consumption, increased mitochondrial membrane potential and reduced susceptibility to stress-induced mitophagy. In cellular models of Friedreich's Ataxia (FA), where loss of the Frataxin (FXN) protein suppresses Fe-S cluster synthesis and mitochondrial respiration, the changes induced by mTORC1 inhibitors lead to improved cell survival. Proteomic-based profiling uncover compositional changes that could underlie mTORC1-dependent modulation of FXN-deficient mitochondria. These studies highlight mTORC1 signaling as a regulator of mitochondrial composition and function, prompting further evaluation of this pathway in the context of mitochondrial disease.
Collapse
|