1
|
O'Sullivan PA, Aidarova A, Afonina IS, Manils J, Thurston TLM, Instrell R, Howell M, Boeing S, Ranawana S, Herpels MB, Chetian R, Bassa M, Flynn H, Frith D, Snijders AP, Howes A, Beyaert R, Bowcock AM, Ley SC. CARD14 signalosome formation is associated with its endosomal relocation and mTORC1-induced keratinocyte proliferation. Biochem J 2024; 481:1143-1171. [PMID: 39145956 PMCID: PMC11555713 DOI: 10.1042/bcj20240058] [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: 02/14/2024] [Revised: 08/13/2024] [Accepted: 08/15/2024] [Indexed: 08/16/2024]
Abstract
Rare mutations in CARD14 promote psoriasis by inducing CARD14-BCL10-MALT1 complexes that activate NF-κB and MAP kinases. Here, the downstream signalling mechanism of the highly penetrant CARD14E138A alteration is described. In addition to BCL10 and MALT1, CARD14E138A associated with several proteins important in innate immune signalling. Interactions with M1-specific ubiquitin E3 ligase HOIP, and K63-specific ubiquitin E3 ligase TRAF6 promoted BCL10 ubiquitination and were essential for NF-κB and MAP kinase activation. In contrast, the ubiquitin binding proteins A20 and ABIN1, both genetically associated with psoriasis development, negatively regulated signalling by inducing CARD14E138A turnover. CARD14E138A localized to early endosomes and was associated with the AP2 adaptor complex. AP2 function was required for CARD14E138A activation of mTOR complex 1 (mTORC1), which stimulated keratinocyte metabolism, but not for NF-κB nor MAP kinase activation. Furthermore, rapamycin ameliorated CARD14E138A-induced keratinocyte proliferation and epidermal acanthosis in mice, suggesting that blocking mTORC1 may be therapeutically beneficial in CARD14-dependent psoriasis.
Collapse
Affiliation(s)
- Paul A. O'Sullivan
- The Francis Crick Institute, London NW1 1AT, U.K
- Institute of Immunity and Transplantation, University College London, London NW3 2PP, U.K
| | - Aigerim Aidarova
- VIB Center for Inflammation Research and Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Inna S. Afonina
- VIB Center for Inflammation Research and Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Joan Manils
- The Francis Crick Institute, London NW1 1AT, U.K
- Institute of Immunity and Transplantation, University College London, London NW3 2PP, U.K
- Immunology Unit, Department of Pathology and Experimental Therapy, School of Medicine, University of Barcelona, Barcelona, Spain
| | - Teresa L. M. Thurston
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London SW7 2AZ, U.K
| | | | | | | | - Sashini Ranawana
- Institute of Immunity and Transplantation, University College London, London NW3 2PP, U.K
| | - Melanie B. Herpels
- Institute of Immunity and Transplantation, University College London, London NW3 2PP, U.K
| | - Riwia Chetian
- Institute of Immunity and Transplantation, University College London, London NW3 2PP, U.K
| | - Matilda Bassa
- Institute of Immunity and Transplantation, University College London, London NW3 2PP, U.K
| | - Helen Flynn
- The Francis Crick Institute, London NW1 1AT, U.K
| | - David Frith
- The Francis Crick Institute, London NW1 1AT, U.K
| | | | - Ashleigh Howes
- National Heart and Lung Institute, Imperial College London, London W12 0NN, U.K
| | - Rudi Beyaert
- VIB Center for Inflammation Research and Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Anne M. Bowcock
- Department of Oncological Science, Dermatology, and Genetics and Genome Sciences, Icahn School of Medicine at Mount Sinai, New York 10029, U.S.A
| | - Steven C. Ley
- Institute of Immunity and Transplantation, University College London, London NW3 2PP, U.K
| |
Collapse
|
2
|
Tang J, Lam GT, Brooks RD, Miles M, Useckaite Z, Johnson IR, Ung BSY, Martini C, Karageorgos L, Hickey SM, Selemidis S, Hopkins AM, Rowland A, Vather R, O'Leary JJ, Brooks DA, Caruso MC, Logan JM. Exploring the role of sporadic BRAF and KRAS mutations during colorectal cancer pathogenesis: A spotlight on the contribution of the endosome-lysosome system. Cancer Lett 2024; 585:216639. [PMID: 38290660 DOI: 10.1016/j.canlet.2024.216639] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/21/2023] [Accepted: 12/30/2023] [Indexed: 02/01/2024]
Abstract
The highly heterogenous nature of colorectal cancer can significantly hinder its early and accurate diagnosis, eventually contributing to high mortality rates. The adenoma-carcinoma sequence and serrated polyp-carcinoma sequence are the two most common sequences in sporadic colorectal cancer. Genetic alterations in adenomatous polyposis coli (APC), v-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (KRAS) and tumour protein 53 (TP53) genes are critical in adenoma-carcinoma sequence, whereas v-Raf murine sarcoma viral oncogene homolog B (BRAF) and MutL Homolog1 (MLH1) are driving oncogenes in the serrated polyp-carcinoma sequence. Sporadic mutations in these genes contribute differently to colorectal cancer pathogenesis by introducing distinct alterations in several signalling pathways that rely on the endosome-lysosome system. Unsurprisingly, the endosome-lysosome system plays a pivotal role in the hallmarks of cancer and contributes to specialised colon function. Thus, the endosome-lysosome system might be distinctively influenced by different mutations and these alterations may contribute to the heterogenous nature of sporadic colorectal cancer. This review highlights potential connections between major sporadic colorectal cancer mutations and the diverse pathogenic mechanisms driven by the endosome-lysosome system in colorectal carcinogenesis.
Collapse
Affiliation(s)
- Jingying Tang
- Clinical and Health Sciences, University of South Australia, North Terrace, Adelaide, South Australia, Australia
| | - Giang T Lam
- Clinical and Health Sciences, University of South Australia, North Terrace, Adelaide, South Australia, Australia
| | - Robert D Brooks
- Clinical and Health Sciences, University of South Australia, North Terrace, Adelaide, South Australia, Australia
| | - Mark Miles
- School of Health and Biomedical Sciences, STEM College, RMIT University, Bundoora, Melbourne, Vic, Australia
| | - Zivile Useckaite
- College of Medicine and Public Health, Flinders University, Flinders Drive, Bedford Park, Adelaide, SA, Australia
| | - Ian Rd Johnson
- Clinical and Health Sciences, University of South Australia, North Terrace, Adelaide, South Australia, Australia
| | - Ben S-Y Ung
- Clinical and Health Sciences, University of South Australia, North Terrace, Adelaide, South Australia, Australia
| | - Carmela Martini
- Clinical and Health Sciences, University of South Australia, North Terrace, Adelaide, South Australia, Australia
| | - Litsa Karageorgos
- Clinical and Health Sciences, University of South Australia, North Terrace, Adelaide, South Australia, Australia
| | - Shane M Hickey
- Clinical and Health Sciences, University of South Australia, North Terrace, Adelaide, South Australia, Australia
| | - Stavros Selemidis
- School of Health and Biomedical Sciences, STEM College, RMIT University, Bundoora, Melbourne, Vic, Australia
| | - Ashley M Hopkins
- College of Medicine and Public Health, Flinders University, Flinders Drive, Bedford Park, Adelaide, SA, Australia
| | - Andrew Rowland
- College of Medicine and Public Health, Flinders University, Flinders Drive, Bedford Park, Adelaide, SA, Australia
| | - Ryash Vather
- Colorectal Unit, Department of Surgery, Royal Adelaide Hospital, Adelaide, South Australia, Australia; Centre for Cancer Biology, University of South Australia, Adelaide, South Australia, Australia
| | - John J O'Leary
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
| | - Douglas A Brooks
- Clinical and Health Sciences, University of South Australia, North Terrace, Adelaide, South Australia, Australia
| | - Maria C Caruso
- Clinical and Health Sciences, University of South Australia, North Terrace, Adelaide, South Australia, Australia
| | - Jessica M Logan
- Clinical and Health Sciences, University of South Australia, North Terrace, Adelaide, South Australia, Australia.
| |
Collapse
|
3
|
Ghosh S, Hom Choudhury S, Mukherjee K, Bhattacharyya SN. HuR-miRNA complex activates RAS GTPase RalA to facilitate endosome targeting and extracellular export of miRNAs. J Biol Chem 2024; 300:105750. [PMID: 38360271 PMCID: PMC10956062 DOI: 10.1016/j.jbc.2024.105750] [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: 10/01/2023] [Revised: 02/01/2024] [Accepted: 02/07/2024] [Indexed: 02/17/2024] Open
Abstract
Extracellular vesicles-mediated exchange of miRNA cargos between diverse types of mammalian cells is a major mechanism of controlling cellular miRNA levels and activity, thus regulating the expression of miRNA-target genes in both donor and recipient cells. Despite tremendous excitement related to extracellular vesicles-associated miRNAs as biomarkers or having therapeutic potential, the mechanism of selective packaging of miRNAs into endosomes and multivesicular bodies for subsequent extracellular export is poorly studied due to the lack of an in vitro assay system. Here, we have developed an in vitro assay with endosomes isolated from mammalian macrophage cells to follow miRNA packaging into endocytic organelles. The synthetic miRNAs, used in the assay, get imported inside the isolated endosomes during the in vitro reaction and become protected from RNase in a time- and concentration-dependent manner. The selective miRNA accumulation inside endosomes requires both ATP and GTP hydrolysis and the miRNA-binding protein HuR. The HuR-miRNA complex binds and stimulates the endosomal RalA GTPase to facilitate the import of miRNAs into endosomes and their subsequent export as part of the extracellular vesicles. The endosomal targeting of miRNAs is also very much dependent on the endosome maturation process that is controlled by Rab5 protein and ATP. In summary, we provide an in vitro method to aid in the investigation of the mechanism of miRNA packaging process for its export from mammalian macrophage cells.
Collapse
Affiliation(s)
- Syamantak Ghosh
- RNA Biology Research Laboratory, Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Sourav Hom Choudhury
- RNA Biology Research Laboratory, Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
| | - Kamalika Mukherjee
- RNA Biology Research Laboratory, Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India; Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center (UNMC), Nebraska, USA.
| | - Suvendra N Bhattacharyya
- RNA Biology Research Laboratory, Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India; Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center (UNMC), Nebraska, USA.
| |
Collapse
|
4
|
Tanimoto A, Yamaguchi Y, Kadowaki T, Sakai E, Oyakawa S, Ono Y, Yoshida N, Tsukuba T. Rab44 negatively regulates myoblast differentiation by controlling fusogenic protein transport and mTORC1 signaling. J Cell Biochem 2023; 124:1486-1502. [PMID: 37566644 DOI: 10.1002/jcb.30457] [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: 12/17/2022] [Revised: 06/27/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023]
Abstract
Skeletal muscle is composed of multinucleated myotubes formed by the fusion of mononucleated myoblasts. Skeletal muscle differentiation, termed as myogenesis, have been investigated using the mouse skeletal myoblast cell line C2C12. It has been reported that several "small" Rab proteins, major membrane-trafficking regulators, possibly regulate membrane protein transport in C2C12 cells; however, the role of Rab proteins in myogenesis remains unexplored. Rab44, a member of "large" Rab GTPases, has recently been identified as a negative regulator of osteoclast differentiation. In this study, using C2C12 cells, we found that Rab44 expression was upregulated during myoblast differentiation into myotubes. Knockdown of Rab44 enhanced myoblast differentiation and myotube formation. Consistent with these results, Rab44 knockdown in myoblasts increased expression levels of several myogenic marker genes. Rab44 knockdown increased the surface accumulation of myomaker and myomixer, two fusogenic proteins required for multinucleation, implying enhanced cell fusion. Conversely, Rab44 overexpression inhibited myoblast differentiation and tube formation, accompanied by decreased expression of some myogenic markers. Furthermore, Rab44 was found to be predominantly localized in lysosomes, and Rab44 overexpression altered the number and size of lysosomes. Considering the underlying molecular mechanism, Rab44 overexpression impaired the signaling pathway of the mechanistic target of rapamycin complex1 (mTORC1) in C2C12 cells. Namely, phosphorylation levels of mTORC1 and downstream mTORC1 substrates, such as S6 and P70-S6K, were notably lower in Rab44 overexpressing cells than those in control cells. These results indicate that Rab44 negatively regulates myoblast differentiation into myotubes by controlling fusogenic protein transport and mTORC1 signaling.
Collapse
Affiliation(s)
- Ayuko Tanimoto
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Yu Yamaguchi
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Tomoko Kadowaki
- Department of Frontier Oral Science, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Eiko Sakai
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Shun Oyakawa
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Yusuke Ono
- Department of Muscle Development and Regeneration, Kumamoto University, Kumamoto, Japan
| | - Noriaki Yoshida
- Department of Orthodontics and Dentofacial Orthopedics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| | - Takayuki Tsukuba
- Department of Dental Pharmacology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
5
|
Lin JX, Xu CY, Wu XM, Che L, Li TY, Mo SM, Guo DB, Lin ZN, Lin YC. Rab7a-mTORC1 signaling-mediated cholesterol trafficking from the lysosome to mitochondria ameliorates hepatic lipotoxicity induced by aflatoxin B1 exposure. CHEMOSPHERE 2023; 320:138071. [PMID: 36754296 DOI: 10.1016/j.chemosphere.2023.138071] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 01/10/2023] [Accepted: 02/05/2023] [Indexed: 06/18/2023]
Abstract
Aflatoxin B1 (AFB1) is a common contaminant in many foodstuffs and is considered a public health concern worldwide due to its hepatotoxicity caused by lipid metabolism disorders. However, the molecular mechanism underlying AFB1-induced lipotoxicity-dependent liver injury via regulating cholesterol metabolism remains unclear. We established a cholesterol trafficking disorder-mediated hepatic lipotoxicity model with AFB1 mixture exposure in vitro (HepaRG and HepG2 cells, 1.6 μM for 36 h) and in vivo (C57BL/6 mice, 3 mg kg-1, i.g., every other day for 6 weeks). In vitro, the interaction between lysosomal Niemann-Pick type C1 (NPC1) protein and mitochondrial translocator protein (TSPO) regulated lipotoxicity induced by AFB1 mixture exposure, including lysosomal membrane permeabilization and mitochondria-dependent necroptosis. Moreover, the downregulation of lysosomal Ras-associated protein 7a (Rab7a) enhanced the mammalian target of rapamycin complex 1 (mTORC1)-mediated disorders of cholesterol trafficking from the lysosome to mitochondria. Furthermore, cholesterol trafficking disorder-mediated hepatic lipotoxicity induced by the low-dose level of AFB1 exposure was relieved by genetic or pharmaceutic activation of Rab7a to inhibit mTORC1 in vitro and ex vivo. In vivo, mTORC1 inhibitor (Torin1, 4 mg kg-1, i.p., every other day for 3 weeks) alleviated the cholesterol trafficking disorder-mediated hepatic lipotoxicity via upregulating the molecular machinery of lysosomes and mitochondria contact mediated by NPC1 and TSPO interaction in the low dose of AFB1 exposure. Altogether, our data suggested a novel mechanism that lysosomal Rab7a-mTORC1 signaling determined the cholesterol trafficking regulated by NPC1-TSPO from the lysosome to mitochondria, which promoted hepatic lipotoxicity via lysosomal quality control and mitochondria-dependent necroptosis signaling pathways in chemical mixture exposure.
Collapse
Affiliation(s)
- Jin-Xian Lin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Chi-Yu Xu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Xin-Mou Wu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Lin Che
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Ting-Yu Li
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Su-Min Mo
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Dong-Bei Guo
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Zhong-Ning Lin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, 361102, China.
| | - Yu-Chun Lin
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen, 361102, China.
| |
Collapse
|
6
|
Kahilainen A, Oostra V, Somervuo P, Minard G, Saastamoinen M. Alternative developmental and transcriptomic responses to host plant water limitation in a butterfly metapopulation. Mol Ecol 2022; 31:5666-5683. [PMID: 34516691 DOI: 10.1111/mec.16178] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 08/06/2021] [Accepted: 09/02/2021] [Indexed: 01/13/2023]
Abstract
Predicting how climate change affects biotic interactions poses a challenge. Plant-insect herbivore interactions are particularly sensitive to climate change, as climate-induced changes in plant quality cascade into the performance of insect herbivores. Whereas the immediate survival of herbivore individuals depends on plastic responses to climate change-induced nutritional stress, long-term population persistence via evolutionary adaptation requires genetic variation for these responses. To assess the prospects for population persistence under climate change, it is therefore crucial to characterize response mechanisms to climate change-induced stressors, and quantify their variability in natural populations. Here, we test developmental and transcriptomic responses to water limitation-induced host plant quality change in a Glanville fritillary butterfly (Melitaea cinxia) metapopulation. We combine nuclear magnetic resonance spectroscopy on the plant metabolome, larval developmental assays and an RNA sequencing analysis of the larval transcriptome. We observed that responses to feeding on water-limited plants, in which amino acids and aromatic compounds are enriched, showed marked variation within the metapopulation, with individuals of some families performing better on control and others on water-limited plants. The transcriptomic responses were concordant with the developmental responses: families exhibiting opposite developmental responses also produced opposite transcriptomic responses (e.g. in growth-associated transcripts). The divergent responses in both larval development and transcriptome are associated with differences between families in amino acid catabolism and storage protein production. The results reveal intrapopulation variability in plasticity, suggesting that the Finnish M. cinxia metapopulation harbours potential for buffering against drought-induced changes in host plant quality.
Collapse
Affiliation(s)
- Aapo Kahilainen
- Organismal and Evolutionary Biology Research Programme, University of Helsinki, P.O. Box 65, Helsinki, FIN-00014, Finland
| | - Vicencio Oostra
- Organismal and Evolutionary Biology Research Programme, University of Helsinki, P.O. Box 65, Helsinki, FIN-00014, Finland.,Department of Evolution, Ecology and Behaviour, University of Liverpool, Crown Street, Liverpool, L69 7ZB, United Kingdom
| | - Panu Somervuo
- Organismal and Evolutionary Biology Research Programme, University of Helsinki, P.O. Box 65, Helsinki, FIN-00014, Finland
| | - Guillaume Minard
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS, INRAe, VetAgro Sup, UMR Ecologie Microbienne, Villeurbanne, France
| | - Marjo Saastamoinen
- Organismal and Evolutionary Biology Research Programme, University of Helsinki, P.O. Box 65, Helsinki, FIN-00014, Finland.,Helsinki Institute of Life Science, University of Helsinki, Finland
| |
Collapse
|
7
|
Hees JT, Harbauer AB. Metabolic Regulation of Mitochondrial Protein Biogenesis from a Neuronal Perspective. Biomolecules 2022; 12:1595. [PMID: 36358945 PMCID: PMC9687362 DOI: 10.3390/biom12111595] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 09/29/2023] Open
Abstract
Neurons critically depend on mitochondria for ATP production and Ca2+ buffering. They are highly compartmentalized cells and therefore a finely tuned mitochondrial network constantly adapting to the local requirements is necessary. For neuronal maintenance, old or damaged mitochondria need to be degraded, while the functional mitochondrial pool needs to be replenished with freshly synthesized components. Mitochondrial biogenesis is known to be primarily regulated via the PGC-1α-NRF1/2-TFAM pathway at the transcriptional level. However, while transcriptional regulation of mitochondrial genes can change the global mitochondrial content in neurons, it does not explain how a morphologically complex cell such as a neuron adapts to local differences in mitochondrial demand. In this review, we discuss regulatory mechanisms controlling mitochondrial biogenesis thereby making a case for differential regulation at the transcriptional and translational level. In neurons, additional regulation can occur due to the axonal localization of mRNAs encoding mitochondrial proteins. Hitchhiking of mRNAs on organelles including mitochondria as well as contact site formation between mitochondria and endolysosomes are required for local mitochondrial biogenesis in axons linking defects in any of these organelles to the mitochondrial dysfunction seen in various neurological disorders.
Collapse
Affiliation(s)
- Jara Tabitha Hees
- TUM Medical Graduate Center, Technical University of Munich, 81675 Munich, Germany
- Max Planck Institute for Biological Intelligence, in Foundation, 82152 Planegg-Martinsried, Germany
| | - Angelika Bettina Harbauer
- Max Planck Institute for Biological Intelligence, in Foundation, 82152 Planegg-Martinsried, Germany
- Institute of Neuronal Cell Biology, Technical University of Munich, 80802 Munich, Germany
- Munich Cluster for Systems Neurology, 81377 Munich, Germany
| |
Collapse
|
8
|
Jiang W, Ou Z, Zhu Q, Zai H. RagC GTPase regulates mTOR to promote chemoresistance in senescence-like HepG2 cells. Front Physiol 2022; 13:949737. [PMID: 36267578 PMCID: PMC9577253 DOI: 10.3389/fphys.2022.949737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 09/20/2022] [Indexed: 11/23/2022] Open
Abstract
Radiotherapy and chemotherapy can arrest cancer cells in a senescence-like state, which can lead to therapy resistance and cancer relapse. mTOR is hyperactivated in senescent cells but the mechanisms remain unclear. In this study, we examine the roles of several mTOR-regulated GTPases in senescence-like liver cancer cells and the mechanisms in drug resistance. We show that although RagC, Rheb, Rab1A, Rab5 and Arf1 GTPases were required for optimal mTOR activation in proliferating HepG2 cells, only RagC and Rheb are required in the senescence-like counterparts. Consistently, the drug resistance of the senescence-like HepG2 can be reduced by knocking down RagC and Rheb but not the other GTPases. Autophagic and lysosomal activity were increased in senescence-like cells; pharmacological inhibition of autophagy-lysosome decreased mTOR activity and preferentially sensitized senescence-like HepG2 cells to chemotherapy drugs including trametinib, cisplatin, and doxorubicin. In liver cancer patients, expression of RagC and Rheb but not other GTPases examined was associated with unfavorable prognosis. Our study therefore has defined a key role of Rag-Rheb GTPase in mediating mTOR activation and drug resistance in senescence-like HepG2 cells, which could have important implications in developing second-line treatments for liver cancer patients.
Collapse
Affiliation(s)
- Wei Jiang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan, China
| | - Zhenglin Ou
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan, China
| | - Qin Zhu
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan, China
| | - Hongyan Zai
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, Hunan, China
- *Correspondence: Hongyan Zai,
| |
Collapse
|
9
|
Iskarpatyoti JA, Shi J, Abraham MA, Rathore APS, Miao Y, Abraham SN. Mast cell regranulation requires a metabolic switch involving mTORC1 and a glucose-6-phosphate transporter. Cell Rep 2022; 40:111346. [PMID: 36170813 PMCID: PMC11218746 DOI: 10.1016/j.celrep.2022.111346] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 06/16/2022] [Accepted: 08/22/2022] [Indexed: 11/17/2022] Open
Abstract
Mast cells (MCs) are granulated cells implicated in inflammatory disorders because of their capacity to degranulate, releasing prestored proinflammatory mediators. As MCs have the unique capacity to reform granules following degranulation in vitro, their potential to regranulate in vivo is linked to their pathogenesis. It is not known what factors regulate regranulation, let alone if regranulation occurs in vivo. We report that mice can undergo multiple bouts of MC regranulation following successive anaphylactic reactions. mTORC1, a nutrient sensor that activates protein and lipid synthesis, is necessary for regranulation. mTORC1 activity is regulated by a glucose-6-phosphate transporter, Slc37a2, which increases intracellular glucose-6-phosphate and ATP during regranulation, two upstream signals of mTOR. Additionally, Slc37a2 concentrates extracellular metabolites within endosomes, which are trafficked into nascent granules. Thus, the metabolic switch associated with MC regranulation is mediated by the interactions of a cellular metabolic sensor and a transporter of extracellular metabolites into MC granules.
Collapse
Affiliation(s)
- Jason A Iskarpatyoti
- Department of Molecular Genetics & Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Jianling Shi
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Mathew A Abraham
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Abhay P S Rathore
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yuxuan Miao
- Ben May Department of Cancer Research, The University of Chicago, Chicago, IL 60637, USA
| | - Soman N Abraham
- Department of Molecular Genetics & Microbiology, Duke University Medical Center, Durham, NC 27710, USA; Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA; Department of Immunology, Duke University Medical Center, Durham, NC 27710, USA; Program in Emerging Infectious Diseases, Duke-National University of Singapore, Singapore 169857, Singapore.
| |
Collapse
|
10
|
Linking Late Endosomal Cholesterol with Cancer Progression and Anticancer Drug Resistance. Int J Mol Sci 2022; 23:ijms23137206. [PMID: 35806209 PMCID: PMC9267071 DOI: 10.3390/ijms23137206] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 06/22/2022] [Accepted: 06/25/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer cells undergo drastic metabolic adaptions to cover increased bioenergetic needs, contributing to resistance to therapies. This includes a higher demand for cholesterol, which often coincides with elevated cholesterol uptake from low-density lipoproteins (LDL) and overexpression of the LDL receptor in many cancers. This implies the need for cancer cells to accommodate an increased delivery of LDL along the endocytic pathway to late endosomes/lysosomes (LE/Lys), providing a rapid and effective distribution of LDL-derived cholesterol from LE/Lys to other organelles for cholesterol to foster cancer growth and spread. LDL-cholesterol exported from LE/Lys is facilitated by Niemann–Pick Type C1/2 (NPC1/2) proteins, members of the steroidogenic acute regulatory-related lipid transfer domain (StARD) and oxysterol-binding protein (OSBP) families. In addition, lysosomal membrane proteins, small Rab GTPases as well as scaffolding proteins, including annexin A6 (AnxA6), contribute to regulating cholesterol egress from LE/Lys. Here, we summarize current knowledge that links upregulated activity and expression of cholesterol transporters and related proteins in LE/Lys with cancer growth, progression and treatment outcomes. Several mechanisms on how cellular distribution of LDL-derived cholesterol from LE/Lys influences cancer cell behavior are reviewed, some of those providing opportunities for treatment strategies to reduce cancer progression and anticancer drug resistance.
Collapse
|
11
|
Altas B, Romanowski AJ, Bunce GW, Poulopoulos A. Neuronal mTOR Outposts: Implications for Translation, Signaling, and Plasticity. Front Cell Neurosci 2022; 16:853634. [PMID: 35465614 PMCID: PMC9021820 DOI: 10.3389/fncel.2022.853634] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/04/2022] [Indexed: 11/13/2022] Open
Abstract
The kinase mTOR is a signaling hub for pathways that regulate cellular growth. In neurons, the subcellular localization of mTOR takes on increased significance. Here, we review findings on the localization of mTOR in axons and offer a perspective on how these may impact our understanding of nervous system development, function, and disease. We propose a model where mTOR accumulates in local foci we term mTOR outposts, which can be found in processes distant from a neuron’s cell body. In this model, pathways that funnel through mTOR are gated by local outposts to spatially select and amplify local signaling. The presence or absence of mTOR outposts in a segment of axon or dendrite may determine whether regional mTOR-dependent signals, such as nutrient and growth factor signaling, register toward neuron-wide responses. In this perspective, we present the emerging evidence for mTOR outposts in neurons, their putative roles as spatial gatekeepers of signaling inputs, and the implications of the mTOR outpost model for neuronal protein synthesis, signal transduction, and synaptic plasticity.
Collapse
|
12
|
Vargas JNS, Sleigh JN, Schiavo G. Coupling axonal mRNA transport and local translation to organelle maintenance and function. Curr Opin Cell Biol 2022; 74:97-103. [PMID: 35220080 PMCID: PMC10477965 DOI: 10.1016/j.ceb.2022.01.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/14/2022] [Accepted: 01/22/2022] [Indexed: 12/14/2022]
Abstract
Neuronal homeostasis requires the transport of various organelles to distal compartments and defects in this process lead to neurological disorders. Although several mechanisms for the delivery of organelles to axons and dendrites have been elucidated, exactly how this process is orchestrated is not well-understood. In this review, we discuss the recent literature supporting a novel paradigm - the co-shuttling of mRNAs with different membrane-bound organelles. This model postulates that the tethering of ribonucleoprotein complexes to endolysosomes and mitochondria allows for the spatiotemporal coupling of organelle transport and the delivery of transcripts to axons. Subcellular translation of these "hitchhiking" transcripts may thus provide a proximal source of proteins required for the maintenance and function of organelles in axons.
Collapse
Affiliation(s)
- Jose Norberto S Vargas
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK; UCL Queen Square Motor Neuron Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK; UK Dementia Research Institute, University College London, London, UK
| | - James N Sleigh
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK; UCL Queen Square Motor Neuron Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK; UK Dementia Research Institute, University College London, London, UK
| | - Giampietro Schiavo
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK; UCL Queen Square Motor Neuron Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK; UK Dementia Research Institute, University College London, London, UK.
| |
Collapse
|
13
|
Wu SY, Chen YL, Lee YR, Lin CF, Lan SH, Lan KY, Chu ML, Lin PW, Yang ZL, Chen YH, Wang WH, Liu HS. The Autophagosomes Containing Dengue Virus Proteins and Full-Length Genomic RNA Are Infectious. Viruses 2021; 13:v13102034. [PMID: 34696464 PMCID: PMC8540618 DOI: 10.3390/v13102034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/30/2021] [Accepted: 10/04/2021] [Indexed: 02/04/2023] Open
Abstract
Autophagic machinery is involved in selective and non-selective recruitment as well as degradation or exocytosis of cargoes, including pathogens. Dengue virus (DENV) infection induces autophagy that enhances virus replication and vesicle release to evade immune system surveillance. This study reveals that DENV2 induces autophagy in lung and liver cancer cells and showed that DENV2 capsid, envelope, NS1, NS3, NS4B and host cell proinflammatory high mobility group box 1 (HMGB1) proteins associated with autophagosomes which were purified by gradient centrifugation. Capsid, NS1 and NS3 proteins showing high colocalization with LC3 protein in the cytoplasm of the infected cells were detected in the purified double-membrane autophagosome by immunogold labeling under transmission electron microscopy. In DENV infected cells, the levels of capsid, envelope, NS1 and HMGB1 proteins are not significantly changed compared to the dramatic accumulation of LC3-II and p62/SQSTM1 proteins when autophagic degradation was blocked by chloroquine, indicating that these proteins are not regulated by autophagic degradation machinery. We further demonstrated that purified autophagosomes were infectious when co-cultured with uninfected cells. Notably, these infectious autophagosomes contain DENV2 proteins, negative-strand and full-length genomic RNAs, but no viral particles. It is possible that the infectivity of the autophagosome originates from the full-length DENV RNA. Moreover, we reveal that DENV2 promotes HMGB1 exocytosis partially through secretory autophagy. In conclusion, we are the first to report that DENV2-induced double-membrane autophagosomes containing viral proteins and full-length RNAs are infectious and not undergoing autophagic degradation. Our novel finding warrants further validation of whether these intracellular vesicles undergo exocytosis to become infectious autophagic vesicles.
Collapse
Affiliation(s)
- Shan-Ying Wu
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (S.-Y.W.); (C.-F.L.)
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Yu-Lun Chen
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan;
| | - Ying-Ray Lee
- Department of Microbiology and Immunology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Chiou-Feng Lin
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; (S.-Y.W.); (C.-F.L.)
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Core Laboratory of Immune Monitoring, Office of Research & Development, Taipei Medical University, Taipei 110, Taiwan
- Center of Infectious Diseases and Signaling Research, National Cheng Kung University, Tainan 701, Taiwan
| | - Sheng-Hui Lan
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (S.-H.L.); (K.-Y.L.); (Z.-L.Y.)
| | - Kai-Ying Lan
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (S.-H.L.); (K.-Y.L.); (Z.-L.Y.)
| | - Man-Ling Chu
- Center for Cancer Research, Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (M.-L.C.); (P.-W.L.)
| | - Pei-Wen Lin
- Center for Cancer Research, Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (M.-L.C.); (P.-W.L.)
| | - Zong-Lin Yang
- Department of Life Sciences and Institute of Genome Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; (S.-H.L.); (K.-Y.L.); (Z.-L.Y.)
| | - Yen-Hsu Chen
- Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-H.C.); (W.-H.W.)
- Sepsis Research Center, Center of Tropical Medicine and Infectious Diseases, Graduate Institute of Medicine, School of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Biological Science and Technology, College of Biological Science and Technology, National Yang Ming Chiao Tung University, HsinChu 300, Taiwan
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 804, Taiwan
| | - Wen-Hung Wang
- Division of Infectious Disease, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (Y.-H.C.); (W.-H.W.)
- Sepsis Research Center, Center of Tropical Medicine and Infectious Diseases, Graduate Institute of Medicine, School of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Hsiao-Sheng Liu
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan;
- Center for Cancer Research, Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (M.-L.C.); (P.-W.L.)
- Master of Science Program in Tropical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Correspondence: ; Tel.: +886-7-3121101 (ext. 2378); Fax: +886-7-3222461
| |
Collapse
|
14
|
Wang W, Bian J, Sun Y, Li Z. The new fate of internalized membrane receptors: Internalized activation. Pharmacol Ther 2021; 233:108018. [PMID: 34626676 DOI: 10.1016/j.pharmthera.2021.108018] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 10/02/2021] [Accepted: 10/04/2021] [Indexed: 12/23/2022]
Abstract
Classically, the fate of internalized membrane receptors includes receptor degradation and receptor recycling. However, recent findings have begun to challenge these views. Much research demonstrated that many internalized membrane receptors can trigger distinct signal activation rather than being desensitized inside the cell. Here, we introduce the concept of "internalized activation" which not only represents a new mode of receptor activation, but also endows the new fate for receptor internalization (from death to life). The new activation mode and fate of membrane receptor are ubiquitous and have unique theoretical significance. We systematically put forward the features, process, and regulation of "internalized activation" and its significance in signal transduction and diseases. "Internalized activation" will provide a completely new understanding for the theory of receptor activation, internalization and novel drug targets for precision medicine.
Collapse
Affiliation(s)
- Wenjing Wang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing Key Laboratory of Cardiovascular Receptors Research, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing 100191, China
| | - Jingwei Bian
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing Key Laboratory of Cardiovascular Receptors Research, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing 100191, China
| | - Yang Sun
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing Key Laboratory of Cardiovascular Receptors Research, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing 100191, China
| | - Zijian Li
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing Key Laboratory of Cardiovascular Receptors Research, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing 100191, China; Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China.
| |
Collapse
|
15
|
Revising Endosomal Trafficking under Insulin Receptor Activation. Int J Mol Sci 2021; 22:ijms22136978. [PMID: 34209489 PMCID: PMC8268289 DOI: 10.3390/ijms22136978] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/25/2021] [Accepted: 06/27/2021] [Indexed: 02/06/2023] Open
Abstract
The endocytosis of ligand-bound receptors and their eventual recycling to the plasma membrane (PM) are processes that have an influence on signalling activity and therefore on many cell functions, including migration and proliferation. Like other tyrosine kinase receptors (TKR), the insulin receptor (INSR) has been shown to be endocytosed by clathrin-dependent and -independent mechanisms. Once at the early endosome (EE), the sorting of the receptor, either to the late endosome (LE) for degradation or back to the PM through slow or fast recycling pathways, will determine the intensity and duration of insulin effects. Both the endocytic and the endosomic pathways are regulated by many proteins, the Arf and Rab families of small GTPases being some of the most relevant. Here, we argue for a specific role for the slow recycling route, whilst we review the main molecular mechanisms involved in INSR endocytosis, sorting and recycling, as well as their possible role in cell functions.
Collapse
|
16
|
Jin J, Kim C, Xia Q, Gould TM, Cao W, Zhang H, Li X, Weiskopf D, Grifoni A, Sette A, Weyand CM, Goronzy JJ. Activation of mTORC1 at late endosomes misdirects T cell fate decision in older individuals. Sci Immunol 2021; 6:eabg0791. [PMID: 34145066 PMCID: PMC8422387 DOI: 10.1126/sciimmunol.abg0791] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Accepted: 05/18/2021] [Indexed: 12/11/2022]
Abstract
The nutrient-sensing mammalian target of rapamycin (mTOR) is integral to cell fate decisions after T cell activation. Sustained mTORC1 activity favors the generation of terminally differentiated effector T cells instead of follicular helper and memory T cells. This is particularly pertinent for T cell responses of older adults who have sustained mTORC1 activation despite dysfunctional lysosomes. Here, we show that lysosome-deficient T cells rely on late endosomes rather than lysosomes as an mTORC1 activation platform, where mTORC1 is activated by sensing cytosolic amino acids. T cells from older adults have an increased expression of the plasma membrane leucine transporter SLC7A5 to provide a cytosolic amino acid source. Hence, SLC7A5 and VPS39 deficiency (a member of the HOPS complex promoting early to late endosome conversion) substantially reduced mTORC1 activities in T cells from older but not young individuals. Late endosomal mTORC1 is independent of the negative-feedback loop involving mTORC1-induced inactivation of the transcription factor TFEB that controls expression of lysosomal genes. The resulting sustained mTORC1 activation impaired lysosome function and prevented lysosomal degradation of PD-1 in CD4+ T cells from older adults, thereby inhibiting their proliferative responses. VPS39 silencing of human T cells improved their expansion to pertussis and to SARS-CoV-2 peptides in vitro. Furthermore, adoptive transfer of CD4+ Vps39-deficient LCMV-specific SMARTA cells improved germinal center responses, CD8+ memory T cell generation, and recall responses to infection. Thus, curtailing late endosomal mTORC1 activity is a promising strategy to enhance T cell immunity.
Collapse
Affiliation(s)
- Jun Jin
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, United States
| | - Chulwoo Kim
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, United States
| | - Qiong Xia
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Timothy M Gould
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, United States
| | - Wenqiang Cao
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, United States
| | - Huimin Zhang
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, United States
| | - Xuanying Li
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, United States
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA, USA
| | - Cornelia M Weyand
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, United States
| | - Jorg J Goronzy
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, USA.
- Department of Medicine, Palo Alto Veterans Administration Healthcare System, Palo Alto, CA, United States
| |
Collapse
|
17
|
Meng D, Yang Q, Melick CH, Park BC, Hsieh T, Curukovic A, Jeong M, Zhang J, James NG, Jewell JL. ArfGAP1 inhibits mTORC1 lysosomal localization and activation. EMBO J 2021; 40:e106412. [PMID: 33988249 PMCID: PMC8204869 DOI: 10.15252/embj.2020106412] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 03/14/2021] [Accepted: 03/24/2021] [Indexed: 12/11/2022] Open
Abstract
The mammalian target of rapamycin complex 1 (mTORC1) integrates nutrients, growth factors, stress, and energy status to regulate cell growth and metabolism. Amino acids promote mTORC1 lysosomal localization and subsequent activation. However, the subcellular location or interacting proteins of mTORC1 under amino acid-deficient conditions is not completely understood. Here, we identify ADP-ribosylation factor GTPase-activating protein 1 (ArfGAP1) as a crucial regulator of mTORC1. ArfGAP1 interacts with mTORC1 in the absence of amino acids and inhibits mTORC1 lysosomal localization and activation. Mechanistically, the membrane curvature-sensing amphipathic lipid packing sensor (ALPS) motifs that bind to vesicle membranes are crucial for ArfGAP1 to interact with and regulate mTORC1 activity. Importantly, ArfGAP1 represses cell growth through mTORC1 and is an independent prognostic factor for the overall survival of pancreatic cancer patients. Our study identifies ArfGAP1 as a critical regulator of mTORC1 that functions by preventing the lysosomal transport and activation of mTORC1, with potential for cancer therapeutics.
Collapse
Affiliation(s)
- Delong Meng
- Department of Molecular BiologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Harold C. Simmons Comprehensive Cancer CenterUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Hamon Center for Regenerative Science and MedicineUniversity of Texas Southwestern Medical CenterDallasTXUSA
| | - Qianmei Yang
- Department of Molecular BiologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Harold C. Simmons Comprehensive Cancer CenterUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Hamon Center for Regenerative Science and MedicineUniversity of Texas Southwestern Medical CenterDallasTXUSA
| | - Chase H Melick
- Department of Molecular BiologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Harold C. Simmons Comprehensive Cancer CenterUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Hamon Center for Regenerative Science and MedicineUniversity of Texas Southwestern Medical CenterDallasTXUSA
| | - Brenden C Park
- Department of Molecular BiologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Harold C. Simmons Comprehensive Cancer CenterUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Hamon Center for Regenerative Science and MedicineUniversity of Texas Southwestern Medical CenterDallasTXUSA
| | - Ting‐Sung Hsieh
- Department of Molecular BiologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Harold C. Simmons Comprehensive Cancer CenterUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Hamon Center for Regenerative Science and MedicineUniversity of Texas Southwestern Medical CenterDallasTXUSA
| | - Adna Curukovic
- Department of Molecular BiologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Harold C. Simmons Comprehensive Cancer CenterUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Hamon Center for Regenerative Science and MedicineUniversity of Texas Southwestern Medical CenterDallasTXUSA
| | - Mi‐Hyeon Jeong
- Department of Molecular BiologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Harold C. Simmons Comprehensive Cancer CenterUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Hamon Center for Regenerative Science and MedicineUniversity of Texas Southwestern Medical CenterDallasTXUSA
| | - Junmei Zhang
- Department of Molecular BiologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
| | - Nicholas G James
- Department of Cell and Molecular BiologyJohn A. Burns School of MedicineUniversity of HawaiiHonoluluHIUSA
| | - Jenna L Jewell
- Department of Molecular BiologyUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Harold C. Simmons Comprehensive Cancer CenterUniversity of Texas Southwestern Medical CenterDallasTXUSA
- Hamon Center for Regenerative Science and MedicineUniversity of Texas Southwestern Medical CenterDallasTXUSA
| |
Collapse
|
18
|
Jeckel P, Kriebel M, Volkmer H. Autism Spectrum Disorder Risk Factor Met Regulates the Organization of Inhibitory Synapses. Front Mol Neurosci 2021; 14:659856. [PMID: 34054427 PMCID: PMC8155383 DOI: 10.3389/fnmol.2021.659856] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/09/2021] [Indexed: 12/27/2022] Open
Abstract
A common hypothesis explains autism spectrum disorder (ASD) as a neurodevelopmental disorder linked to excitatory/inhibitory (E/I) imbalance in neuronal network connectivity. Mutation of genes including Met and downstream signaling components, e.g., PTEN, Tsc2 and, Rheb are involved in the control of synapse formation and stabilization and were all considered as risk genes for ASD. While the impact of Met on glutamatergic synapses was widely appreciated, its contribution to the stability of inhibitory, GABAergic synapses is poorly understood. The stabilization of GABAergic synapses depends on clustering of the postsynaptic scaffolding protein gephyrin. Here, we show in vivo and in vitro that Met is necessary and sufficient for the stabilization of GABAergic synapses via induction of gephyrin clustering. Likewise, we provide evidence for Met-dependent gephyrin clustering via activation of mTOR. Our results support the notion that deficient GABAergic signaling represents a pathomechanism for ASD.
Collapse
Affiliation(s)
- Pauline Jeckel
- Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Martin Kriebel
- Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Hansjürgen Volkmer
- Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| |
Collapse
|
19
|
De D, Mukherjee I, Guha S, Paidi RK, Chakrabarti S, Biswas SC, Bhattacharyya SN. Rheb-mTOR activation rescues Aβ-induced cognitive impairment and memory function by restoring miR-146 activity in glial cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 24:868-887. [PMID: 34094708 PMCID: PMC8141608 DOI: 10.1016/j.omtn.2021.04.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 04/09/2021] [Indexed: 12/22/2022]
Abstract
Deposition of amyloid beta plaques in adult rat or human brain is associated with increased production of proinflammatory cytokines by associated glial cells that are responsible for degeneration of the diseased tissue. The expression of these cytokines is usually under check and is controlled at the post-transcriptional level via several microRNAs. Computational analysis of gene expression profiles of cortical regions of Alzheimer’s disease patients’ brain suggests ineffective target cytokine mRNA suppression by existing micro-ribonucleoproteins (miRNPs) in diseased brain. Exploring the mechanism of amyloid beta-induced cytokine expression, we have identified how the inactivation of the repressive miR-146 miRNPs causes increased production of cytokines in amyloid beta-exposed glial cells. In exploration of the cause of miRNP inactivation, we have noted amyloid beta oligomer-induced sequestration of the mTORC1 complex to early endosomes that results in decreased Ago2 phosphorylation, limited Ago2-miRNA uncoupling, and retarded Ago2-cytokine mRNA interaction in rat astrocytes. Interestingly, constitutive activation of mTORC1 by Rheb activator restricts proinflammatory cytokine production by reactivating miR-146 miRNPs in amyloid beta-exposed glial cells to rescue the disease phenotype in the in vivo rat model of Alzheimer’s disease.
Collapse
Affiliation(s)
- Dipayan De
- RNA Biology Research Laboratory, Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Ishita Mukherjee
- Structural Biology and Bio-informatics Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Subhalakshmi Guha
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Ramesh Kumar Paidi
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Saikat Chakrabarti
- Structural Biology and Bio-informatics Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Subhas C Biswas
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| | - Suvendra N Bhattacharyya
- RNA Biology Research Laboratory, Molecular Genetics Division, CSIR-Indian Institute of Chemical Biology, Kolkata 700032, India
| |
Collapse
|
20
|
Takahara T, Amemiya Y, Sugiyama R, Maki M, Shibata H. Amino acid-dependent control of mTORC1 signaling: a variety of regulatory modes. J Biomed Sci 2020; 27:87. [PMID: 32799865 PMCID: PMC7429791 DOI: 10.1186/s12929-020-00679-2] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 07/30/2020] [Indexed: 01/10/2023] Open
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) is an essential regulator of cell growth and metabolism through the modulation of protein and lipid synthesis, lysosome biogenesis, and autophagy. The activity of mTORC1 is dynamically regulated by several environmental cues, including amino acid availability, growth factors, energy levels, and stresses, to coordinate cellular status with environmental conditions. Dysregulation of mTORC1 activity is closely associated with various diseases, including diabetes, cancer, and neurodegenerative disorders. The discovery of Rag GTPases has greatly expanded our understanding of the regulation of mTORC1 activity by amino acids, especially leucine and arginine. In addition to Rag GTPases, other factors that also contribute to the modulation of mTORC1 activity have been identified. In this review, we discuss the mechanisms of regulation of mTORC1 activity by particular amino acids.
Collapse
Affiliation(s)
- Terunao Takahara
- Department of Applied Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Aichi, 464-8601, Japan.
| | - Yuna Amemiya
- Department of Applied Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Aichi, 464-8601, Japan
| | - Risa Sugiyama
- Department of Applied Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Aichi, 464-8601, Japan
| | - Masatoshi Maki
- Department of Applied Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Aichi, 464-8601, Japan
| | - Hideki Shibata
- Department of Applied Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Furo-cho, Chikusa-ku, Nagoya, Aichi, 464-8601, Japan
| |
Collapse
|
21
|
Drizyte-Miller K, Chen J, Cao H, Schott MB, McNiven MA. The small GTPase Rab32 resides on lysosomes to regulate mTORC1 signaling. J Cell Sci 2020; 133:jcs236661. [PMID: 32295849 PMCID: PMC7295596 DOI: 10.1242/jcs.236661] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 04/01/2020] [Indexed: 12/24/2022] Open
Abstract
Epithelial cells, such as liver-resident hepatocytes, rely heavily on the Rab family of small GTPases to perform membrane trafficking events that dictate cell physiology and metabolism. Not surprisingly, disruption of several Rab proteins can manifest in metabolic diseases or cancer. Rab32 is expressed in many secretory epithelial cells but its role in cellular metabolism is virtually unknown. In this study, we find that Rab32 associates with lysosomes and regulates proliferation and cell size of Hep3B hepatoma and HeLa cells. Specifically, we identify that Rab32 supports the mechanistic target of rapamycin complex 1 (mTORC1) signaling under basal and amino acid-stimulated conditions. Consistent with inhibited mTORC1, an increase in nuclear TFEB localization and lysosome biogenesis is also observed in Rab32-depleted cells. Finally, we find that Rab32 interacts with mTOR kinase, and that loss of Rab32 reduces the association of mTOR and mTORC1 pathway proteins with lysosomes, suggesting that Rab32 regulates lysosomal mTOR trafficking. In summary, these findings suggest that Rab32 functions as a novel regulator of cellular metabolism through supporting mTORC1 signaling.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Kristina Drizyte-Miller
- Biochemistry and Molecular Biology Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905, USA
| | - Jing Chen
- Center for Basic Research in Digestive Diseases, Division of Gastroenterology and Hepatology, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905, USA
| | - Hong Cao
- Center for Basic Research in Digestive Diseases, Division of Gastroenterology and Hepatology, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905, USA
| | - Micah B Schott
- Center for Basic Research in Digestive Diseases, Division of Gastroenterology and Hepatology, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905, USA
| | - Mark A McNiven
- Biochemistry and Molecular Biology Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905, USA
- Center for Basic Research in Digestive Diseases, Division of Gastroenterology and Hepatology, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, 200 1st Street SW, Rochester, MN 55905, USA
| |
Collapse
|
22
|
Palušová V, Renzová T, Verlande A, Vaclová T, Medková M, Cetlová L, Sedláčková M, Hříbková H, Slaninová I, Krutá M, Rotrekl V, Uhlířová H, Křížová A, Chmelík R, Veselý P, Krafčíková M, Trantírek L, Schink KO, Uldrijan S. Dual Targeting of BRAF and mTOR Signaling in Melanoma Cells with Pyridinyl Imidazole Compounds. Cancers (Basel) 2020; 12:cancers12061516. [PMID: 32531927 PMCID: PMC7352453 DOI: 10.3390/cancers12061516] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 06/04/2020] [Accepted: 06/05/2020] [Indexed: 12/19/2022] Open
Abstract
BRAF inhibitors can delay the progression of metastatic melanoma, but resistance usually emerges, leading to relapse. Drugs simultaneously targeting two or more pathways essential for cancer growth could slow or prevent the development of resistant clones. Here, we identified pyridinyl imidazole compounds SB202190, SB203580, and SB590885 as dual inhibitors of critical proliferative pathways in human melanoma cells bearing the V600E activating mutation of BRAF kinase. We found that the drugs simultaneously disrupt the BRAF V600E-driven extracellular signal-regulated kinase (ERK) mitogen-activated protein kinase (MAPK) activity and the mechanistic target of rapamycin complex 1 (mTORC1) signaling in melanoma cells. Pyridinyl imidazole compounds directly inhibit BRAF V600E kinase. Moreover, they interfere with the endolysosomal compartment, promoting the accumulation of large acidic vacuole-like vesicles and dynamic changes in mTOR signaling. A transient increase in mTORC1 activity is followed by the enrichment of the Ragulator complex protein p18/LAMTOR1 at contact sites of large vesicles and delocalization of mTOR from the lysosomes. The induced disruption of the endolysosomal pathway not only disrupts mTORC1 signaling, but also renders melanoma cells sensitive to endoplasmic reticulum (ER) stress. Our findings identify new activities of pharmacologically relevant small molecule compounds and provide a biological rationale for the development of anti-melanoma therapeutics based on the pyridinyl imidazole core.
Collapse
Affiliation(s)
- Veronika Palušová
- Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic; (V.P.); (T.R.); (A.V.); (T.V.); (M.M.); (L.C.); (M.S.); (H.H.); (I.S.); (M.K.); (V.R.)
- International Clinical Research Center, St. Anne’s University Hospital Brno, Pekařská 664/53, 656 91 Brno, Czech Republic
| | - Tereza Renzová
- Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic; (V.P.); (T.R.); (A.V.); (T.V.); (M.M.); (L.C.); (M.S.); (H.H.); (I.S.); (M.K.); (V.R.)
| | - Amandine Verlande
- Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic; (V.P.); (T.R.); (A.V.); (T.V.); (M.M.); (L.C.); (M.S.); (H.H.); (I.S.); (M.K.); (V.R.)
| | - Tereza Vaclová
- Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic; (V.P.); (T.R.); (A.V.); (T.V.); (M.M.); (L.C.); (M.S.); (H.H.); (I.S.); (M.K.); (V.R.)
| | - Michaela Medková
- Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic; (V.P.); (T.R.); (A.V.); (T.V.); (M.M.); (L.C.); (M.S.); (H.H.); (I.S.); (M.K.); (V.R.)
| | - Linda Cetlová
- Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic; (V.P.); (T.R.); (A.V.); (T.V.); (M.M.); (L.C.); (M.S.); (H.H.); (I.S.); (M.K.); (V.R.)
| | - Miroslava Sedláčková
- Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic; (V.P.); (T.R.); (A.V.); (T.V.); (M.M.); (L.C.); (M.S.); (H.H.); (I.S.); (M.K.); (V.R.)
| | - Hana Hříbková
- Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic; (V.P.); (T.R.); (A.V.); (T.V.); (M.M.); (L.C.); (M.S.); (H.H.); (I.S.); (M.K.); (V.R.)
| | - Iva Slaninová
- Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic; (V.P.); (T.R.); (A.V.); (T.V.); (M.M.); (L.C.); (M.S.); (H.H.); (I.S.); (M.K.); (V.R.)
| | - Miriama Krutá
- Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic; (V.P.); (T.R.); (A.V.); (T.V.); (M.M.); (L.C.); (M.S.); (H.H.); (I.S.); (M.K.); (V.R.)
| | - Vladimír Rotrekl
- Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic; (V.P.); (T.R.); (A.V.); (T.V.); (M.M.); (L.C.); (M.S.); (H.H.); (I.S.); (M.K.); (V.R.)
- International Clinical Research Center, St. Anne’s University Hospital Brno, Pekařská 664/53, 656 91 Brno, Czech Republic
| | - Hana Uhlířová
- Institute of Physical Engineering, Faculty of Mechanical Engineering, Brno University of Technology, Technická 2896/2, 616 69 Brno, Czech Republic; (H.U.); (R.C.)
- CEITEC—Central European Institute of Technology, Brno University of Technology, Purkyňova 656/123, 612 00 Brno, Czech Republic; (A.K.); (P.V.)
| | - Aneta Křížová
- CEITEC—Central European Institute of Technology, Brno University of Technology, Purkyňova 656/123, 612 00 Brno, Czech Republic; (A.K.); (P.V.)
| | - Radim Chmelík
- Institute of Physical Engineering, Faculty of Mechanical Engineering, Brno University of Technology, Technická 2896/2, 616 69 Brno, Czech Republic; (H.U.); (R.C.)
- CEITEC—Central European Institute of Technology, Brno University of Technology, Purkyňova 656/123, 612 00 Brno, Czech Republic; (A.K.); (P.V.)
| | - Pavel Veselý
- CEITEC—Central European Institute of Technology, Brno University of Technology, Purkyňova 656/123, 612 00 Brno, Czech Republic; (A.K.); (P.V.)
| | - Michaela Krafčíková
- National Centre for Biomolecular Research, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic;
| | - Lukáš Trantírek
- CEITEC—Central European Institute of Technology, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic;
| | - Kay Oliver Schink
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, University of Oslo, Montebello, N-0379 Oslo, Norway;
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, N-0379 Oslo, Norway
| | - Stjepan Uldrijan
- Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00 Brno, Czech Republic; (V.P.); (T.R.); (A.V.); (T.V.); (M.M.); (L.C.); (M.S.); (H.H.); (I.S.); (M.K.); (V.R.)
- International Clinical Research Center, St. Anne’s University Hospital Brno, Pekařská 664/53, 656 91 Brno, Czech Republic
- Correspondence:
| |
Collapse
|
23
|
Carroll B. Spatial regulation of mTORC1 signalling: Beyond the Rag GTPases. Semin Cell Dev Biol 2020; 107:103-111. [PMID: 32122730 DOI: 10.1016/j.semcdb.2020.02.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/17/2020] [Accepted: 02/20/2020] [Indexed: 12/15/2022]
Abstract
The mechanistic (or mammalian) Target of Rapamycin Complex 1 (mTORC1) is a central regulator of cell growth and metabolism. By integrating mitogenic signals, mTORC1-dependent phosphorylation of substrates dictates the balance between anabolic, pro-growth and catabolic, recycling processes in the cell. The discovery that amino acids activate mTORC1 by promoting its translocation to the lysosome was a fundamental advance in the understanding of mTORC1 signalling. It has since become clear that the lysosome-cytoplasm shuttling of mTORC1 represents just one layer of spatial control of this signalling pathway. This review will focus on exploring the subcellular localisation of mTORC1 and its regulators to multiple sites within the cell. We will discuss how these spatially distinct regions such as endoplasmic reticulum, plasma membrane and the endosomal pathway co-operate to transduce nutrient availability to mTORC1, allowing for tight control of cell growth.
Collapse
Affiliation(s)
- Bernadette Carroll
- School of Biochemistry, Biomedical Sciences Building, University Walk, Bristol, BS8, United Kingdom.
| |
Collapse
|
24
|
Kvainickas A, Nägele H, Qi W, Dokládal L, Jimenez-Orgaz A, Stehl L, Gangurde D, Zhao Q, Hu Z, Dengjel J, De Virgilio C, Baumeister R, Steinberg F. Retromer and TBC1D5 maintain late endosomal RAB7 domains to enable amino acid-induced mTORC1 signaling. J Cell Biol 2019; 218:3019-3038. [PMID: 31431476 PMCID: PMC6719456 DOI: 10.1083/jcb.201812110] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 05/30/2019] [Accepted: 07/08/2019] [Indexed: 02/07/2023] Open
Abstract
Retromer is an evolutionarily conserved multiprotein complex that orchestrates the endocytic recycling of integral membrane proteins. Here, we demonstrate that retromer is also required to maintain lysosomal amino acid signaling through mTORC1 across species. Without retromer, amino acids no longer stimulate mTORC1 translocation to the lysosomal membrane, which leads to a loss of mTORC1 activity and increased induction of autophagy. Mechanistically, we show that its effect on mTORC1 activity is not linked to retromer's role in the recycling of transmembrane proteins. Instead, retromer cooperates with the RAB7-GAP TBC1D5 to restrict late endosomal RAB7 into microdomains that are spatially separated from the amino acid-sensing domains. Upon loss of retromer, RAB7 expands into the ragulator-decorated amino acid-sensing domains and interferes with RAG-GTPase and mTORC1 recruitment. Depletion of retromer in Caenorhabditis elegans reduces mTORC1 signaling and extends the lifespan of the worms, confirming an evolutionarily conserved and unexpected role for retromer in the regulation of mTORC1 activity and longevity.
Collapse
Affiliation(s)
- Arunas Kvainickas
- Center for Biological Systems Analysis, University of Freiburg, Freiburg, Germany
| | - Heike Nägele
- Center for Biological Systems Analysis, University of Freiburg, Freiburg, Germany
| | - Wenjing Qi
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Ladislav Dokládal
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Ana Jimenez-Orgaz
- Center for Biological Systems Analysis, University of Freiburg, Freiburg, Germany
| | - Luca Stehl
- Center for Biological Systems Analysis, University of Freiburg, Freiburg, Germany
| | - Dipak Gangurde
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Qian Zhao
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Zehan Hu
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Jörn Dengjel
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | | | - Ralf Baumeister
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Florian Steinberg
- Center for Biological Systems Analysis, University of Freiburg, Freiburg, Germany
| |
Collapse
|
25
|
Bresnick AR, Backer JM. PI3Kβ-A Versatile Transducer for GPCR, RTK, and Small GTPase Signaling. Endocrinology 2019; 160:536-555. [PMID: 30601996 PMCID: PMC6375709 DOI: 10.1210/en.2018-00843] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 12/20/2018] [Indexed: 12/17/2022]
Abstract
The phosphoinositide 3-kinase (PI3K) family includes eight distinct catalytic subunits and seven regulatory subunits. Only two PI3Ks are directly regulated downstream from G protein-coupled receptors (GPCRs): the class I enzymes PI3Kβ and PI3Kγ. Both enzymes produce phosphatidylinositol 3,4,5-trisposphate in vivo and are regulated by both heterotrimeric G proteins and small GTPases from the Ras or Rho families. However, PI3Kβ is also regulated by direct interactions with receptor tyrosine kinases (RTKs) and their tyrosine phosphorylated substrates, and similar to the class II and III PI3Ks, it binds activated Rab5. The unusually complex regulation of PI3Kβ by small and trimeric G proteins and RTKs leads to a rich landscape of signaling responses at the cellular and organismic levels. This review focuses first on the regulation of PI3Kβ activity in vitro and in cells, and then summarizes the biology of PI3Kβ signaling in distinct tissues and in human disease.
Collapse
Affiliation(s)
- Anne R Bresnick
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York
| | - Jonathan M Backer
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
26
|
Hodson N, Philp A. The Importance of mTOR Trafficking for Human Skeletal Muscle Translational Control. Exerc Sport Sci Rev 2019; 47:46-53. [PMID: 30334852 PMCID: PMC6310455 DOI: 10.1249/jes.0000000000000173] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
This review will critique cell, rodent, and human models of mTOR regulation to discuss why mTOR trafficking may represent a novel and physiologically relevant model of regulation in skeletal muscle. The mechanistic target of rapamycin (mTOR) is a central regulator of muscle protein synthesis, and its activation has long been attributed to its translocation to the lysosome. Here, we present a novel model of mTOR activation in skeletal muscle where the translocation of mTOR and the lysosome toward the cell membrane is a key process in mTOR activation.
Collapse
Affiliation(s)
- Nathan Hodson
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UK
| | - Andrew Philp
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, UK.,Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia
| |
Collapse
|
27
|
Bautista SJ, Boras I, Vissa A, Mecica N, Yip CM, Kim PK, Antonescu CN. mTOR complex 1 controls the nuclear localization and function of glycogen synthase kinase 3β. J Biol Chem 2018; 293:14723-14739. [PMID: 30061153 DOI: 10.1074/jbc.ra118.002800] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 07/19/2018] [Indexed: 01/08/2023] Open
Abstract
Glycogen synthase kinase 3β (GSK3β) phosphorylates and thereby regulates a wide range of protein substrates involved in diverse cellular functions. Some GSK3β substrates, such as c-Myc and Snail, are nuclear transcription factors, suggesting the possibility that GSK3β function is controlled through its nuclear localization. Here, using ARPE-19 and MDA-MB-231 human cell lines, we found that inhibition of mTOR complex 1 (mTORC1) leads to partial redistribution of GSK3β from the cytosol to the nucleus and to a GSK3β-dependent reduction of the levels of both c-Myc and Snail. mTORC1 is known to be controlled by metabolic cues, such as by AMP-activated protein kinase (AMPK) or amino acid abundance, and we observed here that AMPK activation or amino acid deprivation promotes GSK3β nuclear localization in an mTORC1-dependent manner. GSK3β was detected on several distinct endomembrane compartments, including lysosomes. Consistently, disruption of late endosomes/lysosomes through a perturbation of RAS oncogene family member 7 (Rab7) resulted in loss of GSK3β from lysosomes and in enhanced GSK3β nuclear localization as well as GSK3β-dependent reduction of c-Myc levels. These findings indicate that the nuclear localization and function of GSK3β is suppressed by mTORC1 and suggest a link between metabolic conditions sensed by mTORC1 and GSK3β-dependent regulation of transcriptional networks controlling cellular biomass production.
Collapse
Affiliation(s)
- Stephen J Bautista
- From the Department of Chemistry and Biology and Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario M5B 2K3
| | - Ivan Boras
- From the Department of Chemistry and Biology and Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario M5B 2K3
| | - Adriano Vissa
- the Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3E5.,the Program in Cell Biology, Hospital for Sick Children, Toronto, Ontario M5G 0A4
| | - Noa Mecica
- From the Department of Chemistry and Biology and Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario M5B 2K3
| | - Christopher M Yip
- the Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3E5.,the Department of Biochemistry, University of Toronto, Toronto, Ontario M5G 1X8, and.,the Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario M5S 3E5, Canada
| | - Peter K Kim
- the Program in Cell Biology, Hospital for Sick Children, Toronto, Ontario M5G 0A4.,the Department of Biochemistry, University of Toronto, Toronto, Ontario M5G 1X8, and
| | - Costin N Antonescu
- From the Department of Chemistry and Biology and Graduate Program in Molecular Science, Ryerson University, Toronto, Ontario M5B 2K3, .,the Keenan Research Centre for Biomedical Science of St. Michael's Hospital, Toronto, Ontario M5B 1W8
| |
Collapse
|
28
|
Naslavsky N, Caplan S. The enigmatic endosome - sorting the ins and outs of endocytic trafficking. J Cell Sci 2018; 131:131/13/jcs216499. [PMID: 29980602 DOI: 10.1242/jcs.216499] [Citation(s) in RCA: 233] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The early endosome (EE), also known as the sorting endosome (SE) is a crucial station for the sorting of cargoes, such as receptors and lipids, through the endocytic pathways. The term endosome relates to the receptacle-like nature of this organelle, to which endocytosed cargoes are funneled upon internalization from the plasma membrane. Having been delivered by the fusion of internalized vesicles with the EE or SE, cargo molecules are then sorted to a variety of endocytic pathways, including the endo-lysosomal pathway for degradation, direct or rapid recycling to the plasma membrane, and to a slower recycling pathway that involves a specialized form of endosome known as a recycling endosome (RE), often localized to the perinuclear endocytic recycling compartment (ERC). It is striking that 'the endosome', which plays such essential cellular roles, has managed to avoid a precise description, and its characteristics remain ambiguous and heterogeneous. Moreover, despite the rapid advances in scientific methodologies, including breakthroughs in light microscopy, overall, the endosome remains poorly defined. This Review will attempt to collate key characteristics of the different types of endosomes and provide a platform for discussion of this unique and fascinating collection of organelles. Moreover, under-developed, poorly understood and important open questions will be discussed.
Collapse
Affiliation(s)
- Naava Naslavsky
- The Department of Biochemistry and Molecular Biology, The University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Steve Caplan
- The Department of Biochemistry and Molecular Biology, The University of Nebraska Medical Center, Omaha, NE 68198, USA .,The Fred and Pamela Buffett Cancer Center, The University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
29
|
Ranadheera C, Coombs KM, Kobasa D. Comprehending a Killer: The Akt/mTOR Signaling Pathways Are Temporally High-Jacked by the Highly Pathogenic 1918 Influenza Virus. EBioMedicine 2018; 32:142-163. [PMID: 29866590 PMCID: PMC6021456 DOI: 10.1016/j.ebiom.2018.05.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 05/08/2018] [Accepted: 05/21/2018] [Indexed: 02/06/2023] Open
Abstract
Previous transcriptomic analyses suggested that the 1918 influenza A virus (IAV1918), one of the most devastating pandemic viruses of the 20th century, induces a dysfunctional cytokine storm and affects other innate immune response patterns. Because all viruses are obligate parasites that require host cells for replication, we globally assessed how IAV1918 induces host protein dysregulation. We performed quantitative mass spectrometry of IAV1918-infected cells to measure host protein dysregulation. Selected proteins were validated by immunoblotting and phosphorylation levels of members of the PI3K/AKT/mTOR pathway were assessed. Compared to mock-infected controls, >170 proteins in the IAV1918-infected cells were dysregulated. Proteins mapped to amino sugar metabolism, purine metabolism, steroid biosynthesis, transmembrane receptors, phosphatases and transcription regulation. Immunoblotting demonstrated that IAV1918 induced a slight up-regulation of the lamin B receptor whereas all other tested virus strains induced a significant down-regulation. IAV1918 also strongly induced Rab5b expression whereas all other tested viruses induced minor up-regulation or down-regulation. IAV1918 showed early reduced phosphorylation of PI3K/AKT/mTOR pathway members and was especially sensitive to rapamycin. These results suggest the 1918 strain requires mTORC1 activity in early replication events, and may explain the unique pathogenicity of this virus. Proteomic analyses of influenza 1918 virus-infected cells identified >170 dysregulated host proteins. Dysregulated proteins mapped to numerous important cellular pathways. 1918 virus infection showed prominent early reduced phosphorylation of PI3K/Akt/mTOR.
The 1918 influenza pandemic was one of the most devastating infectious disease events of the 20th century, resulting in 20–100 million deaths. Gene-based assays showed severe dysregulation of the host's cytokine responses, but little was known about global protein responses to virus infection. This work identifies unique and temporal alterations in phosphorylation of the PI3K/AKT/mTOR signaling pathway, which is important in determining cell death. This work paves the way for further research on how this pathway influences host mechanisms responsible for aiding virus replication and in determining levels and severity of influenza virus-induced patho
Collapse
Affiliation(s)
- Charlene Ranadheera
- Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba R3E 0J6, Canada; Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba R3E 3R2, Canada
| | - Kevin M Coombs
- Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba R3E 0J6, Canada; Manitoba Centre for Proteomics & Systems Biology, Room 799, 715 McDermot Avenue, Winnipeg, Manitoba R3E 3P4, Canada; Manitoba Institute of Child Health, John Buhler Research Centre, Room 513, 715 McDermot Avenue, Winnipeg, Manitoba R3E 3P4, Canada.
| | - Darwyn Kobasa
- Department of Medical Microbiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba R3E 0J6, Canada; Special Pathogens Program, National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba R3E 3R2, Canada.
| |
Collapse
|
30
|
Yoshida S, Pacitto R, Inoki K, Swanson J. Macropinocytosis, mTORC1 and cellular growth control. Cell Mol Life Sci 2018; 75:1227-1239. [PMID: 29119228 PMCID: PMC5843684 DOI: 10.1007/s00018-017-2710-y] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 10/31/2017] [Accepted: 11/03/2017] [Indexed: 12/15/2022]
Abstract
The growth and proliferation of metazoan cells are driven by cellular nutrient status and by extracellular growth factors. Growth factor receptors on cell surfaces initiate biochemical signals that increase anabolic metabolism and macropinocytosis, an actin-dependent endocytic process in which relatively large volumes of extracellular solutes and nutrients are internalized and delivered efficiently into lysosomes. Macropinocytosis is prominent in many kinds of cancer cells, and supports the growth of cells transformed by oncogenic K-Ras. Growth factor receptor signaling and the overall metabolic status of the cell are coordinated in the cytoplasm by the mechanistic target-of-rapamycin complex-1 (mTORC1), which positively regulates protein synthesis and negatively regulates molecular salvage pathways such as autophagy. mTORC1 is activated by two distinct Ras-related small GTPases, Rag and Rheb, which associate with lysosomal membranes inside the cell. Rag recruits mTORC1 to the lysosomal surface where Rheb directly binds to and activates mTORC1. Rag is activated by both lysosomal luminal and cytosolic amino acids; Rheb activation requires phosphoinositide 3-kinase, Akt, and the tuberous sclerosis complex-1/2. Signals for activation of Rag and Rheb converge at the lysosomal membrane, and several lines of evidence support the idea that growth factor-dependent endocytosis facilitates amino acid transfer into the lysosome leading to the activation of Rag. This review summarizes evidence that growth factor-stimulated macropinocytosis is essential for amino acid-dependent activation of mTORC1, and that increased solute accumulation by macropinocytosis in transformed cells supports unchecked cell growth.
Collapse
Affiliation(s)
- Sei Yoshida
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, 48109-5620, USA
| | - Regina Pacitto
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, 48109-5620, USA
| | - Ken Inoki
- Department of Integrative and Molecular Physiology and Internal Medicine, Life Sciences Institute, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Joel Swanson
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, 48109-5620, USA.
| |
Collapse
|
31
|
A Unique Homeostatic Signaling Pathway Links Synaptic Inactivity to Postsynaptic mTORC1. J Neurosci 2018; 38:2207-2225. [PMID: 29311141 DOI: 10.1523/jneurosci.1843-17.2017] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 11/23/2017] [Accepted: 12/27/2017] [Indexed: 12/31/2022] Open
Abstract
mTORC1-dependent translational control plays a key role in several enduring forms of synaptic plasticity such as long term potentiation (LTP) and mGluR-dependent long term depression. Recent evidence demonstrates an additional role in regulating synaptic homeostasis in response to inactivity, where dendritic mTORC1 serves to modulate presynaptic function via retrograde signaling. Presently, it is unclear whether LTP and homeostatic plasticity use a common route to mTORC1-dependent signaling or whether each engage mTORC1 through distinct pathways. Here, we report a unique signaling pathway that specifically couples homeostatic signaling to postsynaptic mTORC1 after loss of excitatory synaptic input. We find that AMPAR blockade, but not LTP-inducing stimulation, induces phospholipase D (PLD)-dependent synthesis of the lipid second messenger phosphatidic acid (PA) in rat cultured hippocampal neurons of either sex. Pharmacological blockade of PLD1/2 or pharmacogenetic disruption of PA interactions with mTOR eliminates mTORC1 signaling and presynaptic compensation driven by AMPAR blockade, but does not alter mTORC1 activation or functional changes during chemical LTP (cLTP). Overexpression of PLD1, but not PLD2, recapitulates both functional synaptic changes as well as signature cellular adaptations associated with homeostatic plasticity. Finally, transient application of exogenous PA is sufficient to drive rapid presynaptic compensation requiring mTORC1-dependent translation of BDNF in the postsynaptic compartment. These results thus define a unique homeostatic signaling pathway coupling mTORC1 activation to changes in excitatory synaptic drive. Our results further imply that more than one canonical mTORC1 activation pathway may be relevant for the design of novel therapeutic approaches against neurodevelopmental disorders associated with mTORC1 dysregulation.SIGNIFICANCE STATEMENT Homeostatic and Hebbian forms of synaptic plasticity are thought to play complementary roles in regulating neural circuit function, but we know little about how these forms of plasticity are distinguished at the single neuron level. Here, we define a signaling pathway that uniquely links mTORC1 with homeostatic signaling in neurons.
Collapse
|
32
|
Tebar F, Enrich C, Rentero C, Grewal T. GTPases Rac1 and Ras Signaling from Endosomes. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2018; 57:65-105. [PMID: 30097772 DOI: 10.1007/978-3-319-96704-2_3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The endocytic compartment is not only the functional continuity of the plasma membrane but consists of a diverse collection of intracellular heterogeneous complex structures that transport, amplify, sustain, and/or sort signaling molecules. Over the years, it has become evident that early, late, and recycling endosomes represent an interconnected vesicular-tubular network able to form signaling platforms that dynamically and efficiently translate extracellular signals into biological outcome. Cell activation, differentiation, migration, death, and survival are some of the endpoints of endosomal signaling. Hence, to understand the role of the endosomal system in signal transduction in space and time, it is therefore necessary to dissect and identify the plethora of decoders that are operational in the different steps along the endocytic pathway. In this chapter, we focus on the regulation of spatiotemporal signaling in cells, considering endosomes as central platforms, in which several small GTPases proteins of the Ras superfamily, in particular Ras and Rac1, actively participate to control cellular processes like proliferation and cell mobility.
Collapse
Affiliation(s)
- Francesc Tebar
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain.
| | - Carlos Enrich
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
| | - Carles Rentero
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
| | - Thomas Grewal
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
33
|
Wright LM, Carpinone EM, Bennett TL, Hondalus MK, Starai VJ. VapA of Rhodococcus equi binds phosphatidic acid. Mol Microbiol 2017; 107:428-444. [PMID: 29205554 DOI: 10.1111/mmi.13892] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 11/28/2017] [Accepted: 12/03/2017] [Indexed: 12/30/2022]
Abstract
Rhodococcus equi is a multihost, facultative intracellular bacterial pathogen that primarily causes pneumonia in foals less than six months in age and immunocompromised people. Previous studies determined that the major virulence determinant of R. equi is the surface bound virulence associated protein A (VapA). The presence of VapA inhibits the maturation of R. equi-containing phagosomes and promotes intracellular bacterial survival, as determined by the inability of vapA deletion mutants to replicate in host macrophages. While the mechanism of action of VapA remains elusive, we show that soluble recombinant VapA32-189 both rescues the intramacrophage replication defect of a wild type R. equi strain lacking the vapA gene and enhances the persistence of nonpathogenic Escherichia coli in macrophages. During macrophage infection, VapA was observed at both the bacterial surface and at the membrane of the host-derived R. equi containing vacuole, thus providing an opportunity for VapA to interact with host constituents and promote alterations in phagolysosomal function. In support of the observed host membrane binding activity of VapA, we also found that rVapA32-189 interacted specifically with liposomes containing phosphatidic acid in vitro. Collectively, these data demonstrate a lipid binding property of VapA, which may be required for its function during intracellular infection.
Collapse
Affiliation(s)
- Lindsay M Wright
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
| | - Emily M Carpinone
- Department of Microbiology, University of Georgia, Athens, GA 30602, USA
| | - Terry L Bennett
- Department of Microbiology, University of Georgia, Athens, GA 30602, USA
| | - Mary K Hondalus
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
| | - Vincent J Starai
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA.,Department of Microbiology, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
34
|
Patel S, Ashwanikumar N, Robinson E, DuRoss A, Sun C, Murphy-Benenato KE, Mihai C, Almarsson Ö, Sahay G. Boosting Intracellular Delivery of Lipid Nanoparticle-Encapsulated mRNA. NANO LETTERS 2017; 17:5711-5718. [PMID: 28836442 PMCID: PMC5623340 DOI: 10.1021/acs.nanolett.7b02664] [Citation(s) in RCA: 179] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Intracellular delivery of mRNA holds great potential for vaccine1-3 and therapeutic4 discovery and development. Despite increasing recognition of the utility of lipid-based nanoparticles (LNPs) for intracellular delivery of mRNA, particle engineering is hindered by insufficient understanding of endosomal escape, which is believed to be a main limiter of cytosolic availability and activity of the nucleic acid inside the cell. Using a series of CRISPR-based genetic perturbations of the lysosomal pathway, we have identified that late endosome/lysosome (LE/Ly) formation is essential for functional delivery of exogenously presented mRNA. Lysosomes provide a spatiotemporal hub to orchestrate mTOR signaling and are known to control cell proliferation, nutrient sensing, ribosomal biogenesis, and mRNA translation. Through modulation of the mTOR pathway we were able to enhance or inhibit LNP-mediated mRNA delivery. To further boost intracellular delivery of mRNA, we screened 212 bioactive lipid-like molecules that are either enriched in vesicular compartments or modulate cell signaling. Surprisingly, we have discovered that leukotriene-antagonists, clinically approved for treatment of asthma and other lung diseases, enhance intracellular mRNA delivery in vitro (over 3-fold, p < 0.005) and in vivo (over 2-fold, p < 0.005). Understanding LNP-mediated intracellular delivery will inspire the next generation of RNA therapeutics that have high potency and limited toxicity.
Collapse
Affiliation(s)
- Siddharth Patel
- Department of Pharmaceutical Sciences, College of Pharmacy, Collaborative Life Science Building, 2730 SW Moody Ave, Oregon State University, Portland, OR, 97201
| | - N Ashwanikumar
- Department of Pharmaceutical Sciences, College of Pharmacy, Collaborative Life Science Building, 2730 SW Moody Ave, Oregon State University, Portland, OR, 97201
| | - Emily Robinson
- Department of Pharmaceutical Sciences, College of Pharmacy, Collaborative Life Science Building, 2730 SW Moody Ave, Oregon State University, Portland, OR, 97201
| | - Allison DuRoss
- Department of Pharmaceutical Sciences, College of Pharmacy, Collaborative Life Science Building, 2730 SW Moody Ave, Oregon State University, Portland, OR, 97201
| | - Conroy Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, Collaborative Life Science Building, 2730 SW Moody Ave, Oregon State University, Portland, OR, 97201
- Department of Radiation Medicine, School of Medicine, 3181 S.W. Sam Jackson Park Road, Oregon Health Science University, Portland, OR, 97239
| | | | - Cosmin Mihai
- Moderna Therapeutics, 200 Technology Square, Cambridge, MA, 02139
| | - Örn Almarsson
- Moderna Therapeutics, 200 Technology Square, Cambridge, MA, 02139
| | - Gaurav Sahay
- Department of Pharmaceutical Sciences, College of Pharmacy, Collaborative Life Science Building, 2730 SW Moody Ave, Oregon State University, Portland, OR, 97201
- Department of Biomedical Engineering, Collaborative Life Science Building, 2730 SW Moody Ave, Oregon Health Science University, Portland, OR, 97201
| |
Collapse
|
35
|
Shibutani S, Okazaki H, Iwata H. Dynamin-dependent amino acid endocytosis activates mechanistic target of rapamycin complex 1 (mTORC1). J Biol Chem 2017; 292:18052-18061. [PMID: 28808055 DOI: 10.1074/jbc.m117.776443] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 08/03/2017] [Indexed: 11/06/2022] Open
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) is a master regulator of protein synthesis and potential target for modifying cellular metabolism in various conditions, including cancer and aging. mTORC1 activity is tightly regulated by the availability of extracellular amino acids, and previous studies have revealed that amino acids in the extracellular fluid are transported to the lysosomal lumen. There, amino acids induce recruitment of cytoplasmic mTORC1 to the lysosome by the Rag GTPases, followed by mTORC1 activation by the small GTPase Ras homolog enriched in brain (Rheb). However, how the extracellular amino acids reach the lysosomal lumen and activate mTORC1 remains unclear. Here, we show that amino acid uptake by dynamin-dependent endocytosis plays a critical role in mTORC1 activation. We found that mTORC1 is inactivated when endocytosis is inhibited by overexpression of a dominant-negative form of dynamin 2 or by pharmacological inhibition of dynamin or clathrin. Consistently, the recruitment of mTORC1 to the lysosome was suppressed by the dynamin inhibition. The activity and lysosomal recruitment of mTORC1 were rescued by increasing intracellular amino acids via cycloheximide exposure or by Rag overexpression, indicating that amino acid deprivation is the main cause of mTORC1 inactivation via the dynamin inhibition. We further show that endocytosis inhibition does not induce autophagy even though mTORC1 inactivation is known to strongly induce autophagy. These findings open new perspectives for the use of endocytosis inhibitors as potential agents that can effectively inhibit nutrient utilization and shut down the upstream signals that activate mTORC1.
Collapse
Affiliation(s)
- Shusaku Shibutani
- From the Laboratory of Veterinary Hygiene, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi 753-8515, Japan
| | - Hana Okazaki
- From the Laboratory of Veterinary Hygiene, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi 753-8515, Japan
| | - Hiroyuki Iwata
- From the Laboratory of Veterinary Hygiene, Joint Faculty of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi 753-8515, Japan
| |
Collapse
|
36
|
Potheraveedu VN, Schöpel M, Stoll R, Heumann R. Rheb in neuronal degeneration, regeneration, and connectivity. Biol Chem 2017; 398:589-606. [PMID: 28212107 DOI: 10.1515/hsz-2016-0312] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2016] [Accepted: 02/02/2017] [Indexed: 01/31/2023]
Abstract
The small GTPase Rheb was originally detected as an immediate early response protein whose expression was induced by NMDA-dependent synaptic activity in the brain. Rheb's activity is highly regulated by its GTPase activating protein (GAP), the tuberous sclerosis complex protein, which stimulates the conversion from the active, GTP-loaded into the inactive, GDP-loaded conformation. Rheb has been established as an evolutionarily conserved molecular switch protein regulating cellular growth, cell volume, cell cycle, autophagy, and amino acid uptake. The subcellular localization of Rheb and its interacting proteins critically regulate its activity and function. In stem cells, constitutive activation of Rheb enhances differentiation at the expense of self-renewal partially explaining the adverse effects of deregulated Rheb in the mammalian brain. In the context of various cellular stress conditions such as oxidative stress, ER-stress, death factor signaling, and cellular aging, Rheb activation surprisingly enhances rather than prevents cellular degeneration. This review addresses cell type- and cell state-specific function(s) of Rheb and mainly focuses on neurons and their surrounding glial cells. Mechanisms will be discussed in the context of therapy that interferes with Rheb's activity using the antibiotic rapamycin or low molecular weight compounds.
Collapse
Affiliation(s)
- Veena Nambiar Potheraveedu
- Molecular Neurobiochemistry, Faculty of Chemistry and Biochemistry, Ruhr University of Bochum, Universitätstr. 150, D-44780 Bochum
| | - Miriam Schöpel
- Biomolecular NMR, Ruhr University of Bochum, D-44780 Bochum
| | - Raphael Stoll
- Biomolecular NMR, Ruhr University of Bochum, D-44780 Bochum
| | - Rolf Heumann
- Molecular Neurobiochemistry, Faculty of Chemistry and Biochemistry, Ruhr University of Bochum, Universitätstr. 150, D-44780 Bochum
| |
Collapse
|
37
|
The Architecture of the Rag GTPase Signaling Network. Biomolecules 2017; 7:biom7030048. [PMID: 28788436 PMCID: PMC5618229 DOI: 10.3390/biom7030048] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 06/22/2017] [Accepted: 06/27/2017] [Indexed: 12/11/2022] Open
Abstract
The evolutionarily conserved target of rapamycin complex 1 (TORC1) couples an array of intra- and extracellular stimuli to cell growth, proliferation and metabolism, and its deregulation is associated with various human pathologies such as immunodeficiency, epilepsy, and cancer. Among the diverse stimuli impinging on TORC1, amino acids represent essential input signals, but how they control TORC1 has long remained a mystery. The recent discovery of the Rag GTPases, which assemble as heterodimeric complexes on vacuolar/lysosomal membranes, as central elements of an amino acid signaling network upstream of TORC1 in yeast, flies, and mammalian cells represented a breakthrough in this field. Here, we review the architecture of the Rag GTPase signaling network with a special focus on structural aspects of the Rag GTPases and their regulators in yeast and highlight both the evolutionary conservation and divergence of the mechanisms that control Rag GTPases.
Collapse
|
38
|
Chichger H, Braza J, Duong H, Boni G, Harrington EO. Select Rab GTPases Regulate the Pulmonary Endothelium via Endosomal Trafficking of Vascular Endothelial-Cadherin. Am J Respir Cell Mol Biol 2017; 54:769-81. [PMID: 26551054 DOI: 10.1165/rcmb.2015-0286oc] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Pulmonary edema occurs in settings of acute lung injury, in diseases, such as pneumonia, and in acute respiratory distress syndrome. The lung interendothelial junctions are maintained in part by vascular endothelial (VE)-cadherin, an adherens junction protein, and its surface expression is regulated by endocytic trafficking. The Rab family of small GTPases are regulators of endocytic trafficking. The key trafficking pathways are regulated by Rab4, -7, and -9. Rab4 regulates the recycling of endosomes to the cell surface through a rapid-shuttle process, whereas Rab7 and -9 regulate trafficking to the late endosome/lysosome for degradation or from the trans-Golgi network to the late endosome, respectively. We recently demonstrated a role for the endosomal adaptor protein, p18, in regulation of the pulmonary endothelium through enhanced recycling of VE-cadherin to adherens junction. Thus, we hypothesized that Rab4, -7, and -9 regulate pulmonary endothelial barrier function through modulating trafficking of VE-cadherin-positive endosomes. We used Rab mutants with varying activities and associations to the endosome to study endothelial barrier function in vitro and in vivo. Our study demonstrates a key role for Rab4 activation and Rab9 inhibition in regulation of vascular permeability through enhanced VE-cadherin expression at the interendothelial junction. We further showed that endothelial barrier function mediated through Rab4 is dependent on extracellular signal-regulated kinase phosphorylation and activity. Thus, we demonstrate that Rab4 and -9 regulate VE-cadherin levels at the cell surface to modulate the pulmonary endothelium through extracellular signal-regulated kinase-dependent and -independent pathways, respectively. We propose that regulating select Rab GTPases represents novel therapeutic strategies for patients suffering with acute respiratory distress syndrome.
Collapse
Affiliation(s)
- Havovi Chichger
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, Rhode Island; and Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Julie Braza
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, Rhode Island; and Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Huetran Duong
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, Rhode Island; and Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Geraldine Boni
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, Rhode Island; and Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| | - Elizabeth O Harrington
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center, Providence, Rhode Island; and Department of Medicine, Alpert Medical School of Brown University, Providence, Rhode Island
| |
Collapse
|
39
|
Zhikrivetskaya SO, Snezhkina AV, Zaretsky AR, Alekseev BY, Pokrovsky AV, Golovyuk AL, Melnikova NV, Stepanov OA, Kalinin DV, Moskalev AA, Krasnov GS, Dmitriev AA, Kudryavtseva AV. Molecular markers of paragangliomas/pheochromocytomas. Oncotarget 2017; 8:25756-25782. [PMID: 28187001 PMCID: PMC5421967 DOI: 10.18632/oncotarget.15201] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 01/23/2017] [Indexed: 12/14/2022] Open
Abstract
Paragangliomas/pheochromocytomas comprise rare tumors that arise from the extra-adrenal paraganglia, with an incidence of about 2 to 8 per million people each year. Approximately 40% of cases are due to genetic mutations in at least one out of more than 30 causative genes. About 25-30% of pheochromocytomas/paragangliomas develop under the conditions of a hereditary tumor syndrome a third of which are caused by mutations in the VHL gene. Together, the gene mutations in this disorder have implicated multiple processes including signaling pathways, translation initiation, hypoxia regulation, protein synthesis, differentiation, survival, proliferation, and cell growth. The present review contemplates the mutations associated with the development of pheochromocytomas/paragangliomas and their potential to serve as specific markers of these tumors and their progression. These data will improve our understanding of the pathogenesis of these tumors and likely reveal certain features that may be useful for early diagnostics, malignancy prognostics, and the determination of new targets for disease therapeutics.
Collapse
Affiliation(s)
| | | | - Andrew R Zaretsky
- M.M. Shemyakin - Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Boris Y Alekseev
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | | | | | - Nataliya V Melnikova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Oleg A Stepanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| | | | - Alexey A Moskalev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - George S Krasnov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Alexey A Dmitriev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Anna V Kudryavtseva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- National Medical Research Radiological Center, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
40
|
Davis LS, Reimold AM. Transcriptional profiling of leukocytes from rheumatoid arthritis patients before and after anti-tumor necrosis factor therapy: A comparison of anti-nuclear antibody positive and negative subsets. Exp Ther Med 2017; 13:2183-2192. [PMID: 28565826 PMCID: PMC5443193 DOI: 10.3892/etm.2017.4265] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 01/06/2017] [Indexed: 12/13/2022] Open
Abstract
Anti-nuclear antibodies (ANAs) may be induced in patients with rheumatoid arthritis (RA) receiving anti-tumor necrosis factor (TNF) therapy with TNF inhibitors (TNFi), etanercept, infliximab or adalimumab. In the present study, 11 patients who were TNFi drug naive were started on TNFi at a time of high disease activity. Of these, all cases were positive for rheumatoid factor and 9 cases tested were positive for anti-citrullinated peptide (anti-CCP) antibodies prior to TNFi treatment. Peripheral blood mononuclear cells (PBMCs) and serum were collected from all patients before and after TNFi therapy. Serum was assayed for ANAs over time. Total cellular RNA was extracted from PBMCs and assessed using Illumina arrays. Gene expression profiles were examined for alterations in key effector pathways. After 3 or more months on TNFi, 6 patients converted to ANA-positivity. Analysis of transcripts from patients with RA who converted to ANA-positivity after 3 months on TNFi identified complex gene expression profiles that reflected a reduction in cell adhesion, cell stress and lipid metabolism transcripts. In summary, unique transcriptional profiles in PBMCs from patients with RA were observed after TNFi therapy. This pilot study suggests that transcriptional profiling is a precise method of measuring the impact of TNFi therapies and reveals novel pathways that likely influence the immune response.
Collapse
Affiliation(s)
- Laurie S Davis
- Rheumatic Diseases Division, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8884, USA
| | - Andreas M Reimold
- Rheumatic Diseases Division, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390-8884, USA.,Rheumatology Section, Dallas VA Medical Center, Dallas, TX 75216, USA
| |
Collapse
|
41
|
Jacobs BL, McNally RM, Kim KJ, Blanco R, Privett RE, You JS, Hornberger TA. Identification of mechanically regulated phosphorylation sites on tuberin (TSC2) that control mechanistic target of rapamycin (mTOR) signaling. J Biol Chem 2017; 292:6987-6997. [PMID: 28289099 PMCID: PMC5409467 DOI: 10.1074/jbc.m117.777805] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 03/08/2017] [Indexed: 12/31/2022] Open
Abstract
Mechanistic target of rapamycin (mTOR) signaling is necessary to generate a mechanically induced increase in skeletal muscle mass, but the mechanism(s) through which mechanical stimuli regulate mTOR signaling remain poorly defined. Recent studies have suggested that Ras homologue enriched in brain (Rheb), a direct activator of mTOR, and its inhibitor, the GTPase-activating protein tuberin (TSC2), may play a role in this pathway. To address this possibility, we generated inducible and skeletal muscle-specific knock-out mice for Rheb (iRhebKO) and TSC2 (iTSC2KO) and mechanically stimulated muscles from these mice with eccentric contractions (EC). As expected, the knock-out of TSC2 led to an elevation in the basal level of mTOR signaling. Moreover, we found that the magnitude of the EC-induced activation of mTOR signaling was significantly blunted in muscles from both inducible and skeletal muscle-specific knock-out mice for Rheb and iTSC2KO mice. Using mass spectrometry, we identified six sites on TSC2 whose phosphorylation was significantly altered by the EC treatment. Employing a transient transfection-based approach to rescue TSC2 function in muscles of the iTSC2KO mice, we demonstrated that these phosphorylation sites are required for the role that TSC2 plays in the EC-induced activation of mTOR signaling. Importantly, however, these phosphorylation sites were not required for an insulin-induced activation of mTOR signaling. As such, our results not only establish a critical role for Rheb and TSC2 in the mechanical activation of mTOR signaling, but they also expose the existence of a previously unknown branch of signaling events that can regulate the TSC2/mTOR pathway.
Collapse
Affiliation(s)
- Brittany L Jacobs
- From the Department of Comparative Biosciences and.,the School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706
| | - Rachel M McNally
- From the Department of Comparative Biosciences and.,the School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706
| | - Kook-Joo Kim
- From the Department of Comparative Biosciences and.,the School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706
| | - Rocky Blanco
- From the Department of Comparative Biosciences and.,the School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706
| | - Rachel E Privett
- From the Department of Comparative Biosciences and.,the School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706
| | - Jae-Sung You
- From the Department of Comparative Biosciences and.,the School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706
| | - Troy A Hornberger
- From the Department of Comparative Biosciences and .,the School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, 53706
| |
Collapse
|
42
|
Abstract
Cellular adaptation response to a myriad of stressors is key for survival. The lysosomal/autophagy pathway is inextricably connected to the stress response regulation. Two transcription factors, TFEB and TFE3, have recently emerged as master regulators of this degradative pathway. Their function modulating different cellular pathways will be discussed.
Collapse
Affiliation(s)
- José A Martina
- a Cell Biology and Physiology Center , National Heart, Lung, and Blood Institute, National Institutes of Health , Bethesda , MD , USA
| | - Rosa Puertollano
- a Cell Biology and Physiology Center , National Heart, Lung, and Blood Institute, National Institutes of Health , Bethesda , MD , USA
| |
Collapse
|
43
|
Roles of mTOR complexes in the kidney: implications for renal disease and transplantation. Nat Rev Nephrol 2016; 12:587-609. [PMID: 27477490 DOI: 10.1038/nrneph.2016.108] [Citation(s) in RCA: 157] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The mTOR pathway has a central role in the regulation of cell metabolism, growth and proliferation. Studies involving selective gene targeting of mTOR complexes (mTORC1 and mTORC2) in renal cell populations and/or pharmacologic mTOR inhibition have revealed important roles of mTOR in podocyte homeostasis and tubular transport. Important advances have also been made in understanding the role of mTOR in renal injury, polycystic kidney disease and glomerular diseases, including diabetic nephropathy. Novel insights into the roles of mTORC1 and mTORC2 in the regulation of immune cell homeostasis and function are helping to improve understanding of the complex effects of mTOR targeting on immune responses, including those that impact both de novo renal disease and renal allograft outcomes. Extensive experience in clinical renal transplantation has resulted in successful conversion of patients from calcineurin inhibitors to mTOR inhibitors at various times post-transplantation, with excellent long-term graft function. Widespread use of this practice has, however, been limited owing to mTOR-inhibitor- related toxicities. Unique attributes of mTOR inhibitors include reduced rates of squamous cell carcinoma and cytomegalovirus infection compared to other regimens. As understanding of the mechanisms by which mTORC1 and mTORC2 drive the pathogenesis of renal disease progresses, clinical studies of mTOR pathway targeting will enable testing of evolving hypotheses.
Collapse
|
44
|
Gonzalez A, Moya-Alvarado G, Gonzalez-Billaut C, Bronfman FC. Cellular and molecular mechanisms regulating neuronal growth by brain-derived neurotrophic factor. Cytoskeleton (Hoboken) 2016; 73:612-628. [PMID: 27223597 DOI: 10.1002/cm.21312] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 05/20/2016] [Accepted: 05/23/2016] [Indexed: 12/31/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) and its receptors TrkB and p75 regulate dendritic and axonal growth during development and maintenance of the mature nervous system; however, the cellular and molecular mechanisms underlying this process are not fully understood. In recent years, several advances have shed new light on the processes behind the regulation of BDNF-mediated structural plasticity including control of neuronal transcription, local translation of proteins, and regulation of cytoskeleton and membrane dynamics. In this review, we summarize recent advances in the field of BDNF signaling in neurons to induce neuronal growth. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Andres Gonzalez
- MINREB and Center for Ageing and Regeneration (CARE UC), Faculty of Biological Sciences, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Guillermo Moya-Alvarado
- MINREB and Center for Ageing and Regeneration (CARE UC), Faculty of Biological Sciences, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Christian Gonzalez-Billaut
- Laboratory of Cell and Neuronal Dynamics, Department of Biology, Faculty of Sciences, Universidad de Chile and Geroscience Center for Brain Health and Metabolism, Santiago, Chile
| | - Francisca C Bronfman
- MINREB and Center for Ageing and Regeneration (CARE UC), Faculty of Biological Sciences, Department of Physiology, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
45
|
Powis K, De Virgilio C. Conserved regulators of Rag GTPases orchestrate amino acid-dependent TORC1 signaling. Cell Discov 2016; 2:15049. [PMID: 27462445 PMCID: PMC4860963 DOI: 10.1038/celldisc.2015.49] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 12/02/2015] [Indexed: 12/16/2022] Open
Abstract
The highly conserved target of rapamycin complex 1 (TORC1) is the central component of a signaling network that couples a vast range of internal and external stimuli to cell growth, proliferation and metabolism. TORC1 deregulation is associated with a number of human pathologies, including many cancers and metabolic disorders, underscoring its importance in cellular and organismal growth control. The activity of TORC1 is modulated by multiple inputs; however, the presence of amino acids is a stimulus that is essential for its activation. Amino acid sufficiency is communicated to TORC1 via the highly conserved family of Rag GTPases, which assemble as heterodimeric complexes on lysosomal/vacuolar membranes and are regulated by their guanine nucleotide loading status. Studies in yeast, fly and mammalian model systems have revealed a multitude of conserved Rag GTPase modulators, which have greatly expanded our understanding of amino acid sensing by TORC1. Here we review the major known modulators of the Rag GTPases, focusing on recent mechanistic insights that highlight the evolutionary conservation and divergence of amino acid signaling to TORC1.
Collapse
Affiliation(s)
- Katie Powis
- Department of Biology, University of Fribourg , Fribourg, Switzerland
| | | |
Collapse
|
46
|
Role of the mammalian target of rapamycin pathway in lentiviral vector transduction of hematopoietic stem cells. Curr Opin Hematol 2016; 22:302-8. [PMID: 26049750 DOI: 10.1097/moh.0000000000000150] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
PURPOSE OF REVIEW A major goal in repopulating hematopoietic stem cell (HSC) gene therapies is achieving high-efficacy gene transfer, while maintaining robust HSC engraftment and differentiation in vivo. Recent studies have documented that rapamycin treatment of HSC during lentiviral vector transduction enhances gene transfer to human and mouse HSCs and maintains engraftment capacity. In this review, we place into context the role of mammalian target of rapamycin (mTOR) pathways in HSC quiescence and function, endocytic regulation, and lentiviral gene delivery. RECENT FINDINGS Lentiviral vector transduction of human and mouse HSCs is considerably enhanced by rapamycin treatment. Furthermore, rapamycin preserves long-term engraftment of human and mouse HSCs. Investigations of cellular mechanisms that contribute to increased transduction in HSCs uncover a role for mTOR inhibition-dependent activation of endocytosis. SUMMARY Rapamycin enhances lentiviral vector transduction of HSCs through regulation of endocytic activity via mTOR inhibition. An important attribute of rapamycin treatment during transduction is the preservation of HSC function, allowing reconstitution of long-term hematopoiesis in vivo in murine models.
Collapse
|
47
|
Abstract
Rag small GTPases were identified as the sixth subfamily of Ras-related GTPases. Compelling evidence suggests that Rag heterodimer (RagA/B and RagC/D) plays an important role in amino acid signaling toward mechanistic target of rapamycin complex 1 (mTORC1), which is a central player in the control of cell growth in response to a variety of environmental cues, including growth factors, cellular energy/oxygen status, and amino acids. Upon amino acid stimulation, active Rag heterodimer (RagA/B(GTP)-RagC/D(GDP)) recruits mTORC1 to the lysosomal membrane where Rheb resides. In this review, we provide a current understanding on the amino acid-regulated cell growth control via Rag-mTORC1 with recently identified key players, including Ragulator, v-ATPase, and GATOR complexes. Moreover, the functions of Rag in physiological systems and in autophagy are discussed.
Collapse
|
48
|
Vesicular Trafficking Systems Impact TORC1-Controlled Transcriptional Programs in Saccharomyces cerevisiae. G3-GENES GENOMES GENETICS 2016; 6:641-52. [PMID: 26739646 PMCID: PMC4777127 DOI: 10.1534/g3.115.023911] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
The Target of Rapamycin Complex I (TORC1) orchestrates global reprogramming of transcriptional programs in response to myriad environmental conditions, yet, despite the commonality of the TORC1 complex components, different TORC1-inhibitory conditions do not elicit a uniform transcriptional response. In Saccharomyces cerevisiae, TORC1 regulates the expression of nitrogen catabolite repressed (NCR) genes by controlling the nuclear translocation of the NCR transactivator Gln3. Moreover, Golgi-to-endosome trafficking was shown to be required for nuclear translocation of Gln3 upon a shift from rich medium to the poor nitrogen source proline, but not upon rapamycin treatment. Here, we employed microarray profiling to survey the full impact of the vesicular trafficking system on yeast TORC1-orchestrated transcriptional programs. In addition to the NCR genes, we found that ribosomal protein, ribosome biogenesis, phosphate-responsive, and sulfur-containing amino acid metabolism genes are perturbed by disruption of Golgi-to-endosome trafficking following a nutritional shift from rich to poor nitrogen source medium, but not upon rapamycin treatment. Similar to Gln3, defects in Golgi-to-endosome trafficking significantly delayed cytoplasmic–nuclear translocation of Sfp1, but did not detectably affect the cytoplasmic–nuclear or nuclear–cytoplasmic translocation of Met4, which are the transactivators of these genes. Thus, Golgi-to-endosome trafficking defects perturb TORC1 transcriptional programs via multiple mechanisms. Our findings further delineate the downstream transcriptional responses of TORC1 inhibition by rapamycin compared with a nitrogen quality downshift. Given the conservation of both TORC1 and endomembrane networks throughout eukaryotes, our findings may also have implications for TORC1-mediated responses to nutritional cues in mammals and other eukaryotes.
Collapse
|
49
|
Herter JM, Grabie N, Cullere X, Azcutia V, Rosetti F, Bennett P, Herter-Sprie GS, Elyaman W, Luscinskas FW, Lichtman AH, Mayadas TN. AKAP9 regulates activation-induced retention of T lymphocytes at sites of inflammation. Nat Commun 2015; 6:10182. [PMID: 26680259 PMCID: PMC4703868 DOI: 10.1038/ncomms10182] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 11/12/2015] [Indexed: 12/14/2022] Open
Abstract
The mechanisms driving T cell homing to lymph nodes and migration to tissue are well described but little is known about factors that affect T cell egress from tissues. Here, we generate mice with a T cell-specific deletion of the scaffold protein A kinase anchoring protein 9 (AKAP9) and use models of inflammatory disease to demonstrate that AKAP9 is dispensable for T cell priming and migration into tissues and lymph nodes, but is required for T cell retention in tissues. AKAP9 deficiency results in increased T cell egress to draining lymph nodes, which is associated with impaired T cell re-activation in tissues and protection from organ damage. AKAP9-deficient T cells exhibit reduced microtubule-dependent recycling of TCRs back to the cell surface and this affects antigen-dependent activation, primarily by non-classical antigen-presenting cells. Thus, AKAP9-dependent TCR trafficking drives efficient T cell re-activation and extends their retention at sites of inflammation with implications for disease pathogenesis. A-kinase anchoring protein 9 (AKAP9) is a scaffold protein that binds signalling proteins and regulates microtubules. Here the authors show that during inflammation AKAP9 in T cells is required for their reactivation and retention at the inflammation site and that its deletion protects from inflammation-induced organ damage.
Collapse
Affiliation(s)
- Jan M Herter
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, Boston, Massachusetts 02115, USA
| | - Nir Grabie
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, Boston, Massachusetts 02115, USA
| | - Xavier Cullere
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, Boston, Massachusetts 02115, USA
| | - Veronica Azcutia
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, Boston, Massachusetts 02115, USA
| | - Florencia Rosetti
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, Boston, Massachusetts 02115, USA
| | - Paul Bennett
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, Boston, Massachusetts 02115, USA
| | - Grit S Herter-Sprie
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, 450 Brookline Avenue, Boston, Massachusetts 02115, USA
| | - Wassim Elyaman
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, Boston, Massachusetts 02115, USA
| | - Francis W Luscinskas
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, Boston, Massachusetts 02115, USA
| | - Andrew H Lichtman
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, Boston, Massachusetts 02115, USA
| | - Tanya N Mayadas
- Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, 77 Avenue Louis Pasteur, Boston, Massachusetts 02115, USA
| |
Collapse
|
50
|
Caza T, Landas S. Functional and Phenotypic Plasticity of CD4(+) T Cell Subsets. BIOMED RESEARCH INTERNATIONAL 2015; 2015:521957. [PMID: 26583116 PMCID: PMC4637038 DOI: 10.1155/2015/521957] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 05/20/2015] [Accepted: 06/04/2015] [Indexed: 12/19/2022]
Abstract
The remarkable plasticity of CD4(+) T cells allows individuals to respond to environmental stimuli in a context-dependent manner. A balance of CD4(+) T cell subsets is critical to mount responses against pathogen challenges to prevent inappropriate activation, to maintain tolerance, and to participate in antitumor immune responses. Specification of subsets is a process beginning in intrathymic development and continuing within the circulation. It is highly flexible to adapt to differences in nutrient availability and the tissue microenvironment. CD4(+) T cell subsets have significant cross talk, with the ability to "dedifferentiate" given appropriate environmental signals. This ability is dependent on the metabolic status of the cell, with mTOR acting as the rheostat. Autoimmune and antitumor immune responses are regulated by the balance between regulatory T cells and Th17 cells. When a homeostatic balance of subsets is not maintained, immunopathology can result. CD4(+) T cells carry complex roles within tumor microenvironments, with context-dependent immune responses influenced by oncogenic drivers and the presence of inflammation. Here, we examine the signals involved in CD4(+) T cell specification towards each subset, interconnectedness of cytokine networks, impact of mTOR signaling, and cellular metabolism in lineage specification and provide a supplement describing techniques to study these processes.
Collapse
Affiliation(s)
- Tiffany Caza
- Department of Pathology, Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA
| | - Steve Landas
- Department of Pathology, Upstate Medical University, 750 East Adams Street, Syracuse, NY 13210, USA
| |
Collapse
|