1
|
Szalai F, Sztankovics D, Krencz I, Moldvai D, Pápay J, Sebestyén A, Khoor A. Rictor-A Mediator of Progression and Metastasis in Lung Cancer. Cancers (Basel) 2024; 16:543. [PMID: 38339294 PMCID: PMC10854599 DOI: 10.3390/cancers16030543] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
Lung carcinoma is one of the most common cancer types for both men and women. Despite recent breakthroughs in targeted therapy and immunotherapy, it is characterized by a high metastatic rate, which can significantly affect quality of life and prognosis. Rictor (encoded by the RICTOR gene) is known as a scaffold protein for the multiprotein complex mTORC2. Among its diverse roles in regulating essential cellular functions, mTORC2 also facilitates epithelial-mesenchymal transition and metastasis formation. Amplification of the RICTOR gene and subsequent overexpression of the Rictor protein can result in the activation of mTORC2, which promotes cell survival and migration. Based on recent studies, RICTOR amplification or Rictor overexpression can serve as a marker for mTORC2 activation, which in turn provides a promising druggable target. Although selective inhibitors of Rictor and the Rictor-mTOR association are only in a preclinical phase, they seem to be potent novel approaches to reduce tumor cell migration and metastasis formation. Here, we summarize recent advances that support an important role for Rictor and mTORC2 as potential therapeutic targets in the treatment of lung cancer. This is a traditional (narrative) review based on Pubmed and Google Scholar searches for the following keywords: Rictor, RICTOR amplification, mTORC2, Rictor complexes, lung cancer, metastasis, progression, mTOR inhibitors.
Collapse
Affiliation(s)
- Fatime Szalai
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (F.S.); (D.S.); (I.K.); (D.M.); (J.P.); (A.S.)
| | - Dániel Sztankovics
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (F.S.); (D.S.); (I.K.); (D.M.); (J.P.); (A.S.)
| | - Ildikó Krencz
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (F.S.); (D.S.); (I.K.); (D.M.); (J.P.); (A.S.)
| | - Dorottya Moldvai
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (F.S.); (D.S.); (I.K.); (D.M.); (J.P.); (A.S.)
| | - Judit Pápay
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (F.S.); (D.S.); (I.K.); (D.M.); (J.P.); (A.S.)
| | - Anna Sebestyén
- Department of Pathology and Experimental Cancer Research, Semmelweis University, Üllői út 26, H-1085 Budapest, Hungary; (F.S.); (D.S.); (I.K.); (D.M.); (J.P.); (A.S.)
| | - Andras Khoor
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| |
Collapse
|
2
|
Ragupathi A, Kim C, Jacinto E. The mTORC2 signaling network: targets and cross-talks. Biochem J 2024; 481:45-91. [PMID: 38270460 PMCID: PMC10903481 DOI: 10.1042/bcj20220325] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/29/2023] [Accepted: 12/18/2023] [Indexed: 01/26/2024]
Abstract
The mechanistic target of rapamycin, mTOR, controls cell metabolism in response to growth signals and stress stimuli. The cellular functions of mTOR are mediated by two distinct protein complexes, mTOR complex 1 (mTORC1) and mTORC2. Rapamycin and its analogs are currently used in the clinic to treat a variety of diseases and have been instrumental in delineating the functions of its direct target, mTORC1. Despite the lack of a specific mTORC2 inhibitor, genetic studies that disrupt mTORC2 expression unravel the functions of this more elusive mTOR complex. Like mTORC1 which responds to growth signals, mTORC2 is also activated by anabolic signals but is additionally triggered by stress. mTORC2 mediates signals from growth factor receptors and G-protein coupled receptors. How stress conditions such as nutrient limitation modulate mTORC2 activation to allow metabolic reprogramming and ensure cell survival remains poorly understood. A variety of downstream effectors of mTORC2 have been identified but the most well-characterized mTORC2 substrates include Akt, PKC, and SGK, which are members of the AGC protein kinase family. Here, we review how mTORC2 is regulated by cellular stimuli including how compartmentalization and modulation of complex components affect mTORC2 signaling. We elaborate on how phosphorylation of its substrates, particularly the AGC kinases, mediates its diverse functions in growth, proliferation, survival, and differentiation. We discuss other signaling and metabolic components that cross-talk with mTORC2 and the cellular output of these signals. Lastly, we consider how to more effectively target the mTORC2 pathway to treat diseases that have deregulated mTOR signaling.
Collapse
Affiliation(s)
- Aparna Ragupathi
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, U.S.A
| | - Christian Kim
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, U.S.A
| | - Estela Jacinto
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, NJ, U.S.A
| |
Collapse
|
3
|
Bloom GS, Norambuena A. Dysregulation of mTOR by tau in Alzheimer's disease. Cytoskeleton (Hoboken) 2024; 81:30-34. [PMID: 37638691 PMCID: PMC10919542 DOI: 10.1002/cm.21782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/17/2023] [Accepted: 08/18/2023] [Indexed: 08/29/2023]
Abstract
Tau was discovered in the mid 1970's as a microtubule-associated protein that stimulates tubulin polymerization, and subsequently was shown to be expressed primarily in neurons, where it is most concentrated in axons. Interest in tau rose by the late 1980's, when it was shown to be the principal subunit of the neurofibrillary tangles (NFTs) that accumulate in Alzheimer's disease (AD) brain, and achieved new heights by the late 1990's, when numerous tau mutations were found to be highly penetrant for AD-related disorders that also are associated with NFTs and came to be known as non-Alzheimer's tauopathies. The role of tau in neurodegeneration is far more complex than whatever effects on neurons may be caused by NFTs, however, and here we review our work on dysregulation of mTOR by tau in AD. mTOR is a protein kinase and master regulator of myriad aspects of cellular behavior. We have defined a complex signaling network whereby aberrant tau phosphorylation provoked by amyloid-β oligomers (AβOs), the building blocks of the amyloid plaques that form in AD brain, cause post-mitotic neurons to re-enter the cell cycle, but to die eventually instead of dividing, which may account for most neuron death in AD. Remarkably, we found that this same neuronal signaling network also poisons a fundamental cell biological process that we discovered, nutrient-induced mitochondrial activation, or NiMA. Tau-dependent cell cycle re-entry and NiMA inhibition occur in cultured neurons within a few hours of exposure to AβOs, and thus may represent seminal processes in AD pathogenesis.
Collapse
Affiliation(s)
- George S. Bloom
- Department of Biology, University of Virginia, Charlottesville, Virginia, USA
- Department of Cell Biology, University of Virginia, Charlottesville, Virginia, USA
- Department of Neuroscience, University of Virginia, Charlottesville, Virginia, USA
| | - Andrés Norambuena
- Department of Biology, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
4
|
Xu Z, Yan Y, Gu B, Cai W, Wang Y. Up-Regulation of microRNA-424 Causes an Imbalance in AKT Phosphorylation and Impairs Enteric Neural Crest Cell Migration in Hirschsprung Disease. Int J Mol Sci 2023; 24:ijms24076700. [PMID: 37047673 PMCID: PMC10094892 DOI: 10.3390/ijms24076700] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/05/2023] [Accepted: 03/10/2023] [Indexed: 04/07/2023] Open
Abstract
Insights into the role of microRNAs (miRNAs) in disease pathogenesis have made them attractive therapeutic targets, and numerous miRNAs have been functionally linked to Hirschsprung disease (HSCR), a life-threatening genetic disorder due to defective migration, proliferation, and colonization of enteric neural crest cells (ENCCs) in the gut. Recent studies have demonstrated that miR-424 strongly inhibits migration in a variety of cell types and its potential target RICTOR is essential for neural crest cell development. We therefore sought to interrogate how miR-424 and RICTOR contribute to the pathogenesis of HSCR. We utilized HSCR cases and human neural cells to evaluate the miR-424-mediated regulation of RICTOR and the downstream AKT phosphorylation. We further developed an ex vivo model to assess the effects of miR-424 on ENCC migration and proliferation. Then, single-cell atlases of gene expression in both human and mouse fetal intestines were used to determine the characteristics of RICTOR and AKT expression in the developing gut. Our findings demonstrate that miR-424 levels are markedly increased in the colonic tissues of patients with HSCR and that it regulates human neural cell migration by directly targeting RICTOR. Up-regulation of miR-424 leads to decreased AKT phosphorylation levels in a RICTOR-dependent manner, and this, in turn, impairs ENCC proliferation and migration in the developing gut. Interestingly, we further identified prominent RICTOR and AKT expressions in the enteric neurons and other types of enteric neural cells in human and mouse fetal intestines. Our present study reveals the role of the miR-424/RICTOR axis in HSCR pathogenesis and indicates that miR-424 is a promising candidate for the development of targeted therapies against HSCR.
Collapse
Affiliation(s)
- Ze Xu
- Department of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China
- Shanghai Institute for Pediatric Research, Shanghai 200092, China
| | - Yingnan Yan
- Department of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China
| | - Beilin Gu
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China
- Shanghai Institute for Pediatric Research, Shanghai 200092, China
| | - Wei Cai
- Department of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China
- Shanghai Institute for Pediatric Research, Shanghai 200092, China
| | - Yang Wang
- Department of Pediatric Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
- Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai 200092, China
- Shanghai Institute for Pediatric Research, Shanghai 200092, China
| |
Collapse
|
5
|
SMOC2 promotes aggressive behavior of fibroblast-like synoviocytes in rheumatoid arthritis through transcriptional and post-transcriptional regulating MYO1C. Cell Death Dis 2022; 13:1035. [PMID: 36513634 PMCID: PMC9747908 DOI: 10.1038/s41419-022-05479-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 11/23/2022] [Accepted: 11/28/2022] [Indexed: 12/14/2022]
Abstract
Fibroblast-like synoviocytes (FLSs), play a key role in perpetuating synovial inflammation and bone erosion in rheumatoid arthritis (RA), however, the underlying mechanism(s) of RA FLSs activation and aggression remain unclear. Identifying endogenous proteins that selectively target FLSs is urgently needed. Here, we systematically identified that secreted modular calcium-binding protein 2 (SMOC2), was significantly increased in RA FLSs and synovial tissues. SMOC2 knockdown specifically regulated cytoskeleton remodeling and decreased the migration and invasion of RA FLSs. Mechanistically, cytoskeleton-related genes were significantly downregulated in RA FLSs with reduced SMOC2 expression, especially the motor protein myosin1c (MYO1C). SMOC2 controlled MYO1C expression by SRY-related high-mobility group box 4 (SOX4) and AlkB homolog 5 (ALKHB5) mediated-m6A modification through transcriptional and post-transcriptional regulation. Furthermore, intra-articular Ad-shRNA-SMOC2 treatment attenuated synovial inflammation as well as bone and cartilage erosion in rats with collagen-induced arthritis (CIA). Our findings suggest that increased SMOC2 expression in FLSs may contribute to synovial aggression and joint destruction in RA. SMOC2 may serve as a potential target against RA. SMOC2-mediated regulation of the synovial migration and invasion in RA FLSs. In RA FLSs, SMOC2 is significantly increased, leading to the increased level of MYO1C via SOX4-mediated transcriptional regulation and ALKBH5-mediated m6A modification, thereby causing cytoskeleton remodeling and promoting RA FLSs migration and invasion. The Figure was drawn by Figdraw.
Collapse
|
6
|
Gomes KP, Jadli AS, de Almeida LGN, Ballasy NN, Edalat P, Shandilya R, Young D, Belke D, Shearer J, Dufour A, Patel VB. Proteomic Analysis Suggests Altered Mitochondrial Metabolic Profile Associated With Diabetic Cardiomyopathy. Front Cardiovasc Med 2022; 9:791700. [PMID: 35310970 PMCID: PMC8924072 DOI: 10.3389/fcvm.2022.791700] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 01/24/2022] [Indexed: 01/04/2023] Open
Abstract
Diabetic cardiomyopathy (DbCM) occurs independently of cardiovascular diseases or hypertension, leading to heart failure and increased risk for death in diabetic patients. To investigate the molecular mechanisms involved in DbCM, we performed a quantitative proteomic profiling analysis in the left ventricle (LV) of type 2 diabetic mice. Six-month-old C57BL/6J-lepr/lepr (db/db) mice exhibited DbCM associated with diastolic dysfunction and cardiac hypertrophy. Using quantitative shotgun proteomic analysis, we identified 53 differentially expressed proteins in the LVs of db/db mice, majorly associated with the regulation of energy metabolism. The subunits of ATP synthase that form the F1 domain, and Cytochrome c1, a catalytic core subunit of the complex III primarily responsible for electron transfer to Cytochrome c, were upregulated in diabetic LVs. Upregulation of these key proteins may represent an adaptive mechanism by diabetic heart, resulting in increased electron transfer and thereby enhancement of mitochondrial ATP production. Conversely, diabetic LVs also showed a decrease in peptide levels of NADH dehydrogenase 1β subcomplex subunit 11, a subunit of complex I that catalyzes the transfer of electrons to ubiquinone. Moreover, the atypical kinase COQ8A, an essential lipid-soluble electron transporter involved in the biosynthesis of ubiquinone, was also downregulated in diabetic LVs. Our study indicates that despite attempts by hearts from diabetic mice to augment mitochondrial ATP energetics, decreased levels of key components of the electron transport chain may contribute to impaired mitochondrial ATP production. Preserved basal mitochondrial respiration along with the markedly reduced maximal respiratory capacity in the LVs of db/db mice corroborate the association between altered mitochondrial metabolic profile and cardiac dysfunction in DbCM.
Collapse
Affiliation(s)
- Karina P. Gomes
- Department of Physiology and Pharmacology, Cumming School of Medicine, Calgary, AB, Canada
- Libin Cardiovascular Institute, Calgary, AB, Canada
| | - Anshul S. Jadli
- Department of Physiology and Pharmacology, Cumming School of Medicine, Calgary, AB, Canada
- Libin Cardiovascular Institute, Calgary, AB, Canada
| | - Luiz G. N. de Almeida
- Department of Physiology and Pharmacology, Cumming School of Medicine, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, Calgary, AB, Canada
| | - Noura N. Ballasy
- Department of Physiology and Pharmacology, Cumming School of Medicine, Calgary, AB, Canada
- Libin Cardiovascular Institute, Calgary, AB, Canada
| | - Pariya Edalat
- Department of Physiology and Pharmacology, Cumming School of Medicine, Calgary, AB, Canada
- Libin Cardiovascular Institute, Calgary, AB, Canada
| | - Ruchita Shandilya
- Department of Physiology and Pharmacology, Cumming School of Medicine, Calgary, AB, Canada
- Libin Cardiovascular Institute, Calgary, AB, Canada
| | - Daniel Young
- Department of Physiology and Pharmacology, Cumming School of Medicine, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, Calgary, AB, Canada
| | - Darrell Belke
- Libin Cardiovascular Institute, Calgary, AB, Canada
- Department of Cardiac Sciences, Cumming School of Medicine, Calgary, AB, Canada
| | - Jane Shearer
- Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Antoine Dufour
- Department of Physiology and Pharmacology, Cumming School of Medicine, Calgary, AB, Canada
- McCaig Institute for Bone and Joint Health, Calgary, AB, Canada
- Faculty of Kinesiology, University of Calgary, Calgary, AB, Canada
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Vaibhav B. Patel
- Department of Physiology and Pharmacology, Cumming School of Medicine, Calgary, AB, Canada
- Libin Cardiovascular Institute, Calgary, AB, Canada
- *Correspondence: Vaibhav B. Patel ;
| |
Collapse
|
7
|
Moritz S, Krause M, Schlatter J, Cordes N, Vehlow A. Lamellipodin-RICTOR Signaling Mediates Glioblastoma Cell Invasion and Radiosensitivity Downstream of EGFR. Cancers (Basel) 2021; 13:5337. [PMID: 34771501 PMCID: PMC8582497 DOI: 10.3390/cancers13215337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 11/19/2022] Open
Abstract
Glioblastoma is a tumor type of unmet need despite the development of multimodal treatment strategies. The main factors contributing to the poor prognosis of glioblastoma patients are diverse genetic and epigenetic changes driving glioblastoma persistence and recurrence. Complemented are these factors by extracellular cues mediated through cell surface receptors, which further aid in fostering pro-invasion and pro-survival signaling contributing to glioblastoma therapy resistance. The underlying mechanisms conferring this therapy resistance are poorly understood. Here, we show that the cytoskeleton regulator Lamellipodin (Lpd) mediates invasiveness, proliferation and radiosensitivity of glioblastoma cells. Phosphoproteome analysis identified the epidermal growth factor receptor (EGFR) signaling axis commonly hyperactive in glioblastoma to depend on Lpd. Mechanistically, EGFR signaling together with an interaction between Lpd and the Rapamycin-insensitive companion of mammalian target of rapamycin (RICTOR) jointly regulate glioblastoma radiosensitivity. Collectively, our findings demonstrate an essential function of Lpd in the radiation response and invasiveness of glioblastoma cells. Thus, we uncover a novel Lpd-driven resistance mechanism, which adds an additional critical facet to the complex glioblastoma resistance network.
Collapse
Affiliation(s)
- Stefanie Moritz
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, PF 41, 01307 Dresden, Germany; (S.M.); (N.C.)
| | - Matthias Krause
- Randall Centre of Cell and Molecular Biophysics, New Hunt’s House, Guy’s Campus, King’s College London, London SE1 1UL, UK; (M.K.); (J.S.)
| | - Jessica Schlatter
- Randall Centre of Cell and Molecular Biophysics, New Hunt’s House, Guy’s Campus, King’s College London, London SE1 1UL, UK; (M.K.); (J.S.)
| | - Nils Cordes
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, PF 41, 01307 Dresden, Germany; (S.M.); (N.C.)
- Institute of Radiooncology-OncoRay, Helmholtz-Zentrum Dresden-Rossendorf, Bautzner Landstraße 400, 01328 Dresden, Germany
- Department of Radiotherapy and Radiation Oncology, University Hospital Carl Gustav Carus, Technische Universität Dresden, Fetscherstraße 74, PF 50, 01307 Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69192 Heidelberg, Germany
| | - Anne Vehlow
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine, Technische Universität Dresden, Fetscherstraße 74, PF 41, 01307 Dresden, Germany; (S.M.); (N.C.)
- German Cancer Consortium (DKTK), Partner Site Dresden, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69192 Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Partner Site Dresden, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69192 Heidelberg, Germany
| |
Collapse
|
8
|
Zhao L, Zhu L, Oh YT, Qian G, Chen Z, Sun SY. Rictor, an essential component of mTOR complex 2, undergoes caspase-mediated cleavage during apoptosis induced by multiple stimuli. Apoptosis 2021; 26:338-347. [PMID: 33905036 DOI: 10.1007/s10495-021-01676-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/19/2021] [Indexed: 10/21/2022]
Abstract
Caspase-mediated cleavage of proteins ensures the irreversible commitment of cells to undergo apoptosis, and is thus a hallmark of apoptosis. Rapamycin-insensitive companion of mTOR (rictor) functions primarily as a core and essential component of mTOR complex 2 (mTORC2) to critically regulate cellular homeostasis. However, its role in the regulation of apoptosis is largely unknown. In the current study, we found that rictor was cleaved to generate two small fragments at ~ 50 kD and ~ 130 kD in cells undergoing apoptosis upon treatment with different stimuli such as the death ligand, TRAIL, and the small molecule, AZD9291. This cleavage was abolished when caspases were inhibited and could be reproduced when directly incubating rictor protein and caspase-3 in vitro. Furthermore, the cleavage site of caspase-3 on rictor was mapped at D1244 (VGVD). These findings together robustly demonstrate that rictor is a substrate of caspase-3 and undergoes cleavage during apoptosis. These results add new information for understanding the biology of rictor in the regulation of cell survival and growth.
Collapse
Affiliation(s)
- Liqun Zhao
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, 1365-C Clifton Road NE, Atlanta, GA, 30322, USA
| | - Lei Zhu
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, 1365-C Clifton Road NE, Atlanta, GA, 30322, USA
- Research Institute for Pharmaceutical Screening & Evaluation, Wannan Medical College School of Pharmacy and Anhui Province Key Laboratory of Active Biological Macromolecules, Wuhu, Anhui, China
| | - You-Take Oh
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, 1365-C Clifton Road NE, Atlanta, GA, 30322, USA
| | - Guoqing Qian
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, 1365-C Clifton Road NE, Atlanta, GA, 30322, USA
| | - Zhen Chen
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, 1365-C Clifton Road NE, Atlanta, GA, 30322, USA
| | - Shi-Yong Sun
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, 1365-C Clifton Road NE, Atlanta, GA, 30322, USA.
| |
Collapse
|
9
|
Jiffry J, Thavornwatanayong T, Rao D, Fogel EJ, Saytoo D, Nahata R, Guzik H, Chaudhary I, Augustine T, Goel S, Maitra R. Oncolytic Reovirus (pelareorep) Induces Autophagy in KRAS-mutated Colorectal Cancer. Clin Cancer Res 2020; 27:865-876. [PMID: 33168658 DOI: 10.1158/1078-0432.ccr-20-2385] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 09/22/2020] [Accepted: 11/04/2020] [Indexed: 12/19/2022]
Abstract
PURPOSE To explore the effects of pelareorep on autophagy in multiple models of colorectal cancer, including patient-derived peripheral blood mononuclear cells (PBMCs). EXPERIMENTAL DESIGN HCT116 [KRAS mutant (mut)] and Hke3 [KRAS wild-type (WT)] cells were treated with pelareorep (multiplicity of infection, 5) and harvested at 6 and 9 hours. LC3 A/B expression was determined by immunofluorescence and flow cytometry; five autophagic proteins were analyzed by Western blotting. The expression of 88 autophagy genes was determined by qRT-PCR. Syngeneic mouse models, CT26/Balb-C (KRAS mut) and MC38/C57B6 (KRAS WT), were developed and treated with pelareorep (10 × 106 plaque-forming unit/day) intraperitoneally. Protein and RNA were extracted from harvested tumor tissues. PBMCs from five experimental and three control patients were sampled at 0 (pre) and 48 hours, and on days 8 and 15. The gene expression normalized to "pre" was determined using 2-ΔΔC t method. RESULTS Pelareorep induced significant upregulation of LC3 A/B in HCT116 as compared with Hke3 cells by immunofluorescence (3.24 × and 8.67 ×), flow cytometry (2.37 × and 2.58 ×), and autophagosome formation (2.02 × and 1.57 ×), at 6 and 9 hours, respectively; all P < 0.05. Western blot analysis showed an increase in LC3 A/B (2.38 × and 6.82 ×) and Beclin1 (1.17 × and 1.24 ×) at 6 and 9 hours, ATG5 (2.4 ×) and P-62 (1.52 ×) at 6 hours, and VPS-34 (1.39 ×) at 9 hours (all P < 0.05). Induction of 13 transcripts in cell lines (>4 ×; 6 and 9 hours; P < 0.05), 12 transcripts in CT26 (qRT-PCR), and 14 transcripts in human PBMCs (P < 0.05) was observed. LC3 A/B, RICTOR, and RASD1 expression was upregulated in all three model systems. CONCLUSIONS Pelareorep hijacks host autophagic machinery in KRAS-mut conditions to augment its propagation and preferential oncolysis of the cancer cells.
Collapse
Affiliation(s)
| | | | - Devika Rao
- Montefiore Medical Center, Bronx, New York
| | - Elisha J Fogel
- Department of Biology, Yeshiva University, New York, New York
| | | | | | - Hillary Guzik
- Albert Einstein College of Medicine, Bronx, New York
| | | | | | - Sanjay Goel
- Albert Einstein College of Medicine, Bronx, New York. .,Montefiore Medical Center, Bronx, New York
| | - Radhashree Maitra
- Albert Einstein College of Medicine, Bronx, New York. .,Montefiore Medical Center, Bronx, New York.,Department of Biology, Yeshiva University, New York, New York
| |
Collapse
|
10
|
Hsiao WY, Jung SM, Tang Y, Haley JA, Li R, Li H, Calejman CM, Sanchez-Gurmaches J, Hung CM, Luciano AK, DeMambro V, Wellen KE, Rosen CJ, Zhu LJ, Guertin DA. The Lipid Handling Capacity of Subcutaneous Fat Is Programmed by mTORC2 during Development. Cell Rep 2020; 33:108223. [PMID: 33027655 PMCID: PMC7607535 DOI: 10.1016/j.celrep.2020.108223] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 08/12/2020] [Accepted: 09/11/2020] [Indexed: 02/08/2023] Open
Abstract
Overweight and obesity are associated with type 2 diabetes, non-alcoholic fatty liver disease, cardiovascular disease and cancer, but all fat is not equal, as storing excess lipid in subcutaneous white adipose tissue (SWAT) is more metabolically favorable than in visceral fat. Here, we uncover a critical role for mTORC2 in setting SWAT lipid handling capacity. We find that subcutaneous white preadipocytes differentiating without the essential mTORC2 subunit Rictor upregulate mature adipocyte markers but develop a striking lipid storage defect resulting in smaller adipocytes, reduced tissue size, lipid re-distribution to visceral and brown fat, and sex-distinct effects on systemic metabolic fitness. Mechanistically, mTORC2 promotes transcriptional upregulation of select lipid metabolism genes controlled by PPARγ and ChREBP, including genes that control lipid uptake, synthesis, and degradation pathways as well as Akt2, which encodes a major mTORC2 substrate and insulin effector. Further exploring this pathway may uncover new strategies to improve insulin sensitivity.
Collapse
Affiliation(s)
- Wen-Yu Hsiao
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Su Myung Jung
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Yuefeng Tang
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - John A. Haley
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Rui Li
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Huawei Li
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Camila Martinez Calejman
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Joan Sanchez-Gurmaches
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA,Division of Endocrinology, Developmental Biology, Cincinnati Children’s Hospital Research Foundation, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Chien-Min Hung
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Amelia K. Luciano
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | - Kathryn E. Wellen
- Center for Clinical and Translational Research, Maine Medical Center, Scarborough, MN 04074, USA,Department of Cancer Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Clifford J. Rosen
- Center for Clinical and Translational Research, Maine Medical Center, Scarborough, MN 04074, USA
| | - Lihua Julie Zhu
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA,Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA,Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - David A. Guertin
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA,Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01605, USA,Lead Contact,Correspondence:
| |
Collapse
|
11
|
Gkountakos A, Pilotto S, Mafficini A, Vicentini C, Simbolo M, Milella M, Tortora G, Scarpa A, Bria E, Corbo V. Unmasking the impact of Rictor in cancer: novel insights of mTORC2 complex. Carcinogenesis 2019; 39:971-980. [PMID: 29955840 DOI: 10.1093/carcin/bgy086] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 06/16/2018] [Accepted: 06/26/2018] [Indexed: 12/15/2022] Open
Abstract
Genomic alterations affecting components of the mechanistic target of rapamycin (mTOR) pathway are found rather frequently in cancers, suggesting that aberrant pathway activity is implicated in oncogenesis of different tumor types. mTOR functions as the core catalytic kinase of two distinct complexes, mTOR complex 1 (mTORC1) and 2 (mTORC2), which control numerous vital cellular processes. There is growing evidence indicating that Rictor, an essential subunit of the mTORC2 complex, is inappropriately overexpressed across numerous cancer types and this is associated with poor survival. To date, the candidate mechanisms responsible for aberrant Rictor expression described in cancer are two: (i) gene amplification and (ii) epigenetic regulation, mainly by microRNAs. Moreover, different mTOR-independent Rictor-containing complexes with oncogenic role have been documented, revealing alternative routes of Rictor-driven tumorigenesis, but simultaneously, paving the way for identifying novel biomarkers and therapeutic targets. Here, we review the main preclinical and clinical data regarding the role of Rictor in carcinogenesis and metastatic behavior as well as the potentiality of its alteration as a target.
Collapse
Affiliation(s)
- Anastasios Gkountakos
- Section of Pathology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Sara Pilotto
- Medical Oncology Section, Department of Medicine, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Andrea Mafficini
- ARC-NET Applied Research on Cancer Center, University of Verona, Verona, Italy
| | - Caterina Vicentini
- Section of Pathology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy.,ARC-NET Applied Research on Cancer Center, University of Verona, Verona, Italy
| | - Michele Simbolo
- Section of Pathology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Michele Milella
- Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Giampaolo Tortora
- Medical Oncology Section, Department of Medicine, University of Verona, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Aldo Scarpa
- Section of Pathology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy.,ARC-NET Applied Research on Cancer Center, University of Verona, Verona, Italy
| | - Emilio Bria
- Medical Oncology, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Vincenzo Corbo
- Section of Pathology, Department of Diagnostics and Public Health, University of Verona, Verona, Italy.,ARC-NET Applied Research on Cancer Center, University of Verona, Verona, Italy
| |
Collapse
|
12
|
Cota Teixeira S, Silva Lopes D, Santos da Silva M, Cordero da Luz FA, Cirilo Gimenes SN, Borges BC, Alves da Silva A, Alves Martins F, Alves Dos Santos M, Teixeira TL, Oliveira RA, de Melo Rodrigues Ávila V, Barbosa Silva MJ, Elias MC, Martin R, Vieira da Silva C, Knölker HJ. Pentachloropseudilin Impairs Angiogenesis by Disrupting the Actin Cytoskeleton, Integrin Trafficking and the Cell Cycle. Chembiochem 2019; 20:2390-2401. [PMID: 31026110 DOI: 10.1002/cbic.201900203] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Indexed: 12/21/2022]
Abstract
Class 1 myosins (Myo1s) were the first unconventional myosins identified and humans have eight known Myo1 isoforms. The Myo1 family is involved in the regulation of gene expression, cytoskeletal rearrangements, delivery of proteins to the cell surface, cell migration and spreading. Thus, the important role of Myo1s in different biological processes is evident. In this study, we have investigated the effects of pentachloropseudilin (PClP), a reversible and allosteric potent inhibitor of Myo1s, on angiogenesis. We demonstrated that treatment of cells with PClP promoted a decrease in the number of vessels. The observed inhibition of angiogenesis is likely to be related to the inhibition of cell proliferation, migration and adhesion, as well as to alteration of the actin cytoskeleton pattern, as shown on a PClP-treated HUVEC cell line. Moreover, we also demonstrated that PClP treatment partially prevented the delivery of integrins to the plasma membrane. Finally, we showed that PClP caused DNA strand breaks, which are probably repaired during the cell cycle arrest in the G1 phase. Taken together, our results suggest that Myo1s participate directly in the angiogenesis process.
Collapse
Affiliation(s)
- Samuel Cota Teixeira
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Daiana Silva Lopes
- Multidisciplinary Institute of Health, Anísio Teixeira Campus, Federal University of Bahia, Rua Hormindo Barros, 58, Candeias, Vitória da Conquista, 45029-094, BA, Brazil
| | - Marcelo Santos da Silva
- Special Laboratory of Cell Cycle (LECC), Center of Toxins, Immune Response and Cell Signaling (CeTICS), Butantan Institute, Av. Vital Brasil, 1500 - Butantã, São Paulo, 05503-900, SP, Brazil.,The Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, Sir Graeme Davies Building, University of Glasgow, 120 University Place, Glasgow, G12 8TA, UK
| | - Felipe Andrés Cordero da Luz
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Sarah Natalie Cirilo Gimenes
- Imunopathology Laboratory, Butantan Institute, Av. Vital Brasil, 1500 - Butantã, São Paulo, 05503-900, SP, Brazil
| | - Bruna Cristina Borges
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Aline Alves da Silva
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Flávia Alves Martins
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Marlus Alves Dos Santos
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Thaise Lara Teixeira
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Ricardo A Oliveira
- Medical School, Federal University of Uberlândia, Av. Pará, Bloco 2u, 1720 - Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Veridiana de Melo Rodrigues Ávila
- Institute of Biotechnology, Federal University of Uberlândia, Av. Pará, 1720 - Bloco 2E - Sala(s) 246 - Campus Umuarama, Uberlândia, 38405-320, MG, Brazil
| | - Marcelo José Barbosa Silva
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Maria Carolina Elias
- Special Laboratory of Cell Cycle (LECC), Center of Toxins, Immune Response and Cell Signaling (CeTICS), Butantan Institute, Av. Vital Brasil, 1500 - Butantã, São Paulo, 05503-900, SP, Brazil
| | - René Martin
- Fakultät Chemie, Technische Universität Dresden, Bergstraße 66, 01069, Dresden, Germany
| | - Claudio Vieira da Silva
- Department of Immunology, Biomedical Sciences Institute, Federal University of Uberlândia, Rua Piauí, Bloco 2B, sala 200, Campus Umuarama, Uberlândia, 38400-902, MG, Brazil
| | - Hans-Joachim Knölker
- Fakultät Chemie, Technische Universität Dresden, Bergstraße 66, 01069, Dresden, Germany
| |
Collapse
|
13
|
Yu Y, Xiong Y, Ladeiras D, Yang Z, Ming XF. Myosin 1b Regulates Nuclear AKT Activation by Preventing Localization of PTEN in the Nucleus. iScience 2019; 19:39-53. [PMID: 31349190 PMCID: PMC6660601 DOI: 10.1016/j.isci.2019.07.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 05/17/2019] [Accepted: 07/05/2019] [Indexed: 02/08/2023] Open
Abstract
Insulin-induced AKT activation is dependent on phosphoinositide 3-kinase and opposed by tumor suppressor phosphatase and tensin homolog (PTEN). Our previous study demonstrates that myosin 1b (MYO1B) mediates arginase-II-induced activation of mechanistic target of rapamycin complex 1 that is regulated by AKT. However, the role of MYO1B in AKT activation is unknown. Here we show that silencing MYO1B in mouse embryonic fibroblasts (MEF) inhibits insulin-induced nuclear but not cytoplasmic AKT activation accompanied by elevated nuclear PTEN level. Co-immunoprecipitation, co-immunostaining, and proximity ligation assay show an interaction of MYO1B and PTEN resulting in reduced nuclear PTEN. Moreover, the elevated nuclear PTEN upon silencing MYO1B promotes apoptosis of MEFs and melanoma B16F10 cells. Taken together, we demonstrate that MYO1B, by interacting with PTEN, prevents nuclear localization of PTEN contributing to nuclear AKT activation and suppression of cell apoptosis. This may present a therapeutic approach for cancer treatment such as melanoma. MYO1B, by interacting with PTEN, prevents PTEN localization in the nucleus MYO1B prevents nuclear localization of PTEN depending on its motor activity This contributes to nuclear AKT activation and suppression of cell apoptosis Targeting MYO1B may represent a therapeutic approach for cancer treatment
Collapse
Affiliation(s)
- Yi Yu
- Cardiovascular and Aging Research, Department of Endocrinology, Metabolism and Cardiovascular System, Medicine Section, Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 5, 1700 Fribourg, Switzerland
| | - Yuyan Xiong
- Cardiovascular and Aging Research, Department of Endocrinology, Metabolism and Cardiovascular System, Medicine Section, Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 5, 1700 Fribourg, Switzerland
| | - Diogo Ladeiras
- Cardiovascular and Aging Research, Department of Endocrinology, Metabolism and Cardiovascular System, Medicine Section, Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 5, 1700 Fribourg, Switzerland
| | - Zhihong Yang
- Cardiovascular and Aging Research, Department of Endocrinology, Metabolism and Cardiovascular System, Medicine Section, Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 5, 1700 Fribourg, Switzerland.
| | - Xiu-Fen Ming
- Cardiovascular and Aging Research, Department of Endocrinology, Metabolism and Cardiovascular System, Medicine Section, Faculty of Science and Medicine, University of Fribourg, Chemin du Musée 5, 1700 Fribourg, Switzerland.
| |
Collapse
|
14
|
Wright KD, Miller BS, El-Meanawy S, Tsaih SW, Banerjee A, Geurts AM, Sheinin Y, Sun Y, Kalyanaraman B, Rui H, Flister MJ, Sorokin A. The p52 isoform of SHC1 is a key driver of breast cancer initiation. Breast Cancer Res 2019; 21:74. [PMID: 31202267 PMCID: PMC6570928 DOI: 10.1186/s13058-019-1155-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 05/23/2019] [Indexed: 01/23/2023] Open
Abstract
Background SHC1 proteins (also called SHCA) exist in three functionally distinct isoforms (p46SHC, p52SHC, and p66SHC) that serve as intracellular adaptors for several key signaling pathways in breast cancer. Despite the broad evidence implicating SHC1 gene products as a central mediator of breast cancer, testing the isoform-specific roles of SHC1 proteins have been inaccessible due to the lack of isoform-specific inhibitors or gene knockout models. Methods Here, we addressed this issue by generating the first isoform-specific gene knockout models for p52SHC and p66SHC, using germline gene editing in the salt-sensitive rat strain. Compared with the wild-type (WT) rats, we found that genetic ablation of the p52SHC isoform significantly attenuated mammary tumor formation, whereas the p66SHC knockout had no effect. Rats were dosed with 7,12-dimethylbenz(a)anthracene (DMBA) by oral gavage to induce mammary tumors, and progression of tumor development was followed for 15 weeks. At 15 weeks, tumors were excised and analyzed by RNA-seq to determine differences between tumors lacking p66SHC or p52SHC. Results Compared with the wild-type (WT) rats, we found that genetic ablation of the p52SHC isoform significantly attenuated mammary tumor formation, whereas the p66SHC knockout had no effect. These data, combined with p52SHC being the predominant isoform that is upregulated in human and rat tumors, provide the first evidence that p52SHC is the oncogenic isoform of Shc1 gene products in breast cancer. Compared with WT tumors, 893 differentially expressed (DE; FDR < 0.05) genes were detected in p52SHC KO tumors compared with only 18 DE genes in the p66SHC KO tumors, further highlighting that p52SHC is the relevant SHC1 isoform in breast cancer. Finally, gene network analysis revealed that p52SHC KO disrupted multiple key pathways that have been previously implicated in breast cancer initiation and progression, including ESR1 and mTORC2/RICTOR. Conclusion Collectively, these data demonstrate the p52SHC isoform is the key driver of DMBA-induced breast cancer while the expression of p66SHC and p46SHC are not enough to compensate. Electronic supplementary material The online version of this article (doi:10.1186/s13058-019-1155-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kevin D Wright
- Cardiovascular Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Department of Medicine, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Bradley S Miller
- Cardiovascular Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Department of Medicine, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Sarah El-Meanawy
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Free Radical Research Center, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Shirng-Wern Tsaih
- Cardiovascular Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Anjishnu Banerjee
- Institute for Health and Equity, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Aron M Geurts
- Cardiovascular Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Yuri Sheinin
- Department of Pathology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Yunguang Sun
- Department of Pathology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Balaraman Kalyanaraman
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Free Radical Research Center, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Hallgeir Rui
- Department of Pathology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Michael J Flister
- Cardiovascular Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Department of Physiology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Andrey Sorokin
- Cardiovascular Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA. .,Department of Medicine, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| |
Collapse
|
15
|
Pergolizzi B, Panuzzo C, Ali MS, Lo Iacono M, Levron CL, Ponzone L, Prelli M, Cilloni D, Calautti E, Bozzaro S, Bracco E. Mammals and Dictyostelium rictor mutations swapping reveals two essential Gly residues for mTORC2 activity and integrity. J Cell Sci 2019; 132:jcs.236505. [DOI: 10.1242/jcs.236505] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 10/15/2019] [Indexed: 12/27/2022] Open
Abstract
mTORC2 regulates a variety of vital cellular processes, and its aberrant functioning is often associated with various diseases. Rictor is a peculiar and distinguishing mTORC2 component playing a pivotal role in controlling its assembly and activity. Among living organisms Rictor is conserved from unicellular eukaryotes to metazoan. We replaced two distinct, but conserved, glycines in both the Dictyostelium piaA gene and its human ortholog, rictor. The two conserved residues are spaced by approximately 50 aminoacids and both are embedded within a conserved region falling in between the Ras-GEFN2 and Rictor_V domains. The effects of point mutations on the mTORC2 activity and integrity were assessed by biochemical and functional assays.In both cases, the reciprocal exchange between mammals and Dictyostelium rictor and piaA gene point mutations impaired mTORC2 activity and integrity.Our data indicate that the two Gly residues are essential for the maintenance of mTORC2 activity and integrity in organisms that appear to be distantly related, suggesting a primeval role in the assembly and proper TOR complex 2 functioning.
Collapse
Affiliation(s)
- Barbara Pergolizzi
- Department of Clinical and Biological Sciences, University of Torino, AOU S. Luigi, 10043 Orbassano (TO), Italy
| | - Cristina Panuzzo
- Department of Clinical and Biological Sciences, University of Torino, AOU S. Luigi, 10043 Orbassano (TO), Italy
| | - M. Shahzad Ali
- Department of Clinical and Biological Sciences, University of Torino, AOU S. Luigi, 10043 Orbassano (TO), Italy
| | - Marco Lo Iacono
- Department of Clinical and Biological Sciences, University of Torino, AOU S. Luigi, 10043 Orbassano (TO), Italy
| | - Chiara Levra Levron
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Centre, University of Torino, Via Nizza 52, Torino, Italy
| | - Luca Ponzone
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Centre, University of Torino, Via Nizza 52, Torino, Italy
| | - Marta Prelli
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Centre, University of Torino, Via Nizza 52, Torino, Italy
| | - Daniela Cilloni
- Department of Clinical and Biological Sciences, University of Torino, AOU S. Luigi, 10043 Orbassano (TO), Italy
| | - Enzo Calautti
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Centre, University of Torino, Via Nizza 52, Torino, Italy
| | - Salvatore Bozzaro
- Department of Clinical and Biological Sciences, University of Torino, AOU S. Luigi, 10043 Orbassano (TO), Italy
| | - Enrico Bracco
- Department of Oncology, University of Torino, AOU S. Luigi, 10043 Orbassano (TO), Italy
| |
Collapse
|
16
|
Dissanayake WC, Sorrenson B, Cognard E, Hughes WE, Shepherd PR. β-catenin is important for the development of an insulin responsive pool of GLUT4 glucose transporters in 3T3-L1 adipocytes. Exp Cell Res 2018. [PMID: 29540328 DOI: 10.1016/j.yexcr.2018.03.011] [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] [Indexed: 01/01/2023]
Abstract
GLUT4 is unique among specialized glucose transporters in being exclusively expressed in muscle and adipocytes. In the absence of insulin the distribution of GLUT4 is preferentially intracellular and insulin stimulation results in the movement of GLUT4 containing vesicles to the plasma membrane. This process is responsible for the insulin stimulation of glucose uptake in muscle and fat. While signalling pathways triggering the translocation of GLUT4 are well understood, the mechanisms regulating the intracellular retention of GLUT4 are less well understood. Here we report a role for β-catenin in this process. In 3T3-L1 adipocytes in which β-catenin is depleted, the levels of GLUT4 at and near the plasma membrane rise in unstimulated cells while the subsequent increase in GLUT4 at the plasma membrane upon insulin stimulation is reduced. Small molecule approaches to acutely activate or inhibit β-catenin give results that support the results obtained with siRNA and these changes are accompanied by matching changes in glucose transport into these cells. Together these results indicate that β-catenin is a previously unrecognized regulator of the mechanisms that control the insulin sensitive pool of GLUT4 transporters inside these adipocyte cells.
Collapse
Affiliation(s)
- Waruni C Dissanayake
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Brie Sorrenson
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Emmanuelle Cognard
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - William E Hughes
- Department of Medicine, St. Vincent's Hospital, Victoria Street, Sydney 2010, Australia; The Garvan Institute of Medical Research, 384 Victoria Street, Sydney 2010, Australia
| | - Peter R Shepherd
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand.
| |
Collapse
|
17
|
Jaldin-Fincati JR, Pavarotti M, Frendo-Cumbo S, Bilan PJ, Klip A. Update on GLUT4 Vesicle Traffic: A Cornerstone of Insulin Action. Trends Endocrinol Metab 2017; 28:597-611. [PMID: 28602209 DOI: 10.1016/j.tem.2017.05.002] [Citation(s) in RCA: 196] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 05/08/2017] [Accepted: 05/09/2017] [Indexed: 12/20/2022]
Abstract
Glucose transport is rate limiting for dietary glucose utilization by muscle and fat. The glucose transporter GLUT4 is dynamically sorted and retained intracellularly and redistributes to the plasma membrane (PM) by insulin-regulated vesicular traffic, or 'GLUT4 translocation'. Here we emphasize recent findings in GLUT4 translocation research. The application of total internal reflection fluorescence microscopy (TIRFM) has increased our understanding of insulin-regulated events beneath the PM, such as vesicle tethering and membrane fusion. We describe recent findings on Akt-targeted Rab GTPase-activating proteins (GAPs) (TBC1D1, TBC1D4, TBC1D13) and downstream Rab GTPases (Rab8a, Rab10, Rab13, Rab14, and their effectors) along with the input of Rac1 and actin filaments, molecular motors [myosinVa (MyoVa), myosin1c (Myo1c), myosinIIA (MyoIIA)], and membrane fusion regulators (syntaxin4, munc18c, Doc2b). Collectively these findings reveal novel events in insulin-regulated GLUT4 traffic.
Collapse
Affiliation(s)
| | - Martin Pavarotti
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5J 2L4, Canada; IHEM, Universidad Nacional de Cuyo, CONICET, Mendoza 5500, Argentina
| | - Scott Frendo-Cumbo
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5J 2L4, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Philip J Bilan
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5J 2L4, Canada
| | - Amira Klip
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5J 2L4, Canada; Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada; Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada.
| |
Collapse
|
18
|
Benavides-Serrato A, Lee J, Holmes B, Landon KA, Bashir T, Jung ME, Lichtenstein A, Gera J. Specific blockade of Rictor-mTOR association inhibits mTORC2 activity and is cytotoxic in glioblastoma. PLoS One 2017; 12:e0176599. [PMID: 28453552 PMCID: PMC5409528 DOI: 10.1371/journal.pone.0176599] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 04/13/2017] [Indexed: 12/19/2022] Open
Abstract
A small molecule which specifically blocks the interaction of Rictor and mTOR was identified utilizing a high-throughput yeast two-hybrid screen and evaluated as a potential inhibitor of mTORC2 activity in glioblastoma multiforme (GBM). In vitro, CID613034 inhibited mTORC2 kinase activity at submicromolar concentrations and in cellular assays specifically inhibited phosphorylation of mTORC2 substrates, including AKT (Ser-473), NDRG1 (Thr-346) and PKCα (Ser-657), while having no appreciable effects on the phosphorylation status of the mTORC1 substrate S6K (Thr-389) or mTORC1-dependent negative feedback loops. CID613034 demonstrated significant inhibitory effects on cell growth, motility and invasiveness in GBM cell lines and sensitivity correlated with relative Rictor or SIN1 expression. Structure-activity relationship analyses afforded an inhibitor, JR-AB2-011, with improved anti-GBM properties and blocked mTORC2 signaling and Rictor association with mTOR at lower effective concentrations. In GBM xenograft studies, JR-AB2-011 demonstrated significant anti-tumor properties. These data support mTORC2 as a viable therapeutic target in GBM and suggest that targeting protein-protein interactions critical for mTORC2 function is an effective strategy to achieve therapeutic responses.
Collapse
Affiliation(s)
- Angelica Benavides-Serrato
- Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California, United States of America
- Department of Research & Development, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, California, United States of America
| | - Jihye Lee
- Department of Chemistry and Biochemistry, University of California-Los Angeles, Los Angeles, California, United States of America
| | - Brent Holmes
- Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California, United States of America
- Department of Research & Development, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, California, United States of America
| | - Kenna A. Landon
- Department of Research & Development, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, California, United States of America
| | - Tariq Bashir
- Department of Research & Development, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, California, United States of America
| | - Michael E. Jung
- Department of Chemistry and Biochemistry, University of California-Los Angeles, Los Angeles, California, United States of America
- Jonnson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, California, United States of America
- Molecular Biology Institute, University of California-Los Angeles, Los Angeles, California, United States of America
| | - Alan Lichtenstein
- Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California, United States of America
- Department of Research & Development, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, California, United States of America
- Jonnson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, California, United States of America
| | - Joseph Gera
- Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California, United States of America
- Department of Research & Development, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, California, United States of America
- Jonnson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, California, United States of America
- Molecular Biology Institute, University of California-Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
19
|
Ong PS, Wang LZ, Dai X, Tseng SH, Loo SJ, Sethi G. Judicious Toggling of mTOR Activity to Combat Insulin Resistance and Cancer: Current Evidence and Perspectives. Front Pharmacol 2016; 7:395. [PMID: 27826244 PMCID: PMC5079084 DOI: 10.3389/fphar.2016.00395] [Citation(s) in RCA: 122] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 10/07/2016] [Indexed: 12/16/2022] Open
Abstract
The mechanistic target of rapamycin (mTOR), via its two distinct multiprotein complexes, mTORC1, and mTORC2, plays a central role in the regulation of cellular growth, metabolism, and migration. A dysregulation of the mTOR pathway has in turn been implicated in several pathological conditions including insulin resistance and cancer. Overactivation of mTORC1 and disruption of mTORC2 function have been reported to induce insulin resistance. On the other hand, aberrant mTORC1 and mTORC2 signaling via either genetic alterations or increased expression of proteins regulating mTOR and its downstream targets have contributed to cancer development. These underlined the attractiveness of mTOR as a therapeutic target to overcome both insulin resistance and cancer. This review summarizes the evidence supporting the notion of intermittent, low dose rapamycin for treating insulin resistance. It further highlights recent data on the continuous use of high dose rapamycin analogs and related second generation mTOR inhibitors for cancer eradication, for overcoming chemoresistance and for tumor stem cell suppression. Within these contexts, the potential challenges associated with the use of mTOR inhibitors are also discussed.
Collapse
Affiliation(s)
- Pei Shi Ong
- Department of Pharmacy, Faculty of Science, National University of Singapore Singapore, Singapore
| | - Louis Z Wang
- Department of Pharmacy, Faculty of Science, National University of SingaporeSingapore, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of SingaporeSingapore, Singapore
| | - Xiaoyun Dai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore Singapore, Singapore
| | - Sheng Hsuan Tseng
- Department of Pharmacy, Faculty of Science, National University of Singapore Singapore, Singapore
| | - Shang Jun Loo
- Department of Pharmacy, Faculty of Science, National University of Singapore Singapore, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore Singapore, Singapore
| |
Collapse
|
20
|
Lai P, Song Q, Yang C, Li Z, Liu S, Liu B, Li M, Deng H, Cai D, Jin D, Liu A, Bai X. Loss of Rictor with aging in osteoblasts promotes age-related bone loss. Cell Death Dis 2016; 7:e2408. [PMID: 27735936 PMCID: PMC5133960 DOI: 10.1038/cddis.2016.249] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 07/03/2016] [Accepted: 07/05/2016] [Indexed: 01/16/2023]
Abstract
Osteoblast dysfunction is a major cause of age-related bone loss, but the mechanisms underlying changes in osteoblast function with aging are poorly understood. This study demonstrates that osteoblasts in aged mice exhibit markedly impaired adhesion to the bone formation surface and reduced mineralization in vivo and in vitro. Rictor, a specific component of the mechanistic target of rapamycin complex 2 (mTORC2) that controls cytoskeletal organization and cell survival, is downregulated with aging in osteoblasts. Mechanistically, we found that an increased level of reactive oxygen species with aging stimulates the expression of miR-218, which directly targets Rictor and reduces osteoblast bone surface adhesion and survival, resulting in a decreased number of functional osteoblasts and accelerated bone loss in aged mice. Our findings reveal a novel functional pathway important for age-related bone loss and support for miR-218 and Rictor as potential targets for therapeutic intervention for age-related osteoporosis treatment.
Collapse
Affiliation(s)
- Pinling Lai
- Academy of Orthopedics in Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China.,State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Qiancheng Song
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.,Department of Biochemistry, Institute of Genetic Engineering, Southern Medical University, Guangzhou 510515, China
| | - Cheng Yang
- Academy of Orthopedics in Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Zhen Li
- Academy of Orthopedics in Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Sichi Liu
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Bin Liu
- Department of Spine Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | - Mangmang Li
- State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Hongwen Deng
- Academy of Orthopedics in Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Daozhang Cai
- Academy of Orthopedics in Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Dadi Jin
- Academy of Orthopedics in Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Anling Liu
- Department of Biochemistry, Institute of Genetic Engineering, Southern Medical University, Guangzhou 510515, China
| | - Xiaochun Bai
- Academy of Orthopedics in Guangdong Province, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China.,State Key Laboratory of Organ Failure Research, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
21
|
Visuttijai K, Pettersson J, Mehrbani Azar Y, van den Bout I, Örndal C, Marcickiewicz J, Nilsson S, Hörnquist M, Olsson B, Ejeskär K, Behboudi A. Lowered Expression of Tumor Suppressor Candidate MYO1C Stimulates Cell Proliferation, Suppresses Cell Adhesion and Activates AKT. PLoS One 2016; 11:e0164063. [PMID: 27716847 PMCID: PMC5055341 DOI: 10.1371/journal.pone.0164063] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2016] [Accepted: 09/18/2016] [Indexed: 12/12/2022] Open
Abstract
Myosin-1C (MYO1C) is a tumor suppressor candidate located in a region of recurrent losses distal to TP53. Myo1c can tightly and specifically bind to PIP2, the substrate of Phosphoinositide 3-kinase (PI3K), and to Rictor, suggesting a role for MYO1C in the PI3K pathway. This study was designed to examine MYO1C expression status in a panel of well-stratified endometrial carcinomas as well as to assess the biological significance of MYO1C as a tumor suppressor in vitro. We found a significant correlation between the tumor stage and lowered expression of MYO1C in endometrial carcinoma samples. In cell transfection experiments, we found a negative correlation between MYO1C expression and cell proliferation, and MYO1C silencing resulted in diminished cell migration and adhesion. Cells expressing excess of MYO1C had low basal level of phosphorylated protein kinase B (PKB, a.k.a. AKT) and cells with knocked down MYO1C expression showed a quicker phosphorylated AKT (pAKT) response in reaction to serum stimulation. Taken together the present study gives further evidence for tumor suppressor activity of MYO1C and suggests MYO1C mediates its tumor suppressor function through inhibition of PI3K pathway and its involvement in loss of contact inhibition.
Collapse
Affiliation(s)
- Kittichate Visuttijai
- School of Bioscience, Tumor Biology research group, University of Skövde, SE-541 28, Skövde, Sweden
- Department of Medical and Clinical Genetics, Sahlgrenska Academy, University of Gothenburg, SE-405 30, Gothenburg, Sweden
| | - Jennifer Pettersson
- Department of Medical and Clinical Genetics, Sahlgrenska Academy, University of Gothenburg, SE-405 30, Gothenburg, Sweden
| | - Yashar Mehrbani Azar
- School of Bioscience, Tumor Biology research group, University of Skövde, SE-541 28, Skövde, Sweden
| | - Iman van den Bout
- Department of physiology, Faculty of Health Sciences, University of Pretoria, Pretoria, 0007, South Africa
| | - Charlotte Örndal
- Department of Pathology, Sahlgrenska University Hospital, SE-413 45, Gothenburg, Sweden
| | - Janusz Marcickiewicz
- Department of Obstetrics and Gynecology, Halland Hospital Varberg, SE- 432 37, Varberg, Sweden
| | - Staffan Nilsson
- Institute of Mathematical Statistics, Chalmers University of Technology, SE-412 96, Gothenburg, Sweden
| | - Michael Hörnquist
- Department of Science and Technology, University of Linköping, ITN, SE-601 74, Norrköping, Sweden
| | - Björn Olsson
- School of Bioscience, Tumor Biology research group, University of Skövde, SE-541 28, Skövde, Sweden
| | - Katarina Ejeskär
- School of Bioscience, Tumor Biology research group, University of Skövde, SE-541 28, Skövde, Sweden
| | - Afrouz Behboudi
- School of Bioscience, Tumor Biology research group, University of Skövde, SE-541 28, Skövde, Sweden
- * E-mail:
| |
Collapse
|
22
|
Venit T, Kalendová A, Petr M, Dzijak R, Pastorek L, Rohožková J, Malohlava J, Hozák P. Nuclear myosin I regulates cell membrane tension. Sci Rep 2016; 6:30864. [PMID: 27480647 PMCID: PMC4969604 DOI: 10.1038/srep30864] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 07/12/2016] [Indexed: 11/09/2022] Open
Abstract
Plasma membrane tension is an important feature that determines the cell shape and influences processes such as cell motility, spreading, endocytosis and exocytosis. Unconventional class 1 myosins are potent regulators of plasma membrane tension because they physically link the plasma membrane with adjacent cytoskeleton. We identified nuclear myosin 1 (NM1) - a putative nuclear isoform of myosin 1c (Myo1c) - as a new player in the field. Although having specific nuclear functions, NM1 localizes predominantly to the plasma membrane. Deletion of NM1 causes more than a 50% increase in the elasticity of the plasma membrane around the actin cytoskeleton as measured by atomic force microscopy. This higher elasticity of NM1 knock-out cells leads to 25% higher resistance to short-term hypotonic environment and rapid cell swelling. In contrast, overexpression of NM1 in wild type cells leads to an additional 30% reduction of their survival. We have shown that NM1 has a direct functional role in the cytoplasm as a dynamic linker between the cell membrane and the underlying cytoskeleton, regulating the degree of effective plasma membrane tension.
Collapse
Affiliation(s)
- Tomáš Venit
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics, AS CR, v.v.i., Videnska 1083, 142 20 Prague, Czech Republic.,Faculty of Science, Charles University in Prague, Albertov 6, 128 43 Prague, Czech Republic
| | - Alžběta Kalendová
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics, AS CR, v.v.i., Videnska 1083, 142 20 Prague, Czech Republic
| | - Martin Petr
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics, AS CR, v.v.i., Videnska 1083, 142 20 Prague, Czech Republic
| | - Rastislav Dzijak
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics, AS CR, v.v.i., Videnska 1083, 142 20 Prague, Czech Republic
| | - Lukáš Pastorek
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics, AS CR, v.v.i., Videnska 1083, 142 20 Prague, Czech Republic
| | - Jana Rohožková
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics, AS CR, v.v.i., Videnska 1083, 142 20 Prague, Czech Republic
| | - Jakub Malohlava
- Department of Medical Biophysics, Faculty of Medicine and Dentistry, Palacky University in Olomouc, Hnevotinska 3, 775 15 Olomouc, Czech Republic
| | - Pavel Hozák
- Department of Biology of the Cell Nucleus, Institute of Molecular Genetics, AS CR, v.v.i., Videnska 1083, 142 20 Prague, Czech Republic
| |
Collapse
|
23
|
Structural Analysis of the Myo1c and Neph1 Complex Provides Insight into the Intracellular Movement of Neph1. Mol Cell Biol 2016; 36:1639-54. [PMID: 27044863 DOI: 10.1128/mcb.00020-16] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 03/21/2016] [Indexed: 11/20/2022] Open
Abstract
The Myo1c motor functions as a cargo transporter supporting various cellular events, including vesicular trafficking, cell migration, and stereociliary movements of hair cells. Although its partial crystal structures were recently described, the structural details of its interaction with cargo proteins remain unknown. This study presents the first structural demonstration of a cargo protein, Neph1, attached to Myo1c, providing novel insights into the role of Myo1c in intracellular movements of this critical slit diaphragm protein. Using small angle X-ray scattering studies, models of predominant solution conformation of unliganded full-length Myo1c and Myo1c bound to Neph1 were constructed. The resulting structures show an extended S-shaped Myo1c with Neph1 attached to its C-terminal tail. Importantly, binding of Neph1 did not induce a significant shape change in Myo1c, indicating this as a spontaneous process or event. Analysis of interaction surfaces led to the identification of a critical residue in Neph1 involved in binding to Myo1c. Indeed, a point mutant from this site abolished interaction between Neph1 and Myo1c when tested in the in vitro and in live-cell binding assays. Live-cell imaging, including fluorescence recovery after photobleaching, provided further support for the role of Myo1c in intracellular vesicular movement of Neph1 and its turnover at the membrane.
Collapse
|
24
|
Both mTORC1 and mTORC2 are involved in the regulation of cell adhesion. Oncotarget 2016; 6:7136-50. [PMID: 25762619 PMCID: PMC4466674 DOI: 10.18632/oncotarget.3044] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 01/07/2015] [Indexed: 01/11/2023] Open
Abstract
mTOR is a central controller for cell growth/proliferation and survival. Recent studies have shown that mTOR also regulates cell adhesion, yet the underlying mechanism is not known. Here we found that inhibition of mTOR by rapamycin reduced the basal or type I insulin-like growth factor (IGF-1)-stimulated adhesion of cancer cells. Further research revealed that both mTORC1 and mTORC2 were involved in the regulation of cell adhesion, as silencing expression of raptor or rictor inhibited cell adhesion. Also, PP242, an mTORC1/2 kinase inhibitor, inhibited cell adhesion more potently than rapamycin (mTORC1 inhibitor). Of interest, ectopic expression of constitutively active and rapamycin-resistant mutant of p70 kinase 1 (S6K1) or downregulation of eukaryotic initiation factor 4E (eIF4E)-binding protein 1 (4E-BP1) conferred resistance to rapamycin inhibition of cell adhesion, whereas expression of constitutively hypophosphorylated 4E-BP1 (4EBP1-5A) or downregulation of S6K1 suppressed cell adhesion. In contrast, neither genetic manipulation of Akt activity nor pharmacological inhibition of Akt affected cell adhesion. The results suggest that both mTORC1 and mTORC2 are involved in the regulation of cell adhesion; and mTORC1 regulates cell adhesion through S6K1 and 4E-BP1 pathways, but mTORC2 regulates cell adhesion via Akt-independent mechanism.
Collapse
|
25
|
Dong H, Chen Z, Wang C, Xiong Z, Zhao W, Jia C, Lin J, Lin Y, Yuan W, Zhao AZ, Bai X. Rictor Regulates Spermatogenesis by Controlling Sertoli Cell Cytoskeletal Organization and Cell Polarity in the Mouse Testis. Endocrinology 2015; 156:4244-56. [PMID: 26360620 DOI: 10.1210/en.2015-1217] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Maintenance of cell polarity is essential for Sertoli cell and blood-testis barrier (BTB) function and spermatogenesis; however, the signaling mechanisms that regulate the integrity of the cytoskeleton and polarity of Sertoli cells are not fully understood. Here, we demonstrate that rapamycin-insensitive component of target of rapamycin (TOR) (Rictor), a core component of mechanistic TOR complex 2 (mTORC2), was expressed in the seminiferous epithelium during testicular development, and was down-regulated in a cadmium chloride-induced BTB damage model. We then conditionally deleted the Rictor gene in Sertoli cells and mutant mice exhibited azoospermia and were sterile as early as 3 months old. Further study revealed that Rictor may regulate actin organization via both mTORC2-dependent and mTORC2-independent mechanisms, in which the small GTPase, ras-related C3 botulinum toxin substrate 1, and phosphorylation of the actin filament regulatory protein, Paxillin, are involved, respectively. Loss of Rictor in Sertoli cells perturbed actin dynamics and caused microtubule disarrangement, both of which accumulatively disrupted Sertoli cell polarity and BTB integrity, accompanied by testicular developmental defects, spermiogenic arrest and excessive germ cell loss in mutant mice. Together, these findings establish the importance of Rictor/mTORC2 signaling in Sertoli cell function and spermatogenesis through the maintenance of Sertoli cell cytoskeletal dynamics, BTB integrity, and cell polarity.
Collapse
Affiliation(s)
- Heling Dong
- State Key Laboratory of Organ Failure Research (H.D., Z.C., C.W., Z.X., W.Z., C.J., J.L., X.B.), Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; State Key Laboratory of Reproductive Medicine (L.Y., A.Z.Z.), The Center of Metabolic Disease Research, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China; and State Key Laboratory of Experimental Hematology (W.Y.), Institute of Hematology; and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Zhenguo Chen
- State Key Laboratory of Organ Failure Research (H.D., Z.C., C.W., Z.X., W.Z., C.J., J.L., X.B.), Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; State Key Laboratory of Reproductive Medicine (L.Y., A.Z.Z.), The Center of Metabolic Disease Research, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China; and State Key Laboratory of Experimental Hematology (W.Y.), Institute of Hematology; and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Caixia Wang
- State Key Laboratory of Organ Failure Research (H.D., Z.C., C.W., Z.X., W.Z., C.J., J.L., X.B.), Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; State Key Laboratory of Reproductive Medicine (L.Y., A.Z.Z.), The Center of Metabolic Disease Research, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China; and State Key Laboratory of Experimental Hematology (W.Y.), Institute of Hematology; and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Zhi Xiong
- State Key Laboratory of Organ Failure Research (H.D., Z.C., C.W., Z.X., W.Z., C.J., J.L., X.B.), Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; State Key Laboratory of Reproductive Medicine (L.Y., A.Z.Z.), The Center of Metabolic Disease Research, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China; and State Key Laboratory of Experimental Hematology (W.Y.), Institute of Hematology; and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Wanlu Zhao
- State Key Laboratory of Organ Failure Research (H.D., Z.C., C.W., Z.X., W.Z., C.J., J.L., X.B.), Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; State Key Laboratory of Reproductive Medicine (L.Y., A.Z.Z.), The Center of Metabolic Disease Research, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China; and State Key Laboratory of Experimental Hematology (W.Y.), Institute of Hematology; and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Chunhong Jia
- State Key Laboratory of Organ Failure Research (H.D., Z.C., C.W., Z.X., W.Z., C.J., J.L., X.B.), Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; State Key Laboratory of Reproductive Medicine (L.Y., A.Z.Z.), The Center of Metabolic Disease Research, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China; and State Key Laboratory of Experimental Hematology (W.Y.), Institute of Hematology; and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Jun Lin
- State Key Laboratory of Organ Failure Research (H.D., Z.C., C.W., Z.X., W.Z., C.J., J.L., X.B.), Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; State Key Laboratory of Reproductive Medicine (L.Y., A.Z.Z.), The Center of Metabolic Disease Research, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China; and State Key Laboratory of Experimental Hematology (W.Y.), Institute of Hematology; and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Yan Lin
- State Key Laboratory of Organ Failure Research (H.D., Z.C., C.W., Z.X., W.Z., C.J., J.L., X.B.), Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; State Key Laboratory of Reproductive Medicine (L.Y., A.Z.Z.), The Center of Metabolic Disease Research, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China; and State Key Laboratory of Experimental Hematology (W.Y.), Institute of Hematology; and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Weiping Yuan
- State Key Laboratory of Organ Failure Research (H.D., Z.C., C.W., Z.X., W.Z., C.J., J.L., X.B.), Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; State Key Laboratory of Reproductive Medicine (L.Y., A.Z.Z.), The Center of Metabolic Disease Research, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China; and State Key Laboratory of Experimental Hematology (W.Y.), Institute of Hematology; and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Allan Z Zhao
- State Key Laboratory of Organ Failure Research (H.D., Z.C., C.W., Z.X., W.Z., C.J., J.L., X.B.), Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; State Key Laboratory of Reproductive Medicine (L.Y., A.Z.Z.), The Center of Metabolic Disease Research, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China; and State Key Laboratory of Experimental Hematology (W.Y.), Institute of Hematology; and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Xiaochun Bai
- State Key Laboratory of Organ Failure Research (H.D., Z.C., C.W., Z.X., W.Z., C.J., J.L., X.B.), Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China; State Key Laboratory of Reproductive Medicine (L.Y., A.Z.Z.), The Center of Metabolic Disease Research, Nanjing Medical University, Nanjing, Jiangsu Province 210029, China; and State Key Laboratory of Experimental Hematology (W.Y.), Institute of Hematology; and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| |
Collapse
|
26
|
ER sheet–tubule balance is regulated by an array of actin filaments and microtubules. Exp Cell Res 2015; 337:170-8. [DOI: 10.1016/j.yexcr.2015.04.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 04/13/2015] [Indexed: 12/28/2022]
|
27
|
Kee AJ, Yang L, Lucas CA, Greenberg MJ, Martel N, Leong GM, Hughes WE, Cooney GJ, James DE, Ostap EM, Han W, Gunning PW, Hardeman EC. An actin filament population defined by the tropomyosin Tpm3.1 regulates glucose uptake. Traffic 2015; 16:691-711. [PMID: 25783006 PMCID: PMC4945106 DOI: 10.1111/tra.12282] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Revised: 03/10/2015] [Accepted: 03/11/2015] [Indexed: 12/21/2022]
Abstract
Actin has an ill-defined role in the trafficking of GLUT4 glucose transporter vesicles to the plasma membrane (PM). We have identified novel actin filaments defined by the tropomyosin Tpm3.1 at glucose uptake sites in white adipose tissue (WAT) and skeletal muscle. In Tpm 3.1-overexpressing mice, insulin-stimulated glucose uptake was increased; while Tpm3.1-null mice they were more sensitive to the impact of high-fat diet on glucose uptake. Inhibition of Tpm3.1 function in 3T3-L1 adipocytes abrogates insulin-stimulated GLUT4 translocation and glucose uptake. In WAT, the amount of filamentous actin is determined by Tpm3.1 levels and is paralleled by changes in exocyst component (sec8) and Myo1c levels. In adipocytes, Tpm3.1 localizes with MyoIIA, but not Myo1c, and it inhibits Myo1c binding to actin. We propose that Tpm3.1 determines the amount of cortical actin that can engage MyoIIA and generate contractile force, and in parallel limits the interaction of Myo1c with actin filaments. The balance between these actin filament populations may determine the efficiency of movement and/or fusion of GLUT4 vesicles with the PM.
Collapse
Affiliation(s)
- Anthony J. Kee
- Cellular and Genetic Medicine UnitSchool of Medical Sciences, UNSW AustraliaSydneyNSW2052Australia
| | - Lingyan Yang
- Cellular and Genetic Medicine UnitSchool of Medical Sciences, UNSW AustraliaSydneyNSW2052Australia
| | - Christine A. Lucas
- Cellular and Genetic Medicine UnitSchool of Medical Sciences, UNSW AustraliaSydneyNSW2052Australia
| | - Michael J. Greenberg
- The Pennsylvania Muscle Institute and Department of PhysiologyPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPA19104‐6085USA
| | - Nick Martel
- Obesity Research Centre, Institute for Molecular BioscienceThe University of QueenslandSt LuciaQLD4072Australia
| | - Gary M. Leong
- Obesity Research Centre, Institute for Molecular BioscienceThe University of QueenslandSt LuciaQLD4072Australia
- Department of Paediatric Endocrinology and DiabetesMater Children's HospitalSouth BrisbaneQLD4010Australia
| | - William E. Hughes
- Diabetes and Obesity ProgramGarvan Institute of Medical ResearchSydneyNSW2010Australia
| | - Gregory J. Cooney
- Diabetes and Obesity ProgramGarvan Institute of Medical ResearchSydneyNSW2010Australia
| | - David E. James
- Charles Perkins Centre, School of Molecular BioscienceUniversity of SydneySydneyNSW2006Australia
| | - E. Michael Ostap
- The Pennsylvania Muscle Institute and Department of PhysiologyPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPA19104‐6085USA
| | - Weiping Han
- Singapore Bioimaging ConsortiumAgency for Science, Technology and Research (A*STAR)Singapore138667Singapore
| | - Peter W. Gunning
- Oncology Research UnitSchool of Medical Sciences, UNSW AustraliaSydneyNSW2052Australia
| | - Edna C. Hardeman
- Cellular and Genetic Medicine UnitSchool of Medical Sciences, UNSW AustraliaSydneyNSW2052Australia
| |
Collapse
|
28
|
Koo J, Wu X, Mao Z, Khuri FR, Sun SY. Rictor Undergoes Glycogen Synthase Kinase 3 (GSK3)-dependent, FBXW7-mediated Ubiquitination and Proteasomal Degradation. J Biol Chem 2015; 290:14120-14129. [PMID: 25897075 PMCID: PMC4447982 DOI: 10.1074/jbc.m114.633057] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 04/15/2015] [Indexed: 12/15/2022] Open
Abstract
Rictor, an essential component of mTOR complex 2 (mTORC2), plays a pivotal role in regulating mTOR signaling and other biological functions. Posttranslational regulation of rictor (e.g. via degradation) and its underlying mechanism are largely undefined and thus are the focus of this study. Chemical inhibition of the proteasome increased rictor ubiquitination and levels. Consistently, inhibition of FBXW7 with various genetic means including knockdown, knock-out, and enforced expression of a dominant-negative mutant inhibited rictor ubiquitination and increased rictor levels, whereas enforced expression of FBXW7 decreased rictor stability and levels. Moreover, we detected an interaction between FBXW7 and rictor. Hence, rictor is degraded through an FBXW7-mediated ubiquitination/proteasome mechanism. We show that this process is dependent on glycogen synthase kinase 3 (GSK3): GSK3 was associated with rictor and directly phosphorylated the Thr-1695 site in a putative CDC4 phospho-degron motif of rictor; mutation of this site impaired the interaction between rictor and FBXW7, decreased rictor ubiquitination, and increased rictor stability. Finally, enforced activation of Akt enhanced rictor levels and increased mTORC2 activity as evidenced by increased formation of mTORC2 and elevated phosphorylation of Akt, SGK1, and PKCα. Hence we suggest that PI3K/Akt signaling may positively regulate mTORC2 signaling, likely through suppressing GSK3-dependent rictor degradation.
Collapse
Affiliation(s)
- Junghui Koo
- From the Departments of Hematology and Medical Oncology and
| | - Xiaoyun Wu
- From the Departments of Hematology and Medical Oncology and
| | - Zixu Mao
- Pharmacology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, Georgia 30322
| | - Fadlo R Khuri
- From the Departments of Hematology and Medical Oncology and
| | - Shi-Yong Sun
- From the Departments of Hematology and Medical Oncology and
| |
Collapse
|
29
|
Carr TD, Feehan RP, Hall MN, Rüegg MA, Shantz LM. Conditional disruption of rictor demonstrates a direct requirement for mTORC2 in skin tumor development and continued growth of established tumors. Carcinogenesis 2015; 36:487-97. [PMID: 25740823 DOI: 10.1093/carcin/bgv012] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 02/15/2015] [Indexed: 12/31/2022] Open
Abstract
Activation of signaling dependent on the mammalian target of rapamycin (mTOR) has been demonstrated in a variety of human malignancies, and our previous work suggests that mTOR complex (mTORC) 1 and mTORC2 may play unique roles in skin tumorigenesis. The purpose of these studies was to investigate the function of mTORC2-dependent pathways in skin tumor development and the maintenance of established tumors. Using mice that allow spatial and temporal control of mTORC2 in epidermis by conditional knockout of its essential component Rictor, we studied the effect of mTORC2 loss on both epidermal proliferation and chemical carcinogenesis. The results demonstrate that mTORC2 is dispensable for both normal epidermal proliferation and the hyperproliferative response to treatment with tetradecanoyl phorbol acetate (TPA). In contrast, deletion of epidermal Rictor prior to initiation in DMBA/TPA chemical carcinogenesis was sufficient to dramatically delay tumor development and resulted in reduced tumor number and size compared with control groups. Silencing of Rictor expression in tumor-bearing animals triggered regression of established tumors and increased caspase-3 cleavage without changes in proliferation. In vitro experiments demonstrate an increased sensitivity to caspase-dependent apoptosis in the absence of rictor, which is dependent on mTORC2 signaling. These studies demonstrate that mTORC2 activation is essential for keratinocyte survival, and suggest that inhibition of mTORC2 has value in chemoprevention by eliminating carcinogen-damaged cells during the early stages of tumorigenesis, and in therapy of existing tumors by restricting critical pro-survival pathways.
Collapse
Affiliation(s)
- Theresa D Carr
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA and
| | - Robert P Feehan
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA and
| | - Michael N Hall
- Biozentrum, University of Basel, CH-4056 Basel, Switzerland
| | - Markus A Rüegg
- Biozentrum, University of Basel, CH-4056 Basel, Switzerland
| | - Lisa M Shantz
- Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA and
| |
Collapse
|
30
|
Smrz D, Cruse G, Beaven MA, Kirshenbaum A, Metcalfe DD, Gilfillan AM. Rictor negatively regulates high-affinity receptors for IgE-induced mast cell degranulation. THE JOURNAL OF IMMUNOLOGY 2014; 193:5924-32. [PMID: 25378594 DOI: 10.4049/jimmunol.1303495] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Rictor is a regulatory component of the mammalian target of rapamycin (mTOR) complex 2 (mTORC2). We have previously demonstrated that rictor expression is substantially downregulated in terminally differentiated mast cells as compared with their immature or transformed counterparts. However, it is not known whether rictor and mTORC2 regulate mast cell activation. In this article, we show that mast cell degranulation induced by aggregation of high-affinity receptors for IgE (FcεRI) is negatively regulated by rictor independently of mTOR. We found that inhibition of mTORC2 by the dual mTORC1/mTORC2 inhibitor Torin1 or by downregulation of mTOR by short hairpin RNA had no impact on FcεRI-induced degranulation, whereas downregulation of rictor itself resulted in an increased sensitivity (∼50-fold) of cells to FcεRI aggregation with enhancement of degranulation. This was linked to a similar enhancement in calcium mobilization and cytoskeletal rearrangement attributable to increased phosphorylation of LAT and PLCγ1. In contrast, degranulation and calcium responses elicited by the G protein-coupled receptor ligand, C3a, or by thapsigargin, which induces a receptor-independent calcium signal, was unaffected by rictor knockdown. Overexpression of rictor, in contrast with knockdown, suppressed FcεRI-mediated degranulation. Taken together, these data provide evidence that rictor is a multifunctional signaling regulator that can regulate FcεRI-mediated degranulation independently of mTORC2.
Collapse
Affiliation(s)
- Daniel Smrz
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892; and
| | - Glenn Cruse
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892; and
| | - Michael A Beaven
- Laboratory of Molecular Immunology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892
| | - Arnold Kirshenbaum
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892; and
| | - Dean D Metcalfe
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892; and
| | - Alasdair M Gilfillan
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892; and
| |
Collapse
|
31
|
Klip A, Sun Y, Chiu TT, Foley KP. Signal transduction meets vesicle traffic: the software and hardware of GLUT4 translocation. Am J Physiol Cell Physiol 2014; 306:C879-86. [DOI: 10.1152/ajpcell.00069.2014] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Skeletal muscle is the major tissue disposing of dietary glucose, a function regulated by insulin-elicited signals that impart mobilization of GLUT4 glucose transporters to the plasma membrane. This phenomenon, also central to adipocyte biology, has been the subject of intense and productive research for decades. We focus on muscle cell studies scrutinizing insulin signals and vesicle traffic in a spatiotemporal manner. Using the analogy of an integrated circuit to approach the intersection between signal transduction and vesicle mobilization, we identify signaling relays (“software”) that engage structural/mechanical elements (“hardware”) to enact the rapid mobilization and incorporation of GLUT4 into the cell surface. We emphasize how insulin signal transduction switches from tyrosine through lipid and serine phosphorylation down to activation of small G proteins of the Rab and Rho families, describe key negative regulation step of Rab GTPases through the GTPase-activating protein activity of the Akt substrate of 160 kDa (AS160), and focus on the mechanical effectors engaged by Rabs 8A and 10 (the molecular motor myosin Va), and the Rho GTPase Rac1 (actin filament branching and severing through Arp2/3 and cofilin). Finally, we illustrate how actin filaments interact with myosin 1c and α-Actinin4 to promote vesicle tethering as preamble to fusion with the membrane.
Collapse
Affiliation(s)
- Amira Klip
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; and
- Department of Biochemistry, The University of Toronto, Ontario, Canada
| | - Yi Sun
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; and
| | - Tim Ting Chiu
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; and
- Department of Biochemistry, The University of Toronto, Ontario, Canada
| | - Kevin P. Foley
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; and
- Department of Biochemistry, The University of Toronto, Ontario, Canada
| |
Collapse
|
32
|
Wen SY, Li CH, Zhang YL, Bian YH, Ma L, Ge QL, Teng YC, Zhang ZG. Rictor is an independent prognostic factor for endometrial carcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:2068-2078. [PMID: 24966915 PMCID: PMC4069916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 04/10/2014] [Indexed: 06/03/2023]
Abstract
Early-stage endometrial carcinoma (EC) patients have a high cure rate; however, those with high-risk factors may have poor prognosis. Thus, there is an urgent need for searching for new prognostic molecules to more accurately predict survival of patients. We detected the Rictor mRNA expression level in 30 fresh EC tissue and 17 normal endometrial tissue samples with real-time quantitative RT-PCR and Rictor protein expression level in 134 (test cohort) and 115 (validation cohort) paraffin tissue samples by immunohistochemistry, analyzed the correlation between variables and overall survival (OS) using Cox proportional hazards regression, compared the prognostic accuracy of Rictor with other clinicopathological risk factors by logistic regression. The results showed that Rictor mRNA expression of EC is higher than that of normal endometrium; Rictor protein expression level was closely correlated with FIGO stage, grade and vascular invasion in both cohorts; a univariate analysis showed that the pathological type, stage, grade, vascular invasion, lymphatic metastasis and Rictor were predictors of OS in both cohorts; furthermore, multivariate Cox proportional hazards regression analysis indicated that vascular invasion and Rictor were independent prognostic factors for EC in both cohorts; an ROX curve comparison showed that the area under the curve (AUC) for Rictor combined with other clinicopathological prognostic factors was higher than any individual factor or other clinicopathological prognostic factors' combination. Based on the above data, we concluded that Rictor is an independent prognostic factor for EC. It combined with other clinicopathological risk factors was a stronger prognostic model than individual risk factor or their combination.
Collapse
Affiliation(s)
- Shan-Yun Wen
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People’s HospitalShanghai, China
- Department of Obstetrics and Gynecology, Shanghai Songjiang District Central HospitalShanghai, China
| | - Chang-Hua Li
- Department of Obstetrics & Gynecology, Huai’an First People’s Hospital, Nanjing Medical UniversityHuai’an, Jiangsu, PR China
| | - Yan-Li Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
| | - Yu-Hai Bian
- Department of General Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong UniversityShanghai, China
| | - Li Ma
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People’s HospitalShanghai, China
| | - Qiu-Lin Ge
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People’s HospitalShanghai, China
| | - Yin-Cheng Teng
- Department of Obstetrics and Gynecology, Shanghai Jiao Tong University Affiliated Sixth People’s HospitalShanghai, China
| | - Zhi-Gang Zhang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghai, China
| |
Collapse
|
33
|
Xu Y, Lai E, Liu J, Lin J, Yang C, Jia C, Li Y, Bai X, Li M. IKK interacts with rictor and regulates mTORC2. Cell Signal 2013; 25:2239-45. [DOI: 10.1016/j.cellsig.2013.07.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Accepted: 07/11/2013] [Indexed: 12/17/2022]
|
34
|
Alan JK, Struckhoff EC, Lundquist EA. Multiple cytoskeletal pathways and PI3K signaling mediate CDC-42-induced neuronal protrusion in C. elegans. Small GTPases 2013; 4:208-20. [PMID: 24149939 PMCID: PMC4011816 DOI: 10.4161/sgtp.26602] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 09/23/2013] [Accepted: 09/25/2013] [Indexed: 12/30/2022] Open
Abstract
Rho GTPases are key regulators of cellular protrusion and are involved in many developmental events including axon guidance during nervous system development. Rho GTPase pathways display functional redundancy in developmental events, including axon guidance. Therefore, their roles can often be masked when using simple loss-of-function genetic approaches. As a complement to loss-of-function genetics, we constructed a constitutively activated CDC-42(G12V) expressed in C. elegans neurons. CDC-42(G12V) drove the formation of ectopic lamellipodial and filopodial protrusions in the PDE neurons, which resembled protrusions normally found on migrating growth cones of axons. We then used a candidate gene approach to identify molecules that mediate CDC-42(G12V)-induced ectopic protrusions by determining if loss of function of the genes could suppress CDC-42(G12V). Using this approach, we identified 3 cytoskeletal pathways previously implicated in axon guidance, the Arp2/3 complex, UNC-115/abLIM, and UNC-43/Ena. We also identified the Nck-interacting kinase MIG-15/NIK and p21-activated kinases (PAKs), also implicated in axon guidance. Finally, PI3K signaling was required, specifically the Rictor/mTORC2 branch but not the mTORC1 branch that has been implicated in other aspects of PI3K signaling including stress and aging. Our results indicate that multiple pathways can mediate CDC-42-induced neuronal protrusions that might be relevant to growth cone protrusions during axon pathfinding. Each of these pathways involves Rac GTPases, which might serve to integrate the pathways and coordinate the multiple CDC-42 pathways. These pathways might be relevant to developmental events such as axon pathfinding as well as disease states such as metastatic melanoma.
Collapse
Affiliation(s)
| | - Eric C Struckhoff
- Department of Molecular Biosciences; University of Kansas; Lawrence, KS USA
| | - Erik A Lundquist
- Department of Molecular Biosciences; University of Kansas; Lawrence, KS USA
| |
Collapse
|
35
|
Agarwal NK, Kazyken D, Sarbassov DD. Rictor encounters RhoGDI2: the second pilot is taking a lead. Small GTPases 2013; 4:102-5. [PMID: 23354413 DOI: 10.4161/sgtp.23346] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Rictor's role in cell migration has been first indicated in the original chemotaxis studies in Dictyostelium and more recent studies reported that rictor is required for migration of cancer cells. How rictor promotes cell migration remains poorly characterized. Based on our proteomics study we have identified a novel functional role of rictor in regulation of cell migration. Here, we discuss our recent finding that rictor by suppressing RhoGDI2 maintains activity of the Rac1/cdc42 GTPases and promotes cell migration. Our finding outlines a critical role of rictor in the regulation of RhoGDI2 activity. This study opens new avenues in the investigation of cancer metastasis by analyzing the rictor dependent post-translational modification of RhoGDI2.
Collapse
Affiliation(s)
- Nitin K Agarwal
- Department of Hematopathology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | | | | |
Collapse
|
36
|
Mok KW, Mruk DD, Lee WM, Cheng CY. Rictor/mTORC2 regulates blood-testis barrier dynamics via its effects on gap junction communications and actin filament network. FASEB J 2013; 27:1137-52. [PMID: 23288930 DOI: 10.1096/fj.12-212977] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In the mammalian testis, coexisting tight junctions (TJs), basal ectoplasmic specializations, and gap junctions (GJs), together with desmosomes near the basement membrane, constitute the blood-testis barrier (BTB). The most notable feature of the BTB, however, is the extensive network of actin filament bundles, which makes it one of the tightest blood-tissue barriers. The BTB undergoes restructuring to facilitate the transit of preleptotene spermatocytes at stage VIII-IX of the epithelial cycle. Thus, the F-actin network at the BTB undergoes cyclic reorganization via a yet-to-be explored mechanism. Rictor, the key component of mTORC2 that is known to regulate actin cytoskeleton, was shown to express stage-specifically at the BTB in the seminiferous epithelium. Its expression was down-regulated at the BTB in stage VIII-IX tubules, coinciding with BTB restructuring at these stages. Using an in vivo model, a down-regulation of rictor at the BTB was also detected during adjudin-induced BTB disruption, illustrating rictor expression is positively correlated with the status of the BTB integrity. Indeed, the knockdown of rictor by RNAi was found to perturb the Sertoli cell TJ-barrier function in vitro and the BTB integrity in vivo. This loss of barrier function was accompanied by changes in F-actin organization at the Sertoli cell BTB in vitro and in vivo, associated with a loss of interaction between actin and α-catenin or ZO-1. Rictor knockdown by RNAi was also found to impede Sertoli cell-cell GJ communication, disrupting protein distribution (e.g., occludin, ZO-1) at the BTB, illustrating that rictor is a crucial BTB regulator.
Collapse
Affiliation(s)
- Ka-Wai Mok
- Mary M. Wohlford Laboratory for Male Contraceptive Research, Center for Biomedical Research, Population Council, 1230 York Ave, New York, NY 10065, USA
| | | | | | | |
Collapse
|
37
|
Bond LM, Brandstaetter H, Kendrick-Jones J, Buss F. Functional roles for myosin 1c in cellular signaling pathways. Cell Signal 2013; 25:229-35. [PMID: 23022959 PMCID: PMC3715701 DOI: 10.1016/j.cellsig.2012.09.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Accepted: 09/24/2012] [Indexed: 02/01/2023]
Abstract
Cellular signaling pathways underlie the transfer of information throughout the cell and to adjoining cells and so govern most critical cellular functions. Increasing evidence points to the molecular motor myosin 1c as a prominent player in many signaling cascades, from the integrin-dependent signaling involved in cell migration to the signaling events underlying insulin resistance. Myosin 1c functions on these pathways both via an important role in regulating lipid raft recycling and also via direct involvement in signaling cascades. This review provides an overview of the functional involvement of myosin 1c in cellular signaling and discusses the possible potential for myosin 1c as a target for drug-based treatments for human diseases.
Collapse
Affiliation(s)
- Lisa M Bond
- Cambridge Institute for Medical Research, University of Cambridge, Wellcome Trust/MRC Building, Hills Road, Cambridge, CB2 0XY, United Kingdom
| | | | | | | |
Collapse
|
38
|
Greenberg MJ, Ostap EM. Regulation and control of myosin-I by the motor and light chain-binding domains. Trends Cell Biol 2012. [PMID: 23200340 DOI: 10.1016/j.tcb.2012.10.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Members of the myosin-I family of molecular motors are expressed in many eukaryotes, where they are involved in a multitude of critical processes. Humans express eight distinct members of the myosin-I family, making it the second largest family of myosins expressed in humans. Despite the high degree of sequence conservation in the motor and light chain-binding domains (LCBDs) of these myosins, recent studies have revealed surprising diversity of function and regulation arising from isoform-specific differences in these domains. Here we review the regulation of myosin-I function and localization by the motor and LCBDs.
Collapse
Affiliation(s)
- Michael J Greenberg
- The Pennsylvania Muscle Institute and Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6085, USA
| | | |
Collapse
|
39
|
Bashir T, Cloninger C, Artinian N, Anderson L, Bernath A, Holmes B, Benavides-Serrato A, Sabha N, Nishimura RN, Guha A, Gera J. Conditional astroglial Rictor overexpression induces malignant glioma in mice. PLoS One 2012; 7:e47741. [PMID: 23077666 PMCID: PMC3471885 DOI: 10.1371/journal.pone.0047741] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Accepted: 09/14/2012] [Indexed: 11/18/2022] Open
Abstract
Background Hyperactivation of the mTORC2 signaling pathway has been shown to contribute to the oncogenic properties of gliomas. Moreover, overexpression of the mTORC2 regulatory subunit Rictor has been associated with increased proliferation and invasive character of these tumor cells. Methodology/Principal Findings To determine whether Rictor overexpression was sufficient to induce glioma formation in mice, we inserted a Cre-lox-regulated human Rictor transgene into the murine ROSA26 locus. This floxed Rictor strain was crossed with mice expressing the Cre recombinase driven from the glial fibrillary acidic protein (GFAP) promoter whose expression is limited to the glial cell compartment. Double transgenic GFAP-Cre/RictorloxP/loxP mice developed multifocal infiltrating glioma containing elevated mTORC2 activity and typically involved the subventricular zone (SVZ) and lateral ventricle. Analysis of Rictor-dependent signaling in these tumors demonstrated that in addition to elevated mTORC2 activity, an mTORC2-independent marker of cortical actin network function, was also elevated. Upon histological examination of the neoplasms, many displayed oligodendroglioma-like phenotypes and expressed markers associated with oligodendroglial lineage tumors. To determine whether upstream oncogenic EGFRvIII signaling would alter tumor phenotypes observed in the GFAP-Cre/RictorloxP/loxP mice, transgenic GFAP-EGFRvIII; GFAP-Cre/RictorloxP/loxP mice were generated. These mice developed mixed astrocytic-oligodendroglial tumors, however glioma formation was accelerated and correlated with increased mTORC2 activity. Additionally, the subventricular zone within the GFAP-Cre/RictorloxP/loxP mouse brain was markedly expanded, and a further proliferation within this compartment of the brain was observed in transgenic GFAP-EGFRvIII; GFAP-Cre/RictorloxP/loxP mice. Conclusion/Significance These data collectively establish Rictor as a novel oncoprotein and support the role of dysregulated Rictor expression in gliomagenesis via mTOR-dependent and mTOR-independent mechanisms. Furthermore, oncogenic EGFRvIII signaling appears to potentiate the in vivo proliferative capacity of GFAP-Cre/RictorloxP/loxP gliomas.
Collapse
Affiliation(s)
- Tariq Bashir
- Department of Research and Development, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, California, United States of America
| | - Cheri Cloninger
- Department of Research and Development, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, California, United States of America
| | - Nicholas Artinian
- Department of Research and Development, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, California, United States of America
| | - Lauren Anderson
- Department of Research and Development, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, California, United States of America
| | - Andrew Bernath
- Department of Research and Development, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, California, United States of America
| | - Brent Holmes
- Department of Research and Development, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, California, United States of America
| | - Angelica Benavides-Serrato
- Department of Research and Development, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, California, United States of America
| | - Nesrin Sabha
- Labatt Brain Tumor Research Centre, Hospital for Sick Children Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Robert N. Nishimura
- Department of Research and Development, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, California, United States of America
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Abhijit Guha
- Labatt Brain Tumor Research Centre, Hospital for Sick Children Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Joseph Gera
- Department of Research and Development, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, California, United States of America
- Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California, United States of America
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
40
|
Fan Y, Eswarappa SM, Hitomi M, Fox PL. Myo1c facilitates G-actin transport to the leading edge of migrating endothelial cells. ACTA ACUST UNITED AC 2012; 198:47-55. [PMID: 22778278 PMCID: PMC3392929 DOI: 10.1083/jcb.201111088] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Addition of actin monomer (G-actin) to growing actin filaments (F-actin) at the leading edge generates force for cell locomotion. The polymerization reaction and its regulation have been studied in depth. However, the mechanism responsible for transport of G-actin substrate to the cell front is largely unknown; random diffusion, facilitated transport via myosin II contraction, local synthesis as a result of messenger ribonucleic acid localization, or F-actin turnover all might contribute. By tracking a photoactivatable, nonpolymerizable actin mutant, we show vectorial transport of G-actin in live migrating endothelial cells (ECs). Mass spectrometric analysis identified Myo1c, an unconventional F-actin-binding motor protein, as a major G-actin-interacting protein. The cargo-binding tail domain of Myo1c interacted with G-actin, and the motor domain was required for the transport. Local microinjection of Myo1c promoted G-actin accumulation and plasma membrane ruffling, and Myo1c knockdown confirmed its contribution to G-actin delivery to the leading edge and for cell motility. In addition, there is no obvious requirement for myosin II contractile-based transport of G-actin in ECs. Thus, Myo1c-facilitated G-actin transport might be a critical node for control of cell polarity and motility.
Collapse
Affiliation(s)
- Yi Fan
- Department of Cell Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | | | | | | |
Collapse
|
41
|
Boguslavsky S, Chiu T, Foley KP, Osorio-Fuentealba C, Antonescu CN, Bayer KU, Bilan PJ, Klip A. Myo1c binding to submembrane actin mediates insulin-induced tethering of GLUT4 vesicles. Mol Biol Cell 2012; 23:4065-78. [PMID: 22918957 PMCID: PMC3469521 DOI: 10.1091/mbc.e12-04-0263] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
GLUT4-containing vesicles cycle between the plasma membrane and intracellular compartments. Insulin promotes GLUT4 exocytosis by regulating GLUT4 vesicle arrival at the cell periphery and its subsequent tethering, docking, and fusion with the plasma membrane. The molecular machinery involved in GLUT4 vesicle tethering is unknown. We show here that Myo1c, an actin-based motor protein that associates with membranes and actin filaments, is required for insulin-induced vesicle tethering in muscle cells. Myo1c was found to associate with both mobile and tethered GLUT4 vesicles and to be required for vesicle capture in the total internal reflection fluorescence (TIRF) zone beneath the plasma membrane. Myo1c knockdown or overexpression of an actin binding-deficient Myo1c mutant abolished insulin-induced vesicle immobilization, increased GLUT4 vesicle velocity in the TIRF zone, and prevented their externalization. Conversely, Myo1c overexpression immobilized GLUT4 vesicles in the TIRF zone and promoted insulin-induced GLUT4 exposure to the extracellular milieu. Myo1c also contributed to insulin-dependent actin filament remodeling. Thus we propose that interaction of vesicular Myo1c with cortical actin filaments is required for insulin-mediated tethering of GLUT4 vesicles and for efficient GLUT4 surface delivery in muscle cells.
Collapse
Affiliation(s)
- Shlomit Boguslavsky
- Cell Biology Program, Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Agarwal NK, Chen CH, Cho H, Boulbès DR, Spooner E, Sarbassov DD. Rictor regulates cell migration by suppressing RhoGDI2. Oncogene 2012; 32:2521-6. [PMID: 22777355 PMCID: PMC3470753 DOI: 10.1038/onc.2012.287] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Rictor and its binding partner Sin1 are indispensable components of mTORC2 (mammalian Target of Rapamycin Complex 2). The mTORC2 signaling complex functions as the regulatory kinase of the distinct members of AGC kinase family known to regulate cell proliferation and survival. In the early chemotaxis studies in Dictyostelium, the rictor's ortholog has been identified as a regulator of cell migration. How rictor regulates cell migration is poorly characterized. Here we show that rictor regulates cell migration by controlling a potent inhibitor of Rho proteins known as the Rho-GDP dissociation inhibitor 2 (RhoGDI2). Based on our proteomics study we identified that the rictor-dependent deficiency in cell migration is caused by up-regulation of RhoGDI2 leading to a low activity of Rac and Cdc42. We found that a suppression of RhoGDI2 by rictor is not related to the Sin1 or raptor function that excludes a role of mTORC2 or mTORC1 in regulation of RhoGDI2. Our study reveals that rictor by suppressing RhoGDI2 promotes activity of the Rho proteins and cell migration.
Collapse
Affiliation(s)
- N K Agarwal
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | |
Collapse
|
43
|
Pereira MJ, Palming J, Rizell M, Aureliano M, Carvalho E, Svensson MK, Eriksson JW. mTOR inhibition with rapamycin causes impaired insulin signalling and glucose uptake in human subcutaneous and omental adipocytes. Mol Cell Endocrinol 2012; 355:96-105. [PMID: 22333157 DOI: 10.1016/j.mce.2012.01.024] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Revised: 01/17/2012] [Accepted: 01/27/2012] [Indexed: 02/07/2023]
Abstract
Rapamycin is an immunosuppressive agent used after organ transplantation, but its molecular effects on glucose metabolism needs further evaluation. We explored rapamycin effects on glucose uptake and insulin signalling proteins in adipocytes obtained via subcutaneous (n=62) and omental (n=10) fat biopsies in human donors. At therapeutic concentration (0.01 μM) rapamycin reduced basal and insulin-stimulated glucose uptake by 20-30%, after short-term (15 min) or long-term (20 h) culture of subcutaneous (n=23 and n=10) and omental adipocytes (n=6 and n=7). Rapamycin reduced PKB Ser473 and AS160 Thr642 phosphorylation, and IRS2 protein levels in subcutaneous adipocytes. Additionally, it reduced mTOR-raptor, mTOR-rictor and mTOR-Sin1 interactions, suggesting decreased mTORC1 and mTORC2 formation. Rapamycin also reduced IR Tyr1146 and IRS1 Ser307/Ser616/Ser636 phosphorylation, whereas no effects were observed on the insulin stimulated IRS1-Tyr and TSC2 Thr1462 phosphorylation. This is the first study to show that rapamycin reduces glucose uptake in human adipocytes through impaired insulin signalling and this may contribute to the development of insulin resistance associated with rapamycin therapy.
Collapse
Affiliation(s)
- Maria J Pereira
- The Lundberg Laboratory for Diabetes Research, Department of Molecular and Clinical Medicine, The Sahlgrenska Academy at University of Gothenburg, 413 45 Gothenburg, Sweden.
| | | | | | | | | | | | | |
Collapse
|
44
|
Rictor regulates FBXW7-dependent c-Myc and cyclin E degradation in colorectal cancer cells. Biochem Biophys Res Commun 2012; 418:426-32. [PMID: 22285861 DOI: 10.1016/j.bbrc.2012.01.054] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2012] [Accepted: 01/11/2012] [Indexed: 11/23/2022]
Abstract
Rictor (Rapamycin-insensitive companion of mTOR) forms a complex with mTOR and phosphorylates and activates Akt. Activation of Akt induces expression of c-Myc and cyclin E, which are overexpressed in colorectal cancer and play an important role in colorectal cancer cell proliferation. Here, we show that rictor associates with FBXW7 to form an E3 complex participating in the regulation of c-Myc and cyclin E degradation. The Rictor-FBXW7 complex is biochemically distinct from the previously reported mTORC2 and can be immunoprecipitated independently of mTORC2. Moreover, knocking down of rictor in serum-deprived colorectal cancer cells results in the decreased ubiquitination and increased protein levels of c-Myc and cyclin E while overexpression of rictor induces the degradation of c-Myc and cyclin E proteins. Genetic knockout of FBXW7 blunts the effects of rictor, suggesting that rictor regulation of c-Myc and cyclin E requires FBXW7. Our findings identify rictor as an important component of FBXW7 E3 ligase complex participating in the regulation of c-Myc and cyclin E protein ubiquitination and degradation. Importantly, our results suggest that elevated growth factor signaling may contribute to decrease rictor/FBXW7-mediated ubiquitination of c-Myc and cyclin E, thus leading to accumulation of cyclin E and c-Myc in colorectal cancer cells.
Collapse
|
45
|
Boulbés DR, Shaiken T, Sarbassov DD. Endoplasmic reticulum is a main localization site of mTORC2. Biochem Biophys Res Commun 2011; 413:46-52. [PMID: 21867682 DOI: 10.1016/j.bbrc.2011.08.034] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Accepted: 08/07/2011] [Indexed: 11/25/2022]
Abstract
The Akt kinase is a critical effector in growth factor signaling. Activation of Akt driven by the growth factor dependent PI3K (phosphatidylinositol-3-OH kinase) is coupled to the plasma membrane translocation and phosphorylation of Akt on two sites by PDK1 (phosphoinositide-dependent protein kinase-1) on Thr-308 and by mTORC2 (mammalian Target of Rapamycin Complex 2) on Ser-473. In our study we examined the sub-cellular localization of mTORC2 and identified that this kinase complex predominantly resides on endoplasmic reticulum (ER). Our immunostaining analysis did not show a substantial co-localization of the mTORC2 component rictor with Golgi, lysosome, clathrin-coated vesicles, early endosomes, or plasma membrane but indicated a strong co-localization of rictor with ribosomal protein S6 and ER marker. Our biochemical study also identified the mTORC2 components rictor, SIN1, and mTOR as the highly abundant proteins in the ER fraction, whereas only small amount of these proteins are detected in the plasma membrane and cytosolic fractions. We found that growth factor signaling does not alter the ER localization of mTORC2 and also does not induce its translocation to the plasma membrane. Based on our study we suggest that the mTORC2-dependent phosphorylation of Akt on Ser-473 takes place on the surface of ER.
Collapse
Affiliation(s)
- Delphine R Boulbés
- Department of Molecular and Cellular Oncology, University of Texas M. D. Anderson Cancer Center, Houston, TX 77030, USA.
| | | | | |
Collapse
|
46
|
Abstract
The mechanistic target of rapamycin (mTOR) plays a central role in cellular growth and metabolism. mTOR forms two distinct protein complexes, mTORC1 and mTORC2. Much is known about the regulation and functions of mTORC1 due to availability of a natural compound, rapamycin, that inhibits this complex. Studies that define mTORC2 cellular functions and signaling have lagged behind. The development of pharmacological inhibitors that block mTOR kinase activity, and thereby inhibit both mTOR complexes, along with availability of mice with genetic knockouts in mTOR complex components have now provided new insights on mTORC2 function and regulation. Since prolonged effects of rapamycin can also disrupt mTORC2, it is worth re-evaluating the contribution of this less-studied mTOR complex in cancer, metabolic disorders and aging. In this review, we focus on recent developments on mammalian mTORC2 signaling mechanisms and its cellular and tissue-specific functions.
Collapse
Affiliation(s)
- Won Jun Oh
- Department of Physiology and Biophysics, UMDNJ-Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | | |
Collapse
|
47
|
Huang YC, Chang WL, Huang SF, Lin CY, Lin HC, Chang TC. Pachymic acid stimulates glucose uptake through enhanced GLUT4 expression and translocation. Eur J Pharmacol 2010; 648:39-49. [PMID: 20816811 DOI: 10.1016/j.ejphar.2010.08.021] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2009] [Revised: 07/14/2010] [Accepted: 08/24/2010] [Indexed: 12/28/2022]
Abstract
In an effort to investigate the effect and mechanism of Poria cocos on glucose uptake, six lanostane-type triterpenoids were isolated and analyzed. Among them, pachymic acid displayed the most significant stimulating activity on glucose uptake in 3T3-L1 adipocytes. The effect of pachymic acid on the expression profile of glucose transporters in differentiated 3T3-L1 adipocytes was also analyzed. Our results demonstrated that pachymic acid induced an increase in GLUT4, but not GLUT1, expression at both the mRNA and protein levels. The role of GLUT4 was further confirmed using the lentiviral vector-derived GLUT4 short hairpin RNA (shRNA). The stimulating activity of pachymic acid on glucose uptake was abolished when the endogenous GLUT4 expression was suppressed in 3T3-L1 adipocytes. In addition to increased GLUT4 expression, pachymic acid stimulated GLUT4 redistribution from intracellular vesicles to the plasma membrane in adipocytes. Exposure of the differentiated adipocytes to pachymic acid increased the phosphorylation of insulin receptor substrate (IRS)-1, AKT and AMP-activated kinase (AMPK). The involvement of PI3K and AMPK in the action of pachymic acid was further confirmed as PI3K and AMPK inhibitors completely blocked the pachymic acid-mediated activities in adipocytes. In addition, pachymic acid was shown to induce triglyceride accumulation and inhibit lipolysis in differentiated adipocytes. Taken together, we demonstrated the insulin-like activities of this compound in stimulating glucose uptake, GLUT4 gene expression and translocation, and promoting triglyceride accumulation in adipocytes. Our study provides important insights into the underlying mechanism of hypoglycemic activity of P. cocos.
Collapse
Affiliation(s)
- Yu-Chuan Huang
- Institute of Preventive Medicine, National Defense Medical Center, Taipei, Taiwan, ROC.
| | | | | | | | | | | |
Collapse
|
48
|
Komaba S, Coluccio LM. Localization of myosin 1b to actin protrusions requires phosphoinositide binding. J Biol Chem 2010; 285:27686-93. [PMID: 20610386 DOI: 10.1074/jbc.m109.087270] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Myosin 1b (Myo1b), a class I myosin, is a widely expressed, single-headed, actin-associated molecular motor. Transient kinetic and single-molecule studies indicate that it is kinetically slow and responds to tension. Localization and subcellular fractionation studies indicate that Myo1b associates with the plasma membrane and certain subcellular organelles such as endosomes and lysosomes. Whether Myo1b directly associates with membranes is unknown. We demonstrate here that full-length rat Myo1b binds specifically and with high affinity to phosphatidylinositol 4,5-bisphosphate (PIP(2)) and phosphatidylinositol 3,4,5-triphosphate (PIP(3)), two phosphoinositides that play important roles in cell signaling. Binding is not Ca(2+)-dependent and does not involve the calmodulin-binding IQ region in the neck domain of Myo1b. Furthermore, the binding site is contained entirely within the C-terminal tail region, which contains a putative pleckstrin homology domain. Single mutations in the putative pleckstrin homology domain abolish binding of the tail domain of Myo1b to PIP(2) and PIP(3) in vitro. These same mutations alter the distribution of Myc-tagged Myo1b at membrane protrusions in HeLa cells where PIP(2) localizes. In addition, we found that motor activity is required for Myo1b localization in filopodia. These results suggest that binding of Myo1b to phosphoinositides plays an important role in vivo by regulating localization to actin-enriched membrane projections.
Collapse
Affiliation(s)
- Shigeru Komaba
- Boston Biomedical Research Institute, Watertown, Massachusetts 02472, USA
| | | |
Collapse
|
49
|
Boulbes D, Chen CH, Shaikenov T, Agarwal NK, Peterson TR, Addona TA, Keshishian H, Carr SA, Magnuson MA, Sabatini DM, Sarbassov DD. Rictor phosphorylation on the Thr-1135 site does not require mammalian target of rapamycin complex 2. Mol Cancer Res 2010; 8:896-906. [PMID: 20501647 DOI: 10.1158/1541-7786.mcr-09-0409] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In animal cells, growth factors coordinate cell proliferation and survival by regulating the phosphoinositide 3-kinase/Akt signaling pathway. Deregulation of this signaling pathway is common in a variety of human cancers. The PI3K-dependent signaling kinase complex defined as mammalian target of rapamycin complex 2 (mTORC2) functions as a regulatory Ser-473 kinase of Akt. We find that activation of mTORC2 by growth factor signaling is linked to the specific phosphorylation of its component rictor on Thr-1135. The phosphorylation of this site is induced by the growth factor stimulation and expression of the oncogenic forms of ras or PI3K. Rictor phosphorylation is sensitive to the inhibition of PI3K, mTOR, or expression of integrin-linked kinase. The substitution of wild-type rictor with its specific phospho-mutants in rictor null mouse embryonic fibroblasts did not alter the growth factor-dependent phosphorylation of Akt, indicating that the rictor Thr-1135 phosphorylation is not critical in the regulation of the mTORC2 kinase activity. We found that this rictor phosphorylation takes place in the mTORC2-deficient cells, suggesting that this modification might play a role in the regulation of not only mTORC2 but also the mTORC2-independent function of rictor.
Collapse
Affiliation(s)
- Delphine Boulbes
- Department of Molecular and Cellular Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Bai X, Jiang Y. Key factors in mTOR regulation. Cell Mol Life Sci 2010; 67:239-53. [PMID: 19823764 PMCID: PMC4780839 DOI: 10.1007/s00018-009-0163-7] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2009] [Revised: 09/22/2009] [Accepted: 09/23/2009] [Indexed: 12/12/2022]
Abstract
Mammalian target of rapamycin (mTOR) is a protein serine/threonine kinase that controls a wide range of growth-related cellular processes. In the past several years, many factors have been identified that are involved in controlling mTOR activity. Those factors in turn are regulated by diverse signaling cascades responsive to changes in intracellular and environmental conditions. The molecular connections between mTOR and its regulators form a complex signaling network that governs cellular metabolism, growth and proliferation. In this review, we discuss some key factors in mTOR regulation and mechanisms by which these factors control mTOR activity.
Collapse
Affiliation(s)
- Xiaochun Bai
- Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, 510515 Guangzhou, China
| | - Yu Jiang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213 USA
| |
Collapse
|