1
|
Jin Q, Qi D, Zhang M, Qu H, Dong Y, Sun M, Quan C. CLDN6 inhibits breast cancer growth and metastasis through SREBP1-mediated RAS palmitoylation. Cell Mol Biol Lett 2024; 29:112. [PMID: 39169280 PMCID: PMC11337767 DOI: 10.1186/s11658-024-00629-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 08/08/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Breast cancer (BC) ranks as the third most fatal malignant tumor worldwide, with a strong reliance on fatty acid metabolism. CLDN6, a candidate BC suppressor gene, was previously identified as a regulator of fatty acid biosynthesis; however, the underlying mechanism remains elusive. In this research, we aim to clarify the specific mechanism through which CLDN6 modulates fatty acid anabolism and its impact on BC growth and metastasis. METHODS Cell function assays, tumor xenograft mouse models, and lung metastasis mouse models were conducted to evaluate BC growth and metastasis. Human palmitic acid assay, triglyceride assay, Nile red staining, and oil red O staining were employed to investigate fatty acid anabolism. Reverse transcription polymerase chain reaction (RT-PCR), western blot, immunohistochemistry (IHC) assay, nuclear fractionation, immunofluorescence (IF), immunoprecipitation and acyl-biotin exchange (IP-ABE), chromatin immunoprecipitation (ChIP), dual luciferase reporter assay, and co-immunoprecipitation (Co-IP) were applied to elucidate the underlying molecular mechanism. Moreover, tissue microarrays of BC were analyzed to explore the clinical implications. RESULTS We identified that CLDN6 inhibited BC growth and metastasis by impeding RAS palmitoylation both in vitro and in vivo. We proposed a unique theory suggesting that CLDN6 suppressed RAS palmitoylation through SREBP1-modulated de novo palmitic acid synthesis. Mechanistically, CLDN6 interacted with MAGI2 to prevent KLF5 from entering the nucleus, thereby restraining SREBF1 transcription. The downregulation of SREBP1 reduced de novo palmitic acid synthesis, hindering RAS palmitoylation and subsequent endosomal sorting complex required for transport (ESCRT)-mediated plasma membrane localization required for RAS oncogenic activation. Besides, targeting inhibition of RAS palmitoylation synergized with CLDN6 to repress BC progression. CONCLUSIONS Our findings provide compelling evidence that CLDN6 suppresses the palmitic acid-induced RAS palmitoylation through the MAGI2/KLF5/SREBP1 axis, thereby impeding BC malignant progression. These results propose a new insight that monitoring CLDN6 expression alongside targeting inhibition of palmitic acid-mediated palmitoylation could be a viable strategy for treating oncogenic RAS-driven BC.
Collapse
Affiliation(s)
- Qiu Jin
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, Jilin, China
| | - Da Qi
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, Jilin, China
| | - Mingzi Zhang
- The Zilkha Neurogenetic Institute, Department of Physiology and Neuroscience Keck School of Medicine of the University of Southern California, 1501 San Pablo Street, Los Angeles, 90033, CA, US
| | - Huinan Qu
- Department of Histology and Embryology, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, Jilin, China
| | - Yuan Dong
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, Jilin, China
| | - Minghao Sun
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, Jilin, China
| | - Chengshi Quan
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, 126 Xinmin Avenue, Changchun, 130021, Jilin, China.
| |
Collapse
|
2
|
Chen J, Chen S, Li B, Zhou S, Lin H. A pyroptosis-related signature predicts prognosis and indicates immune microenvironment infiltration in glioma. Cancer Med 2023; 12:5071-5087. [PMID: 36161280 PMCID: PMC9972150 DOI: 10.1002/cam4.5247] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 08/08/2022] [Accepted: 09/01/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Glioma, the most common malignant brain tumor, leads to high recurrence rates and disabilities in patients. Pyroptosis is an inflammasomes-induced programmed cell death in response to infection or chemotherapy. However, the role of pyroptosis in glioma has not yet been elucidated. METHODS RNA-seq data and clinical information of 660 gliomas and 847 samples were downloaded from the TCGA and CGGA, respectively. Then, data of 104 normal brain tissues was retrieved from the GTEx for differential expression analysis. Twelve pairs of peritumoral tissue and glioma samples were used for validation. Gene alteration status of differentially expressed pyroptosis-related regulators in gliomas was detected in cBioPortal algorithm. Consensus clustering was employed to classify gliomas based on differentially expressed pyroptosis-related regulators. Subsequently, a PS-signature was constructed using LASSO-congressional analysis for clinical application. The immune infiltration of glioma microenvironment (TME) was explored using ESTIMATE, CIBERSORT, and the other immune signatures. RESULTS cBioPortal algorithm revealed alteration of these regulators was correlated to better prognosis of gliomas. Then, our study showed that pyroptosis-related regulators can be used to sort out patients into two clusters with distinct prognostic outcome and immune status. Moreover, a PS-signature for predicting the prognosis of glioma patients was developed based on the identified subtypes. The high PS-score group showed more abundant inflammatory cell infiltration and stronger immune response, but with poorer prognosis of gliomas. CONCLUSION The findings of this study provide a therapeutic basis for future research on pyroptosis and unravel the relationship between pyroptosis and glioma prognosis. The risk signature can be utilized as a prognostic biomarker for glioma.
Collapse
Affiliation(s)
- Jia Chen
- The Fourth People's Hospital of ChengduChengduChina
- The Clinical Hospital of Chengdu Brain Science InstituteMOE Key Lab for Neuroinformation, University of Electronic Science and Technology of ChinaChengduChina
| | - Shanwei Chen
- Department of Neurosurgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical SciencesGuangzhouChina
- Shantou University Medical CollegeShantouChina
| | - Bingxian Li
- Department of Neurology, Shantou Central HospitalShantouChina
| | - Shaojiong Zhou
- Department of Neurology, Shantou Central HospitalShantouChina
| | - Han Lin
- Department of Neurosurgery, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
3
|
Carleton AE, Duncan MC, Taniguchi K. Human epiblast lumenogenesis: From a cell aggregate to a lumenal cyst. Semin Cell Dev Biol 2022; 131:117-123. [PMID: 35637065 PMCID: PMC9529837 DOI: 10.1016/j.semcdb.2022.05.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/13/2022] [Accepted: 05/13/2022] [Indexed: 12/14/2022]
Abstract
The formation of a central lumen in the human epiblast is a critical step for development. However, because the lumen forms in the epiblast coincident with implantation, the molecular and cellular events of this early lumenogenesis process cannot be studied in vivo. Recent developments using new model systems have revealed insight into the underpinnings of epiblast formation. To provide an up-to-date comprehensive review of human epiblast lumenogenesis, we highlight recent findings from human and mouse models with an emphasis on new molecular understanding of a newly described apicosome compartment, a novel 'formative' state of pluripotency that coordinates with epiblast polarization, and new evidence about the physical and polarized trafficking mechanisms contributing to lumenogenesis.
Collapse
Affiliation(s)
- Amber E. Carleton
- Departments of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin USA
| | - Mara C. Duncan
- Departments of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan USA,Co-corresponding authors
| | - Kenichiro Taniguchi
- Departments of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin USA,Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin USA,Co-corresponding authors
| |
Collapse
|
4
|
Neggers JE, Paolella BR, Asfaw A, Rothberg MV, Skipper TA, Yang A, Kalekar RL, Krill-Burger JM, Dharia NV, Kugener G, Kalfon J, Yuan C, Dumont N, Gonzalez A, Abdusamad M, Li YY, Spurr LF, Wu WW, Durbin AD, Wolpin BM, Piccioni F, Root DE, Boehm JS, Cherniack AD, Tsherniak A, Hong AL, Hahn WC, Stegmaier K, Golub TR, Vazquez F, Aguirre AJ. Synthetic Lethal Interaction between the ESCRT Paralog Enzymes VPS4A and VPS4B in Cancers Harboring Loss of Chromosome 18q or 16q. Cell Rep 2020; 33:108493. [PMID: 33326793 PMCID: PMC8374858 DOI: 10.1016/j.celrep.2020.108493] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 09/04/2020] [Accepted: 11/17/2020] [Indexed: 12/26/2022] Open
Abstract
Few therapies target the loss of tumor suppressor genes in cancer. We examine CRISPR-SpCas9 and RNA-interference loss-of-function screens to identify new therapeutic targets associated with genomic loss of tumor suppressor genes. The endosomal sorting complexes required for transport (ESCRT) ATPases VPS4A and VPS4B score as strong synthetic lethal dependencies. VPS4A is essential in cancers harboring loss of VPS4B adjacent to SMAD4 on chromosome 18q and VPS4B is required in tumors with co-deletion of VPS4A and CDH1 (E-cadherin) on chromosome 16q. We demonstrate that more than 30% of cancers selectively require VPS4A or VPS4B. VPS4A suppression in VPS4B-deficient cells selectively leads to ESCRT-III filament accumulation, cytokinesis defects, nuclear deformation, G2/M arrest, apoptosis, and potent tumor regression. CRISPR-SpCas9 screening and integrative genomic analysis reveal other ESCRT members, regulators of abscission, and interferon signaling as modifiers of VPS4A dependency. We describe a compendium of synthetic lethal vulnerabilities and nominate VPS4A and VPS4B as high-priority therapeutic targets for cancers with 18q or 16q loss. Neggers, Paolella, and colleagues identify the ATPases VPS4A and VPS4B as selective vulnerabilities and potential therapeutic targets in cancers harboring loss of chromosome 18q or 16q. In VPS4B-deficient cancers, VPS4A suppression leads to ESCRT-III dysfunction, nuclear deformation, and abscission defects. Moreover, ESCRT proteins and interferons can modulate dependency on VPS4A.
Collapse
Affiliation(s)
- Jasper E Neggers
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Brenton R Paolella
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Adhana Asfaw
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Michael V Rothberg
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Thomas A Skipper
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Annan Yang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Radha L Kalekar
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - John M Krill-Burger
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Neekesh V Dharia
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Cancer and Blood Disorders Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Guillaume Kugener
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jérémie Kalfon
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Chen Yuan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Nancy Dumont
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Alfredo Gonzalez
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Mai Abdusamad
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Yvonne Y Li
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Liam F Spurr
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Westley W Wu
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Adam D Durbin
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Cancer and Blood Disorders Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Federica Piccioni
- Genetic Perturbation Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - David E Root
- Genetic Perturbation Platform, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jesse S Boehm
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Andrew D Cherniack
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Aviad Tsherniak
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Andrew L Hong
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Cancer and Blood Disorders Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - William C Hahn
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Kimberly Stegmaier
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; Cancer and Blood Disorders Center, Boston Children's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Todd R Golub
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Francisca Vazquez
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
| | - Andrew J Aguirre
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
5
|
Li L, Lin L, Veeraraghavan J, Hu Y, Wang X, Lee S, Tan Y, Schiff R, Wang XS. Therapeutic role of recurrent ESR1-CCDC170 gene fusions in breast cancer endocrine resistance. Breast Cancer Res 2020; 22:84. [PMID: 32771039 PMCID: PMC7414578 DOI: 10.1186/s13058-020-01325-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 07/27/2020] [Indexed: 01/07/2023] Open
Abstract
Background Endocrine therapy is the most common treatment for estrogen receptor (ER)-positive breast cancer, but its effectiveness is limited by high rates of primary and acquired resistance. There are likely many genetic causes, and recent studies suggest the important role of ESR1 mutations and fusions in endocrine resistance. Previously, we reported a recurrent ESR1 fusion called ESR1-CCDC170 in 6–8% of the luminal B breast cancers that has a worse clinical outcome after endocrine therapy. Despite being the most frequent ESR1 fusion, its functional role in endocrine resistance has not been studied in vivo, and the engaged mechanism and therapeutic relevance remain uncharacterized. Methods The endocrine sensitivities of HCC1428 or T47D breast cancer cells following genetic perturbations of ESR1-CCDC170 were assessed using clonogenic assays and/or xenograft mouse models. The underlying mechanisms were investigated by reverse phase protein array, western blotting, immunoprecipitation, and bimolecular fluorescence complementation assays. The sensitivity of ESR1-CCDC170 expressing breast cancer cells to concomitant treatments of tamoxifen and HER/SRC inhibitors was assessed by clonogenic assays. Results Our results suggested that different ESR1-CCDC170 fusions endow different levels of reduced endocrine sensitivity in vivo, resulting in significant survival disadvantages. Further investigation revealed a novel mechanism that ESR1-CCDC170 binds to HER2/HER3/SRC and activates SRC/PI3K/AKT signaling. Silencing of ESR1-CCDC170 in the fusion-positive cell line, HCC1428, downregulates HER2/HER3, represses pSRC/pAKT, and improves endocrine sensitivity. More important, breast cancer cells expressing ectopic or endogenous ESR1-CCDC170 are highly sensitive to treatment regimens combining endocrine agents with the HER2 inhibitor lapatinib and/or the SRC inhibitor dasatinib. Conclusion ESR1-CCDC170 may endow breast cancer cell survival under endocrine therapy via maintaining/activating HER2/HER3/SRC/AKT signaling which implies a potential therapeutic strategy for managing these fusion positive tumors.
Collapse
Affiliation(s)
- Li Li
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15232, USA.,Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15261, USA.,Women's Cancer Research Center, Magee-Womens Research Institute, Pittsburgh, PA, 15213, USA.,Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Ling Lin
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15232, USA.,Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15261, USA.,Women's Cancer Research Center, Magee-Womens Research Institute, Pittsburgh, PA, 15213, USA
| | - Jamunarani Veeraraghavan
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.,Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yiheng Hu
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15232, USA.,Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15261, USA.,Women's Cancer Research Center, Magee-Womens Research Institute, Pittsburgh, PA, 15213, USA.,Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xian Wang
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15232, USA.,Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15261, USA.,Women's Cancer Research Center, Magee-Womens Research Institute, Pittsburgh, PA, 15213, USA.,Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Sanghoon Lee
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15232, USA.,Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, 15206, USA
| | - Ying Tan
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Rachel Schiff
- Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.,Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Xiao-Song Wang
- Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15232, USA. .,Department of Pathology, University of Pittsburgh, Pittsburgh, PA, 15261, USA. .,Women's Cancer Research Center, Magee-Womens Research Institute, Pittsburgh, PA, 15213, USA. .,Lester & Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA. .,Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA. .,Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, PA, 15206, USA.
| |
Collapse
|
6
|
Yang T, Li W, Li Y, Liu X, Yang D. The ESCRT System Plays an Important Role in the Germination in Candida albicans by Regulating the Expression of Hyphal-Specific Genes and the Localization of Polarity-Related Proteins. Mycopathologia 2020; 185:439-454. [PMID: 32279163 DOI: 10.1007/s11046-020-00442-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 03/12/2020] [Indexed: 11/30/2022]
Abstract
Candida albicans is an important opportunistic fungal pathogen, and its pathogenicity is closely related to its ability to form hyphae. ESCRT system was initially discovered as a membrane-budding machinery involved in the formation of multivesicular bodies. More recently, the role of ESCRT is vastly expanded. Early reports showed that the ESCRT system is involved in inducing hyphae under neutral-alkaline environment via the Rim101 pathway. We previously found that in the environment that contains serum, one ESCRT protein, Vps4, is essential for polarity maintenance during hyphal formation, as its deletion causes the formation of multiple hyphae. In this study, we found that Vps4 is also essential for the proper localization of Cdc42 and Cdc3, which may be related to its role in polarity maintenance. We also discovered that deletions of the ESCRT proteins significantly delay germination and cause downregulation of hyphal-specific genes, most prominent of which is HGC1. Since Hgc1 is essential for many aspects of hyphal growth, its downregulation could explain our observed phenotypes. Our further studies show that ESCRT proteins are involved in the dynamics of Ras1. Deletions of VPS4 or SNF7 significantly decrease the recovery rate of GFP-Ras1 in the fluorescence recovery after photobleaching experiment. The decreased Ras1 dynamics may disrupt the signaling pathway and lead to downregulation of hyphal-specific genes. Therefore, in this study we discovered a novel and Rim101 independent mechanism used by the ESCRT system to regulate hyphal induction and polarity maintenance, which could provide insights on the pathogenicity mechanism of Candia albicans.
Collapse
Affiliation(s)
- Tianran Yang
- Gene Engineering and Biotechnology Beijing Key Laboratory, College of Life Sciences, Beijing Normal University, Beijing, 100875, China.,Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, 100875, China
| | - Wanjie Li
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, 100875, China
| | - Yi Li
- Gene Engineering and Biotechnology Beijing Key Laboratory, College of Life Sciences, Beijing Normal University, Beijing, 100875, China.,Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, 100875, China
| | - Xin Liu
- Gene Engineering and Biotechnology Beijing Key Laboratory, College of Life Sciences, Beijing Normal University, Beijing, 100875, China.,Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, 100875, China
| | - Dong Yang
- Gene Engineering and Biotechnology Beijing Key Laboratory, College of Life Sciences, Beijing Normal University, Beijing, 100875, China. .,Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, College of Life Sciences, Beijing Normal University, Beijing, 100875, China.
| |
Collapse
|
7
|
Zheng ZY, Anurag M, Lei JT, Cao J, Singh P, Peng J, Kennedy H, Nguyen NC, Chen Y, Lavere P, Li J, Du XH, Cakar B, Song W, Kim BJ, Shi J, Seker S, Chan DW, Zhao GQ, Chen X, Banks KC, Lanman RB, Shafaee MN, Zhang XHF, Vasaikar S, Zhang B, Hilsenbeck SG, Li W, Foulds CE, Ellis MJ, Chang EC. Neurofibromin Is an Estrogen Receptor-α Transcriptional Co-repressor in Breast Cancer. Cancer Cell 2020; 37:387-402.e7. [PMID: 32142667 PMCID: PMC7286719 DOI: 10.1016/j.ccell.2020.02.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 11/15/2019] [Accepted: 02/06/2020] [Indexed: 12/18/2022]
Abstract
We report that neurofibromin, a tumor suppressor and Ras-GAP (GTPase-activating protein), is also an estrogen receptor-α (ER) transcriptional co-repressor through leucine/isoleucine-rich motifs that are functionally independent of GAP activity. GAP activity, in turn, does not affect ER binding. Consequently, neurofibromin depletion causes estradiol hypersensitivity and tamoxifen agonism, explaining the poor prognosis associated with neurofibromin loss in endocrine therapy-treated ER+ breast cancer. Neurofibromin-deficient ER+ breast cancer cells initially retain sensitivity to selective ER degraders (SERDs). However, Ras activation does play a role in acquired SERD resistance, which can be reversed upon MEK inhibitor addition, and SERD/MEK inhibitor combinations induce tumor regression. Thus, neurofibromin is a dual repressor for both Ras and ER signaling, and co-targeting may treat neurofibromin-deficient ER+ breast tumors.
Collapse
Affiliation(s)
- Ze-Yi Zheng
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Meenakshi Anurag
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Jonathan T Lei
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA; Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jin Cao
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Purba Singh
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Jianheng Peng
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA; Department of Physical Examination, the First Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Hilda Kennedy
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Nhu-Chau Nguyen
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Yue Chen
- Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA, USA
| | - Philip Lavere
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Jing Li
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Xin-Hui Du
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA; Department of Bone and Soft Tissue, Zhengzhou University Affiliated Henan Cancer Hospital and College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, P. R. China
| | - Burcu Cakar
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Wei Song
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Beom-Jun Kim
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Jiejun Shi
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Sinem Seker
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Doug W Chan
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Guo-Qiang Zhao
- Department of Bone and Soft Tissue, Zhengzhou University Affiliated Henan Cancer Hospital and College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, P. R. China
| | - Xi Chen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | | | | | - Maryam Nemati Shafaee
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Xiang H-F Zhang
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Suhas Vasaikar
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Bing Zhang
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Susan G Hilsenbeck
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Wei Li
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Charles E Foulds
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
| | - Matthew J Ellis
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA; Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| | - Eric C Chang
- Lester and Sue Smith Breast Center and Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
8
|
Han Q, Lv L, Wei J, Lei X, Lin H, Li G, Cao J, Xie J, Yang W, Wu S, You J, Lu J, Liu P, Min J. Vps4A mediates the localization and exosome release of β-catenin to inhibit epithelial-mesenchymal transition in hepatocellular carcinoma. Cancer Lett 2019; 457:47-59. [PMID: 31059752 DOI: 10.1016/j.canlet.2019.04.035] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 04/05/2019] [Accepted: 04/26/2019] [Indexed: 12/20/2022]
Abstract
We previously reported that Vps4A acted as a tumor suppressor by influencing the microRNA profiles of exosomes and their parental cells in hepatocellular carcinoma (HCC). However, the underlying mechanism and if Vps4A contributes to sorting proteins into exosomes are not well known. Here, we performed mass spectrometry analysis of the immunoprecipitated Vps4A complex and confirmed that Vps4A was associated with β-catenin and CHMP4B. Through this interaction, Vps4A promoted the plasma membrane (PM) localization and exosome release of β-catenin. Silencing Vps4A or CHMP4B decreased the PM localization and exosome sorting of β-catenin. Vps4A overexpression decreased β-catenin signaling pathway and inhibited epithelial-mesenchymal transition (EMT) and motility of HCC cells. And, silencing Vps4A or CHMP4B promoted EMT in HCC. Furthermore, the expression of Vps4A was significantly related to that of several EMT markers in HCC tissues and the level of exosomal β-catenin in patients with metastatic HCC was significantly lower compared to that of control patients. In conclusion, through the interaction with CHMP4B and β-catenin, Vps4A regulates the PM localization and exosome sorting of β-catenin, consequently decreases β-catenin signaling, and thereby inhibits EMT and metastasis in HCC.
Collapse
Affiliation(s)
- Qingfang Han
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China; Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Guangzhou, 510120, China
| | - Lihong Lv
- Clinical Trial Institution of Pharmaceuticals, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Jinxing Wei
- Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Guangzhou, 510120, China
| | - Xin Lei
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China
| | - Haoming Lin
- Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Guangzhou, 510120, China
| | - Guolin Li
- Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Guangzhou, 510120, China
| | - Jun Cao
- Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Guangzhou, 510120, China
| | - Jiyan Xie
- Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Guangzhou, 510120, China
| | - Weibang Yang
- Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Guangzhou, 510120, China
| | - Shaobin Wu
- Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Guangzhou, 510120, China
| | - Jia You
- Laboratory of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Jing Lu
- Laboratory of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Peiqing Liu
- Laboratory of Pharmacology and Toxicology, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Jun Min
- Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Guangzhou, 510120, China.
| |
Collapse
|
9
|
Abstract
The three RAS genes - HRAS, NRAS and KRAS - are collectively mutated in one-third of human cancers, where they act as prototypic oncogenes. Interestingly, there are rather distinct patterns to RAS mutations; the isoform mutated as well as the position and type of substitution vary between different cancers. As RAS genes are among the earliest, if not the first, genes mutated in a variety of cancers, understanding how these mutation patterns arise could inform on not only how cancer begins but also the factors influencing this event, which has implications for cancer prevention. To this end, we suggest that there is a narrow window or 'sweet spot' by which oncogenic RAS signalling can promote tumour initiation in normal cells. As a consequence, RAS mutation patterns in each normal cell are a product of the specific RAS isoform mutated, as well as the position of the mutation and type of substitution to achieve an ideal level of signalling.
Collapse
Affiliation(s)
- Siqi Li
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Allan Balmain
- Helen Diller Family Comprehensive Cancer Center and Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Christopher M Counter
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
10
|
Zheng ZY, Li J, Li F, Zhu Y, Cui K, Wong ST, Chang EC, Liao YH. Induction of N-Ras degradation by flunarizine-mediated autophagy. Sci Rep 2018; 8:16932. [PMID: 30446677 PMCID: PMC6240051 DOI: 10.1038/s41598-018-35237-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 10/29/2018] [Indexed: 12/13/2022] Open
Abstract
Ras GTPases are powerful drivers for tumorigenesis, but directly targeting Ras for treating cancer remains challenging. The growth and transforming activity of the aggressive basal-like breast cancer (BLBC) are driven by N-Ras. To target N-Ras in BLBC, this study screened existing pharmacologically active compounds for the new ability to induce N-Ras degradation, which led to the identification of flunarizine (FLN), previously approved for treating migraine and epilepsy. The FLN-induced N-Ras degradation was not affected by a 26S-proteasome inhibitor. Rather, it was blocked by autophagy inhibitors. Furthermore, N-Ras can be seen co-localized with active autophagosomes upon FLN treatment, suggesting that FLN alters the autophagy pathway to degrade N-Ras. Importantly, FLN treatment recapitulated the effect of N-RAS silencing in vitro by selectively inhibiting the growth of BLBC cells, but not that of breast cancer cells of other subtypes. In addition, in vivo FLN inhibited tumor growth of a BLBC xenograft model. In conclusion, this proof-of-principle study presents evidence that the autophagy pathway can be coerced by small molecule inhibitors, such as FLN, to degrade Ras as a strategy to treat cancer. FLN has low toxicity and should be further investigated to enrich the toolbox of cancer therapeutics.
Collapse
Affiliation(s)
- Ze-Yi Zheng
- Lester and Sue Smith Breast Center, and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jing Li
- Lester and Sue Smith Breast Center, and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
- Department of Oncology and Hematology, Hospital (TCM) Affiliated to Southwest Medical University, Luzhou, Sichuan, 646000, P. R. China
| | - Fuhai Li
- Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Yanqiao Zhu
- Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Kemi Cui
- Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Stephen T Wong
- Department of Systems Medicine and Bioengineering, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Eric C Chang
- Lester and Sue Smith Breast Center, and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Yi-Hua Liao
- Department of Dermatology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, 10002, Taiwan.
| |
Collapse
|
11
|
Tebar F, Enrich C, Rentero C, Grewal T. GTPases Rac1 and Ras Signaling from Endosomes. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2018; 57:65-105. [PMID: 30097772 DOI: 10.1007/978-3-319-96704-2_3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The endocytic compartment is not only the functional continuity of the plasma membrane but consists of a diverse collection of intracellular heterogeneous complex structures that transport, amplify, sustain, and/or sort signaling molecules. Over the years, it has become evident that early, late, and recycling endosomes represent an interconnected vesicular-tubular network able to form signaling platforms that dynamically and efficiently translate extracellular signals into biological outcome. Cell activation, differentiation, migration, death, and survival are some of the endpoints of endosomal signaling. Hence, to understand the role of the endosomal system in signal transduction in space and time, it is therefore necessary to dissect and identify the plethora of decoders that are operational in the different steps along the endocytic pathway. In this chapter, we focus on the regulation of spatiotemporal signaling in cells, considering endosomes as central platforms, in which several small GTPases proteins of the Ras superfamily, in particular Ras and Rac1, actively participate to control cellular processes like proliferation and cell mobility.
Collapse
Affiliation(s)
- Francesc Tebar
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain.
| | - Carlos Enrich
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
| | - Carles Rentero
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Centre de Recerca Biomèdica CELLEX, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Casanova 143, 08036, Barcelona, Spain
| | - Thomas Grewal
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
12
|
Juan T, Fürthauer M. Biogenesis and function of ESCRT-dependent extracellular vesicles. Semin Cell Dev Biol 2017; 74:66-77. [PMID: 28807885 DOI: 10.1016/j.semcdb.2017.08.022] [Citation(s) in RCA: 271] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 08/04/2017] [Accepted: 08/07/2017] [Indexed: 12/18/2022]
Abstract
From bacteria to humans, cells secrete a large variety of membrane-bound extracellular vesicles. Only relatively recently has it however started to become clear that the exovesicular transport of proteins and RNAs is important for normal physiology and numerous pathological conditions. Extracellular vesicles can be formed through the release of the intralumenal vesicles of multivesicular endosomes as so-called exosomes, or through direct, ectosomal, budding from the cell surface. Through their ability to promote the bending of membranes away from the cytoplasm, the components of the Endosomal Sorting Complex Required for Transport (ESCRT) have been implicated in both exo- and ectosomal biogenesis. Studies of the ESCRT machinery may therefore provide important insights into the formation and function of extracellular vesicles. In the present review, we first describe the cell biological mechanisms through which ESCRT components contribute to the biogenesis of different types of extracellular vesicles. We then discuss how recent functional studies have started to uncover important roles of ESCRT-dependent extracellular vesicles in a wide variety of processes, including the transport of developmental signaling molecules and embryonic morphogenesis, the regulation of social behavior and host-pathogen interactions, as well as the etiology and progression of neurodegenerative pathologies and cancer.
Collapse
Affiliation(s)
- Thomas Juan
- Université Côte d'Azur, CNRS, Inserm, iBV, France
| | | |
Collapse
|
13
|
Szymanska E, Budick-Harmelin N, Miaczynska M. Endosomal "sort" of signaling control: The role of ESCRT machinery in regulation of receptor-mediated signaling pathways. Semin Cell Dev Biol 2017; 74:11-20. [PMID: 28797837 DOI: 10.1016/j.semcdb.2017.08.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2017] [Revised: 07/24/2017] [Accepted: 08/04/2017] [Indexed: 12/31/2022]
Abstract
The endosomal sorting complexes required for transport (ESCRTs) machinery consists of four protein assemblies (ESCRT-0 to -III subcomplexes) which mediate various processes of membrane remodeling in the cell. In the endocytic pathway, ESCRTs sort cargo destined for degradation into intraluminal vesicles (ILVs) of endosomes. Cargos targeted by ESCRTs include various signaling molecules, mainly internalized cell-surface receptors but also some cytosolic proteins. It is therefore expected that aberrant trafficking caused by ESCRT dysfunction affects different signaling pathways. Here we review how perturbation of ESCRT activity alters intracellular transport of membrane receptors, causing their accumulation on endocytic compartments, decreased degradation and/or altered recycling to the plasma membrane. We further describe how perturbed trafficking of receptors impacts the activity of their downstream signaling pathways, with or without changes in transcriptional responses. Finally, we present evidence that ESCRT components can also control activity and intracellular distribution of cytosolic signaling proteins (kinases, other effectors and soluble receptors). The underlying mechanisms involve sequestration of such proteins in ILVs, their sorting for degradation or towards non-lysosomal destinations, and regulating their availability in various cellular compartments. All these ESCRT-mediated processes can modulate final outputs of multiple signaling pathways.
Collapse
Affiliation(s)
- Ewelina Szymanska
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland
| | - Noga Budick-Harmelin
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland; Cell Research and Immunology Department, George S. Wise Faculty of Life Sciences, Tel Aviv University, Israel
| | - Marta Miaczynska
- Laboratory of Cell Biology, International Institute of Molecular and Cell Biology, Warsaw, Poland.
| |
Collapse
|
14
|
Ren J, Yuan D, Xie L, Tao X, Duan C, Bao Y, He Y, Ge J, Lu H. Up-regulation of Vps4A promotes neuronal apoptosis after intracerebral hemorrhage in adult rats. Metab Brain Dis 2017; 32:565-575. [PMID: 28064406 DOI: 10.1007/s11011-016-9943-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 12/26/2016] [Indexed: 10/20/2022]
Abstract
Vps4, vacuolar protein sorting 4, belongs to ATPases Associated with diverse cellular Activities (AAA) protein family which is made up of Vps4A and Vps4B. Previous studies demonstrated that Vps4A plays vital roles in diverse aspects such as virus budding, the efficient transport of H-Ras to the PM (plasma membrane) and the involvement in the MVB (multivesiculate bodies) pathway. Interestingly, Vps4A is also expressed in the brain. However, the distribution and function of Vps4A in ICH diseases remain unclear. In this study, we show that Vps4A may be involved in neuronal apoptosis during pathophysiological processes of intracerebral hemorrhage (ICH). Based on the results of Western blot and immunohistochemistry, we found a remarkable up-regulation of Vps4A expression surrounding the hematoma after ICH. Double labeled immunofluorescence showed that Vps4A was co-expressed with NeuN but rarely with astrocytes and microglia. Morever, we detected that neuronal apoptosis marker active caspase-3 had co-localizations with Vps4A. Additionaly, Vps4A knockdown in vitro specifically leads to decreasing neuronal apoptosis coupled with increased Akt phosphorylation. All datas suggested that Vps4A was involved in promoting neuronal apoptosis via inhibiting Akt phosphorylation after ICH.
Collapse
Affiliation(s)
- Jianbing Ren
- Department of Rehabilitation, the Second People's Hospital of Nantong, Nantong, Jiangsu Province, 226001, China
| | - Debin Yuan
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, Jiangsu Province, 226001, China
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Lili Xie
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, Jiangsu Province, 226001, China
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Xuelei Tao
- Department of Rehabilitation, the Second People's Hospital of Nantong, Nantong, Jiangsu Province, 226001, China
| | - Chenwei Duan
- Department of Rehabilitation, the Second People's Hospital of Nantong, Nantong, Jiangsu Province, 226001, China
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Yifeng Bao
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, Jiangsu Province, 226001, China
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Yunfeng He
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong, Jiangsu Province, 226001, China
- Department of Neurosurgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, 226001, China
| | - Jianbin Ge
- Department of Rehabilitation, the Second People's Hospital of Nantong, Nantong, Jiangsu Province, 226001, China.
| | - Hongjian Lu
- Department of Rehabilitation, the Second People's Hospital of Nantong, Nantong, Jiangsu Province, 226001, China.
| |
Collapse
|
15
|
Nakhaeizadeh H, Amin E, Nakhaei-Rad S, Dvorsky R, Ahmadian MR. The RAS-Effector Interface: Isoform-Specific Differences in the Effector Binding Regions. PLoS One 2016; 11:e0167145. [PMID: 27936046 PMCID: PMC5147862 DOI: 10.1371/journal.pone.0167145] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 11/09/2016] [Indexed: 12/31/2022] Open
Abstract
RAS effectors specifically interact with the GTP-bound form of RAS in response to extracellular signals and link them to downstream signaling pathways. The molecular nature of effector interaction by RAS is well-studied but yet still incompletely understood in a comprehensive and systematic way. Here, structure-function relationships in the interaction between different RAS proteins and various effectors were investigated in detail by combining our in vitro data with in silico data. Equilibrium dissociation constants were determined for the binding of HRAS, KRAS, NRAS, RRAS1 and RRAS2 to both the RAS binding (RB) domain of CRAF and PI3Kα, and the RAS association (RA) domain of RASSF5, RALGDS and PLCε, respectively, using fluorescence polarization. An interaction matrix, constructed on the basis of available crystal structures, allowed identification of hotspots as critical determinants for RAS-effector interaction. New insights provided by this study are the dissection of the identified hotspots in five distinct regions (R1 to R5) in spite of high sequence variability not only between, but also within, RB/RA domain-containing effectors proteins. Finally, we propose that intermolecular β-sheet interaction in R1 is a central recognition region while R3 may determine specific contacts of RAS versus RRAS isoforms with effectors.
Collapse
Affiliation(s)
- Hossein Nakhaeizadeh
- Institute of Biochemistry and Molecular Biology II, Medical Faculty of the Heinrich-Heine University, Düsseldorf, Germany
| | - Ehsan Amin
- Institute of Biochemistry and Molecular Biology II, Medical Faculty of the Heinrich-Heine University, Düsseldorf, Germany
| | - Saeideh Nakhaei-Rad
- Institute of Biochemistry and Molecular Biology II, Medical Faculty of the Heinrich-Heine University, Düsseldorf, Germany
| | - Radovan Dvorsky
- Institute of Biochemistry and Molecular Biology II, Medical Faculty of the Heinrich-Heine University, Düsseldorf, Germany
| | - Mohammad Reza Ahmadian
- Institute of Biochemistry and Molecular Biology II, Medical Faculty of the Heinrich-Heine University, Düsseldorf, Germany
- * E-mail:
| |
Collapse
|
16
|
Luhtala N, Aslanian A, Yates JR, Hunter T. Secreted Glioblastoma Nanovesicles Contain Intracellular Signaling Proteins and Active Ras Incorporated in a Farnesylation-dependent Manner. J Biol Chem 2016; 292:611-628. [PMID: 27909058 DOI: 10.1074/jbc.m116.747618] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 11/11/2016] [Indexed: 12/21/2022] Open
Abstract
Glioblastomas (GBMs) are malignant brain tumors with a median survival of less than 18 months. Redundancy of signaling pathways represented within GBMs contributes to their therapeutic resistance. Exosomes are extracellular nanovesicles released from cells and present in human biofluids that represent a possible biomarker of tumor signaling state that could aid in personalized treatment. Herein, we demonstrate that mouse GBM cell-derived extracellular nanovesicles resembling exosomes from an H-RasV12 myr-Akt mouse model for GBM are enriched for intracellular signaling cascade proteins (GO: 0007242) and Ras protein signal transduction (GO: 0007265), and contain active Ras. Active Ras isolated from human and mouse GBM extracellular nanovesicles lysates using the Ras-binding domain of Raf also coprecipitates with ESCRT (endosomal sorting complex required for transport)-associated exosome proteins Vps4a and Alix. Although we initially hypothesized a role for active Ras protein signaling in exosome biogenesis, we found that GTP binding of K-Ras was dispensable for its packaging within extracellular nanovesicles and for the release of Alix. By contrast, farnesylation of K-Ras was required for its packaging within extracellular nanovesicles, yet expressing a K-Ras farnesylation mutant did not decrease the number of nanovesicles or the amount of Alix protein released per cell. Overall, these results emphasize the primary importance of membrane association in packaging of extracellular nanovesicle factors and indicate that screening nanovesicles within human fluids could provide insight into tissue origin and the wiring of signaling proteins at membranes to predict onset and behavior of cancer and other diseases linked to deregulated membrane signaling states.
Collapse
Affiliation(s)
- Natalie Luhtala
- From the Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California 92037 and
| | - Aaron Aslanian
- From the Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California 92037 and.,the Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California 92037
| | - John R Yates
- the Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California 92037
| | - Tony Hunter
- From the Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, California 92037 and
| |
Collapse
|
17
|
Kubala MH, Norwood SJ, Gomez GA, Jones A, Johnston W, Yap AS, Mureev S, Alexandrov K. Mammalian farnesyltransferase α subunit regulates vacuolar protein sorting-associated protein 4A (Vps4A)--dependent intracellular trafficking through recycling endosomes. Biochem Biophys Res Commun 2015; 468:580-6. [PMID: 26551458 DOI: 10.1016/j.bbrc.2015.10.148] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 10/27/2015] [Indexed: 11/19/2022]
Abstract
The protein farnesyltransferase (FTase) mediates posttranslational modification of proteins with isoprenoid lipids. FTase is a heterodimer and although the β subunit harbors the active site, it requires the α subunit for its activity. Here we explore the other functions of the FTase α subunit in addition to its established role in protein prenylation. We found that in the absence of the β subunit, the α subunit of FTase forms a stable autonomous dimeric structure in solution. We identify interactors of FTase α using mass spectrometry, followed by rapid in vitro analysis using the Leishmania tarentolae cell - free system. Vps4A was validated for direct binding to the FTase α subunit both in vitro and in vivo. Analysis of the interaction with Vps4A in Hek 293 cells demonstrated that FTase α controls trafficking of transferrin receptor upstream of this protein. These results point to the existence of previously undetected biological functions of the FTase α subunit that includes control of intracellular membrane trafficking.
Collapse
Affiliation(s)
- Marta H Kubala
- Institute for Molecular Bioscience, University of Queensland, Brisbane, St. Lucia, Queensland, 4072, Australia
| | - Suzanne J Norwood
- Institute for Molecular Bioscience, University of Queensland, Brisbane, St. Lucia, Queensland, 4072, Australia
| | - Guillermo A Gomez
- Institute for Molecular Bioscience, University of Queensland, Brisbane, St. Lucia, Queensland, 4072, Australia
| | - Alun Jones
- Institute for Molecular Bioscience, University of Queensland, Brisbane, St. Lucia, Queensland, 4072, Australia
| | - Wayne Johnston
- Institute for Molecular Bioscience, University of Queensland, Brisbane, St. Lucia, Queensland, 4072, Australia
| | - Alpha S Yap
- Institute for Molecular Bioscience, University of Queensland, Brisbane, St. Lucia, Queensland, 4072, Australia
| | - Sergey Mureev
- Institute for Molecular Bioscience, University of Queensland, Brisbane, St. Lucia, Queensland, 4072, Australia
| | - Kirill Alexandrov
- Institute for Molecular Bioscience, University of Queensland, Brisbane, St. Lucia, Queensland, 4072, Australia.
| |
Collapse
|
18
|
Wild-Type N-Ras, Overexpressed in Basal-like Breast Cancer, Promotes Tumor Formation by Inducing IL-8 Secretion via JAK2 Activation. Cell Rep 2015; 12:511-24. [PMID: 26166574 DOI: 10.1016/j.celrep.2015.06.044] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 05/21/2015] [Accepted: 06/14/2015] [Indexed: 01/09/2023] Open
Abstract
Basal-like breast cancers (BLBCs) are aggressive, and their drivers are unclear. We have found that wild-type N-RAS is overexpressed in BLBCs but not in other breast cancer subtypes. Repressing N-RAS inhibits transformation and tumor growth, whereas overexpression enhances these processes even in preinvasive BLBC cells. We identified N-Ras-responsive genes, most of which encode chemokines; e.g., IL8. Expression levels of these chemokines and N-RAS in tumors correlate with outcome. N-Ras, but not K-Ras, induces IL-8 by binding and activating the cytoplasmic pool of JAK2; IL-8 then acts on both the cancer cells and stromal fibroblasts. Thus, BLBC progression is promoted by increasing activities of wild-type N-Ras, which mediates autocrine/paracrine signaling that can influence both cancer and stroma cells.
Collapse
|
19
|
Legent K, Liu HH, Treisman JE. Drosophila Vps4 promotes Epidermal growth factor receptor signaling independently of its role in receptor degradation. Development 2015; 142:1480-91. [PMID: 25790850 DOI: 10.1242/dev.117960] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Accepted: 02/20/2015] [Indexed: 12/12/2022]
Abstract
Endocytic trafficking of signaling receptors is an important mechanism for limiting signal duration. Components of the Endosomal Sorting Complexes Required for Transport (ESCRT), which target ubiquitylated receptors to intra-lumenal vesicles (ILVs) of multivesicular bodies, are thought to terminate signaling by the epidermal growth factor receptor (EGFR) and direct it for lysosomal degradation. In a genetic screen for mutations that affect Drosophila eye development, we identified an allele of Vacuolar protein sorting 4 (Vps4), which encodes an AAA ATPase that interacts with the ESCRT-III complex to drive the final step of ILV formation. Photoreceptors are largely absent from Vps4 mutant clones in the eye disc, and even when cell death is genetically prevented, the mutant R8 photoreceptors that develop fail to recruit surrounding cells to differentiate as R1-R7 photoreceptors. This recruitment requires EGFR signaling, suggesting that loss of Vps4 disrupts the EGFR pathway. In imaginal disc cells mutant for Vps4, EGFR and other receptors accumulate in endosomes and EGFR target genes are not expressed; epistasis experiments place the function of Vps4 at the level of the receptor. Surprisingly, Vps4 is required for EGFR signaling even in the absence of Shibire, the Dynamin that internalizes EGFR from the plasma membrane. In ovarian follicle cells, in contrast, Vps4 does not affect EGFR signaling, although it is still essential for receptor degradation. Taken together, these findings indicate that Vps4 can promote EGFR activity through an endocytosis-independent mechanism.
Collapse
Affiliation(s)
- Kevin Legent
- Kimmel Center for Biology and Medicine of the Skirball Institute and Department of Cell Biology, NYU School of Medicine, 540 First Avenue, New York, NY 10016, USA
| | - Hui Hua Liu
- Kimmel Center for Biology and Medicine of the Skirball Institute and Department of Cell Biology, NYU School of Medicine, 540 First Avenue, New York, NY 10016, USA
| | - Jessica E Treisman
- Kimmel Center for Biology and Medicine of the Skirball Institute and Department of Cell Biology, NYU School of Medicine, 540 First Avenue, New York, NY 10016, USA
| |
Collapse
|
20
|
Mattissek C, Teis D. The role of the endosomal sorting complexes required for transport (ESCRT) in tumorigenesis. Mol Membr Biol 2014; 31:111-9. [PMID: 24641493 PMCID: PMC4059258 DOI: 10.3109/09687688.2014.894210] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Revised: 01/30/2014] [Accepted: 02/07/2014] [Indexed: 11/30/2022]
Abstract
The endosomal sorting complexes required for transport (ESCRT) are needed for three distinct cellular functions in higher eukaryotes: (i) Multivesicular body formation for the degradation of transmembrane proteins in lysosomes, (ii) midbody abscission during cytokinesis and (iii) retroviral budding. Not surprisingly, loss of ESCRT function has severe consequences, which include the failure to down-regulate growth factor receptors leading to deregulated mitogenic signaling. While it is clear that the function of the ESCRT machinery is important for embryonic development, its role in cancer is more controversial. Various experimental approaches in different model organisms arrive at partially divergent conclusions regarding the contribution of ESCRTs to tumorigenesis. Therefore the aim of this review is to provide an overview on different model systems used to study the role of the ESCRT machinery in cancer development, to highlight common grounds and present certain controversies in the field.
Collapse
Affiliation(s)
- Claudia Mattissek
- Division of Cell Biology, Biocenter, Innsbruck Medical University
InnsbruckAustria
| | - David Teis
- Division of Cell Biology, Biocenter, Innsbruck Medical University
InnsbruckAustria
| |
Collapse
|
21
|
Young E, Zheng ZY, Wilkins AD, Jeong HT, Li M, Lichtarge O, Chang EC. Regulation of Ras localization and cell transformation by evolutionarily conserved palmitoyltransferases. Mol Cell Biol 2014; 34:374-85. [PMID: 24248599 PMCID: PMC3911504 DOI: 10.1128/mcb.01248-13] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 10/16/2013] [Accepted: 11/09/2013] [Indexed: 01/06/2023] Open
Abstract
Ras can act on the plasma membrane (PM) to mediate extracellular signaling and tumorigenesis. To identify key components controlling Ras PM localization, we performed an unbiased screen to seek Schizosaccharomyces pombe mutants with reduced PM Ras. Five mutants were found with mutations affecting the same gene, S. pombe erf2 (sp-erf2), encoding sp-Erf2, a palmitoyltransferase, with various activities. sp-Erf2 localizes to the trans-Golgi compartment, a process which is mediated by its third transmembrane domain and the Erf4 cofactor. In fission yeast, the human ortholog zDHHC9 rescues the phenotypes of sp-erf2 null cells. In contrast, expressing zDHHC14, another sp-Erf2-like human protein, did not rescue Ras1 mislocalization in these cells. Importantly, ZDHHC9 is widely overexpressed in cancers. Overexpressing ZDHHC9 promotes, while repressing it diminishes, Ras PM localization and transformation of mammalian cells. These data strongly demonstrate that sp-Erf2/zDHHC9 palmitoylates Ras proteins in a highly selective manner in the trans-Golgi compartment to facilitate PM targeting via the trans-Golgi network, a role that is most certainly critical for Ras-driven tumorigenesis.
Collapse
Affiliation(s)
- Evelin Young
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
| | - Ze-Yi Zheng
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
| | - Angela D. Wilkins
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- CIBR Center for Computational and Integrative Biomedical Research, Baylor College of Medicine, Houston, Texas, USA
| | - Hee-Tae Jeong
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
| | - Min Li
- Department of Oncology, Nanjing Hospital of Traditional Chinese Medicine, Nanjing, Jiangsu, China
| | - Olivier Lichtarge
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA
- CIBR Center for Computational and Integrative Biomedical Research, Baylor College of Medicine, Houston, Texas, USA
| | - Eric C. Chang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
22
|
A bimolecular fluorescent complementation screen reveals complex roles of endosomes in Ras-mediated signaling. Methods Enzymol 2014; 535:25-38. [PMID: 24377915 DOI: 10.1016/b978-0-12-397925-4.00002-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
While Ras GTPases are best known for mediating growth factor signaling on the plasma membrane, these proteins also have surprisingly complex activities in the endosome. Assisted by a method called bimolecular fluorescent complementation (BiFC), which can detect weak and transient protein-protein interactions and reveal where the binding takes place in live cells, we have identified three effectors, Cdc42, CHMP6, and VPS4A that interact with Ras proteins in endosomes. These effectors are all necessary for Ras-induced transformation, suggesting that for Ras proteins to efficiently induce tumor formation, they must also activate effectors in cytoplasm, such as those in endosomes. Here, we describe how BiFC can be used to detect and screen for Ras effectors and for readily revealing where in the cell the binding occurs.
Collapse
|
23
|
Abstract
The endosomal sorting complexes required for transport (ESCRT) pathway was initially defined in yeast genetic screens that identified the factors necessary to sort membrane proteins into intraluminal endosomal vesicles. Subsequent studies have revealed that the mammalian ESCRT pathway also functions in a series of other key cellular processes, including formation of extracellular microvesicles, enveloped virus budding, and the abscission stage of cytokinesis. The core ESCRT machinery comprises Bro1 family proteins and ESCRT-I, ESCRT-II, ESCRT-III, and VPS4 complexes. Site-specific adaptors recruit these soluble factors to assemble on different cellular membranes, where they carry out membrane fission reactions. ESCRT-III proteins form filaments that draw membranes together from the cytoplasmic face, and mechanistic models have been advanced to explain how ESCRT-III filaments and the VPS4 ATPase can work together to catalyze membrane fission.
Collapse
Affiliation(s)
- John McCullough
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, Utah 84112-5650, USA
| | | | | |
Collapse
|
24
|
Makvandi-Nejad S, Hoffman GE, Allen JJ, Chu E, Gu E, Chandler AM, Loredo AI, Bellone RR, Mezey JG, Brooks SA, Sutter NB. Four loci explain 83% of size variation in the horse. PLoS One 2012; 7:e39929. [PMID: 22808074 PMCID: PMC3394777 DOI: 10.1371/journal.pone.0039929] [Citation(s) in RCA: 151] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 05/29/2012] [Indexed: 01/09/2023] Open
Abstract
Horse body size varies greatly due to intense selection within each breed. American Miniatures are less than one meter tall at the withers while Shires and Percherons can exceed two meters. The genetic basis for this variation is not known. We hypothesize that the breed population structure of the horse should simplify efforts to identify genes controlling size. In support of this, here we show with genome-wide association scans (GWAS) that genetic variation at just four loci can explain the great majority of horse size variation. Unlike humans, which are naturally reproducing and possess many genetic variants with weak effects on size, we show that horses, like other domestic mammals, carry just a small number of size loci with alleles of large effect. Furthermore, three of our horse size loci contain the LCORL, HMGA2 and ZFAT genes that have previously been found to control human height. The LCORL/NCAPG locus is also implicated in cattle growth and HMGA2 is associated with dog size. Extreme size diversification is a hallmark of domestication. Our results in the horse, complemented by the prior work in cattle and dog, serve to pinpoint those very few genes that have played major roles in the rapid evolution of size during domestication.
Collapse
Affiliation(s)
- Shokouh Makvandi-Nejad
- Department of Clinical Sciences, Cornell University, Ithaca, New York, United States of America
| | - Gabriel E. Hoffman
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, United States of America
| | - Jeremy J. Allen
- Department of Clinical Sciences, Cornell University, Ithaca, New York, United States of America
| | - Erin Chu
- Department of Clinical Sciences, Cornell University, Ithaca, New York, United States of America
| | - Esther Gu
- Department of Clinical Sciences, Cornell University, Ithaca, New York, United States of America
| | - Alyssa M. Chandler
- Department of Clinical Sciences, Cornell University, Ithaca, New York, United States of America
| | - Ariel I. Loredo
- Department of Clinical Sciences, Cornell University, Ithaca, New York, United States of America
- Biology Department, La Sierra University, Riverside, California, United States of America
| | - Rebecca R. Bellone
- Department of Biology, University of Tampa, Tampa, Florida, United States of America
| | - Jason G. Mezey
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, New York, United States of America
| | - Samantha A. Brooks
- Department of Animal Science, Cornell University, Ithaca, New York, United States of America
| | - Nathan B. Sutter
- Department of Clinical Sciences, Cornell University, Ithaca, New York, United States of America
- * E-mail:
| |
Collapse
|
25
|
Abstract
Ras proteins are best known to function on the plasma membrane to mediate growth factor signaling. Controlling the length of time that Ras proteins stay on the plasma membrane is an effective way to properly modulate the intensity and duration of growth factor signaling. It has been shown previously that H- and N-Ras proteins in the GTP-bound state can be ubiquitylated via a K-63 linkage, which leads to endosome internalization and results in a negative-feedback loop for efficient signal attenuation. In a more recent study, two new Ras effectors have been isolated, CHMP6 and VPS4A, which are components of the ESCRT-III complex, best known for mediating protein sorting in the endosomes. Surprisingly, these molecules are required for efficient Ras-induced transformation. They apparently do so by controlling recycling of components of the Ras pathway back to the plasma membrane, thus creating a positive-feedback loop to enhance growth factor signaling. These results suggest the fates of endosomal Ras proteins are complex and dynamic — they can be either stored and/or destroyed or recycled. Further work is needed to decipher how the fate of these endosomal Ras proteins is determined.
Collapse
Affiliation(s)
- Ze-Yi Zheng
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA
| | | | | | | |
Collapse
|