1
|
Ma C, Luo C, Deng F, Yu C, Chen Y, Zhong G, Zhan Y, Nie L, Huang Y, Xia Y, Cai Z, Xu K, Cai H, Wang F, Lu Z, Zeng X, Zhu Y, Liu S. Major vault protein directly enhances adaptive immunity induced by Influenza A virus or indirectly through innate immunity. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167441. [PMID: 39069011 DOI: 10.1016/j.bbadis.2024.167441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 07/05/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024]
Abstract
As we previously revealed, major vault protein (MVP) is a virus-induced host factor, and its expression is crucial for innate immune responses. Nevertheless, the function of MVP in adaptive immunity is poorly known. Here, we demonstrate that Mvp knockout mice had attenuated antibody responses and reduced survival after rechallenge with homologous influenza A virus (IAV) relative to wild-type mice. Analysis of B cell populations showed that MVP promoted germinal center (GC) responses to develop optimal antiviral humoral immunity. Although MVP-deficient T cells and dendritic cells (DCs) were not intrinsically damaged, MVP promoted activating effector T cells and T follicular helper responses and regulated specific DC subsets. These findings suggest that MVP directs an effective adaptive immune response against IAV by directly engaging in GC reactions or indirectly augmenting cellular immunity via innate immune pathways.
Collapse
Affiliation(s)
- Caijiao Ma
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Chuanjin Luo
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Feiyan Deng
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Chen Yu
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yumeng Chen
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Gechang Zhong
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yuxin Zhan
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Longyu Nie
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yu Huang
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yongfang Xia
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Zeng Cai
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Ke Xu
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Huanhuan Cai
- Institute of Myocardial Injury and Repair, Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430072, China
| | - Fubing Wang
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan 430072, China
| | - Zhibing Lu
- Institute of Myocardial Injury and Repair, Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430072, China
| | - Xiangtai Zeng
- Department of General Surgery, Ganzhou Key Laboratory of Thyroid Cancer, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi 341000, China
| | - Ying Zhu
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China.
| | - Shi Liu
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, College of Life Sciences, Wuhan University, Wuhan 430072, China; Institute of Myocardial Injury and Repair, Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430072, China; Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan 430072, China; Department of General Surgery, Ganzhou Key Laboratory of Thyroid Cancer, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi 341000, China.
| |
Collapse
|
2
|
Lee YF, Phua CZJ, Yuan J, Zhang B, Lee MY, Kannan S, Chiu YHJ, Koh CWQ, Yap CK, Lim EKH, Chen J, Lim Y, Lee JJH, Skanderup AJ, Wang Z, Zhai W, Tan NS, Verma CS, Tay Y, Tan DSW, Tam WL. PARP4 interacts with hnRNPM to regulate splicing during lung cancer progression. Genome Med 2024; 16:91. [PMID: 39034402 PMCID: PMC11265163 DOI: 10.1186/s13073-024-01328-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/02/2024] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND The identification of cancer driver genes from sequencing data has been crucial in deepening our understanding of tumor biology and expanding targeted therapy options. However, apart from the most commonly altered genes, the mechanisms underlying the contribution of other mutations to cancer acquisition remain understudied. Leveraging on our whole-exome sequencing of the largest Asian lung adenocarcinoma (LUAD) cohort (n = 302), we now functionally assess the mechanistic role of a novel driver, PARP4. METHODS In vitro and in vivo tumorigenicity assays were used to study the functional effects of PARP4 loss and mutation in multiple lung cancer cell lines. Interactomics analysis by quantitative mass spectrometry was conducted to identify PARP4's interaction partners. Transcriptomic data from cell lines and patient tumors were used to investigate splicing alterations. RESULTS PARP4 depletion or mutation (I1039T) promotes the tumorigenicity of KRAS- or EGFR-driven lung cancer cells. Disruption of the vault complex, with which PARP4 is commonly associated, did not alter tumorigenicity, indicating that PARP4's tumor suppressive activity is mediated independently. The splicing regulator hnRNPM is a potentially novel PARP4 interaction partner, the loss of which likewise promotes tumor formation. hnRNPM loss results in splicing perturbations, with a propensity for dysregulated intronic splicing that was similarly observed in PARP4 knockdown cells and in LUAD cohort patients with PARP4 copy number loss. CONCLUSIONS PARP4 is a novel modulator of lung adenocarcinoma, where its tumor suppressive activity is mediated not through the vault complex-unlike conventionally thought, but in association with its novel interaction partner hnRNPM, thus suggesting a role for splicing dysregulation in LUAD tumorigenesis.
Collapse
Affiliation(s)
- Yi Fei Lee
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Singapore
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Cheryl Zi Jin Phua
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Singapore
| | - Ju Yuan
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Singapore
| | - Bin Zhang
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore, 117599, Singapore
- Computational Bioscience Research Center, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- Computer Science Program, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - May Yin Lee
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Singapore
| | - Srinivasaraghavan Kannan
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, Matrix, Singapore, 138671, Singapore
| | - Yui Hei Jasper Chiu
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Singapore
| | - Casslynn Wei Qian Koh
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Singapore
| | - Choon Kong Yap
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Singapore
| | - Edwin Kok Hao Lim
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Singapore
| | - Jianbin Chen
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Singapore
| | - Yuhua Lim
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Singapore
| | - Jane Jia Hui Lee
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Singapore
| | - Anders Jacobsen Skanderup
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Singapore
| | - Zhenxun Wang
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Singapore
- Centre for Vision Research, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Weiwei Zhai
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Singapore
- Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China
| | - Nguan Soon Tan
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore, 308232, Singapore
| | - Chandra S Verma
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, Matrix, Singapore, 138671, Singapore
- Department of Biological Sciences, National University of Singapore, 16 Science Drive 4, Singapore, 117558, Singapore
| | - Yvonne Tay
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore, 117599, Singapore
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore, 117599, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 8 Medical Drive, Singapore, 117597, Singapore
| | - Daniel Shao Weng Tan
- Division of Medical Oncology, National Cancer Centre Singapore, 30 Hospital Boulevard, Singapore, 168583, Singapore
| | - Wai Leong Tam
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, Genome, Singapore, 138672, Singapore.
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Singapore, 117599, Singapore.
- NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore, 117599, Singapore.
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 8 Medical Drive, Singapore, 117597, Singapore.
| |
Collapse
|
3
|
Slinning MS, Nthiga TM, Eichner C, Khadija S, Rome LH, Nilsen F, Dondrup M. Major vault protein is part of an extracellular cement material in the Atlantic salmon louse (Lepeophtheirus salmonis). Sci Rep 2024; 14:15240. [PMID: 38956386 PMCID: PMC11219742 DOI: 10.1038/s41598-024-65683-0] [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: 03/06/2024] [Accepted: 06/24/2024] [Indexed: 07/04/2024] Open
Abstract
Major vault protein (MVP) is the main component of the vault complex, which is a highly conserved ribonucleoprotein complex found in most eukaryotic organisms. MVP or vaults have previously been found to be overexpressed in multidrug-resistant cancer cells and implicated in various cellular processes such as cell signaling and innate immunity. The precise function of MVP is, however, poorly understood and its expression and probable function in lower eukaryotes are not well characterized. In this study, we report that the Atlantic salmon louse expresses three full-length MVP paralogues (LsMVP1-3). Furthermore, we extended our search and identified MVP orthologues in several other ecdysozoan species. LsMVPs were shown to be expressed in various tissues at both transcript and protein levels. In addition, evidence for LsMVP to assemble into vaults was demonstrated by performing differential centrifugation. LsMVP was found to be highly expressed in cement, an extracellular material produced by a pair of cement glands in the adult female salmon louse. Cement is important for the formation of egg strings that serve as protective coats for developing embryos. Our results imply a possible novel function of LsMVP as a secretory cement protein. LsMVP may play a role in structural or reproductive functions, although this has to be further investigated.
Collapse
Affiliation(s)
- Malene Skuseth Slinning
- Sea Lice Research Centre (SLRC), Department of Biological Sciences, University of Bergen, Pb. 7803, 5020, Bergen, Norway
| | - Thaddaeus Mutugi Nthiga
- Sea Lice Research Centre (SLRC), Department of Biological Sciences, University of Bergen, Pb. 7803, 5020, Bergen, Norway
| | - Christiane Eichner
- Sea Lice Research Centre (SLRC), Department of Biological Sciences, University of Bergen, Pb. 7803, 5020, Bergen, Norway
| | - Syeda Khadija
- Department of Biological Chemistry, David Geffen School of Medicine and the California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Leonard H Rome
- Department of Biological Chemistry, David Geffen School of Medicine and the California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Frank Nilsen
- Sea Lice Research Centre (SLRC), Department of Biological Sciences, University of Bergen, Pb. 7803, 5020, Bergen, Norway
| | - Michael Dondrup
- SLRC, Computational Biology Unit (CBU), Department of Informatics, University of Bergen, Pb. 7803, 5020, Bergen, Norway.
| |
Collapse
|
4
|
Sakamoto K, Yamamoto Y, Inaba H, Matsuura K. Strategy toward In-Cell Self-Assembly of an Artificial Viral Capsid from a Fluorescent Protein-Modified β-Annulus Peptide. ACS Synth Biol 2024; 13:1842-1850. [PMID: 38729919 DOI: 10.1021/acssynbio.4c00135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2024]
Abstract
In-cell self-assembly of natural viral capsids is an event that can be visualized under transmission electron microscopy (TEM) observations. By mimicking the self-assembly of natural viral capsids, various artificial protein- and peptide-based nanocages were developed; however, few studies have reported the in-cell self-assembly of such nanocages. Our group developed a β-Annulus peptide that can form a nanocage called artificial viral capsid in vitro, but in-cell self-assembly of the capsid has not been achieved. Here, we designed an artificial viral capsid decorated with a fluorescent protein, StayGold, to visualize in-cell self-assembly. Fluorescence anisotropy measurements and fluorescence resonance energy transfer imaging, in addition to TEM observations of the cells and super-resolution microscopy, revealed that StayGold-conjugated β-Annulus peptides self-assembled into the StayGold-decorated artificial viral capsid in a cell. Using these techniques, we achieved the in-cell self-assembly of an artificial viral capsid.
Collapse
Affiliation(s)
- Kentarou Sakamoto
- Department of Chemistry and Biotechnology, Graduate School of Engineering, Tottori University, Tottori 680-8552, Japan
| | - Yuka Yamamoto
- Department of Chemistry and Biotechnology, Graduate School of Engineering, Tottori University, Tottori 680-8552, Japan
| | - Hiroshi Inaba
- Department of Chemistry and Biotechnology, Graduate School of Engineering, Tottori University, Tottori 680-8552, Japan
- Centre for Research on Green Sustainable Chemistry, Tottori University, Tottori 680-8552, Japan
| | - Kazunori Matsuura
- Department of Chemistry and Biotechnology, Graduate School of Engineering, Tottori University, Tottori 680-8552, Japan
- Centre for Research on Green Sustainable Chemistry, Tottori University, Tottori 680-8552, Japan
| |
Collapse
|
5
|
Tomaino G, Pantaleoni C, D’Urzo A, Santambrogio C, Testa F, Ciprandi M, Cotugno D, Frascotti G, Vanoni M, Tortora P. An Efficient Method for Vault Nanoparticle Conjugation with Finely Adjustable Amounts of Antibodies and Small Molecules. Int J Mol Sci 2024; 25:6629. [PMID: 38928334 PMCID: PMC11203631 DOI: 10.3390/ijms25126629] [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: 04/30/2024] [Revised: 06/07/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Vaults are eukaryotic ribonucleoproteins consisting of 78 copies of the major vault protein (MVP), which assemble into a nanoparticle with an about 60 nm volume-based size, enclosing other proteins and RNAs. Regardless of their physiological role(s), vaults represent ideal, natural hollow nanoparticles, which are produced by the assembly of the sole MVP. Here, we have expressed in Komagataella phaffi and purified an MVP variant carrying a C-terminal Z peptide (vault-Z), which can tightly bind an antibody's Fc portion, in view of targeted delivery. Via surface plasmon resonance analysis, we could determine a 2.5 nM affinity to the monoclonal antibody Trastuzumab (Tz)/vault-Z 1:1 interaction. Then, we characterized the in-solution interaction via co-incubation, ultracentrifugation, and analysis of the pelleted proteins. This showed virtually irreversible binding up to an at least 10:1 Tz/vault-Z ratio. As a proof of concept, we labeled the Fc portion of Tz with a fluorophore and conjugated it with the nanoparticle, along with either Tz or Cetuximab, another monoclonal antibody. Thus, we could demonstrate antibody-dependent, selective uptake by the SKBR3 and MDA-MB 231 breast cancer cell lines. These investigations provide a novel, flexible technological platform that significantly extends vault-Z's applications, in that it can be stably conjugated with finely adjusted amounts of antibodies as well as of other molecules, such as fluorophores, cell-targeting peptides, or drugs, using the Fc portion as a scaffold.
Collapse
Affiliation(s)
- Giulia Tomaino
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy; (G.T.); (C.P.); (A.D.); (C.S.); (F.T.); (M.C.); (D.C.); (M.V.)
| | - Camilla Pantaleoni
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy; (G.T.); (C.P.); (A.D.); (C.S.); (F.T.); (M.C.); (D.C.); (M.V.)
| | - Annalisa D’Urzo
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy; (G.T.); (C.P.); (A.D.); (C.S.); (F.T.); (M.C.); (D.C.); (M.V.)
| | - Carlo Santambrogio
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy; (G.T.); (C.P.); (A.D.); (C.S.); (F.T.); (M.C.); (D.C.); (M.V.)
| | - Filippo Testa
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy; (G.T.); (C.P.); (A.D.); (C.S.); (F.T.); (M.C.); (D.C.); (M.V.)
| | - Matilde Ciprandi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy; (G.T.); (C.P.); (A.D.); (C.S.); (F.T.); (M.C.); (D.C.); (M.V.)
| | - Davide Cotugno
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy; (G.T.); (C.P.); (A.D.); (C.S.); (F.T.); (M.C.); (D.C.); (M.V.)
| | - Gianni Frascotti
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy; (G.T.); (C.P.); (A.D.); (C.S.); (F.T.); (M.C.); (D.C.); (M.V.)
| | - Marco Vanoni
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy; (G.T.); (C.P.); (A.D.); (C.S.); (F.T.); (M.C.); (D.C.); (M.V.)
- ISBE-SYSBIO Centre for Systems Biology, 20126 Milan, Italy
| | - Paolo Tortora
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milan, Italy; (G.T.); (C.P.); (A.D.); (C.S.); (F.T.); (M.C.); (D.C.); (M.V.)
| |
Collapse
|
6
|
Aghajani Mir M. Vault RNAs (vtRNAs): Rediscovered non-coding RNAs with diverse physiological and pathological activities. Genes Dis 2024; 11:772-787. [PMID: 37692527 PMCID: PMC10491885 DOI: 10.1016/j.gendis.2023.01.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Accepted: 01/16/2023] [Indexed: 04/05/2023] Open
Abstract
The physicochemical characteristics of RNA admit non-coding RNAs to perform a different range of biological acts through various mechanisms and are involved in regulating a diversity of fundamental processes. Notably, some reports of pathological conditions have proved abnormal expression of many non-coding RNAs guides the ailment. Vault RNAs are a class of non-coding RNAs containing stem regions or loops with well-conserved sequence patterns that play a fundamental role in the function of vault particles through RNA-ligand, RNA-RNA, or RNA-protein interactions. Taken together, vault RNAs have been proposed to be involved in a variety of functions such as cell proliferation, nucleocytoplasmic transport, intracellular detoxification processes, multidrug resistance, apoptosis, and autophagy, and serve as microRNA precursors and signaling pathways. Despite decades of investigations devoted, the biological function of the vault particle or the vault RNAs is not yet completely cleared. In this review, the current scientific assertions of the vital vault RNAs functions were discussed.
Collapse
Affiliation(s)
- Mahsa Aghajani Mir
- Deputy of Research and Technology, Health Research Institute, Babol University of Medical Sciences, Babol 47176-4774, Iran
| |
Collapse
|
7
|
Yu C, Zhu Q, Ma C, Luo C, Nie L, Cai H, Wang Q, Wang F, Ren H, Yan H, Xu K, Zhou L, Zhang C, Lu G, Lu Z, Zhu Y, Liu S. Major vault protein regulates tumor-associated macrophage polarization through interaction with signal transducer and activator of transcription 6. Front Immunol 2024; 14:1289795. [PMID: 38264642 PMCID: PMC10803552 DOI: 10.3389/fimmu.2023.1289795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 12/21/2023] [Indexed: 01/25/2024] Open
Abstract
Tumor-associated macrophages (TAMs) are critical in the tumor microenvironment (TME) of hepatocellular carcinoma (HCC). Major vault protein (MVP) mediates multidrug resistance, cell growth and development, and viral immunity. However, the relationship between MVP and TAMs polarization has not been clarified in HCC. We found that MVP significantly increased M2-TAMs infiltration levels in tumor tissues of HCC patients. MVP promoted HCC proliferation, metastasis, and invasion by regulating M2 polarization in vivo and in vitro. Mechanistically, MVP associated with signal transducer and activator of transcription 6 (STAT6) and enhanced STAT6 phosphorylation. STAT6 translocated from the cytosol to the nucleus and regulated M2 macrophage-associated gene transcription. These findings suggest that MVP modulates the macrophage M2 transcriptional program, revealing its potential role in the TAMs of TME.
Collapse
Affiliation(s)
- Chen Yu
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan, China
| | - Qingmei Zhu
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan, China
| | - Caijiao Ma
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan, China
| | - Chuanjin Luo
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan, China
| | - Longyu Nie
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan, China
| | - Huanhuan Cai
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, China
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qiming Wang
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan, Changsha, China
| | - Fubing Wang
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China
| | - Hong Ren
- Shanghai Children’s Medical Center, Affiliated Hospital to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huan Yan
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan, China
| | - Ke Xu
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan, China
| | - Li Zhou
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan, China
| | - Caiyan Zhang
- Shanghai Children’s Medical Center, Affiliated Hospital to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guoping Lu
- Shanghai Children’s Medical Center, Affiliated Hospital to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhibing Lu
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, China
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ying Zhu
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan, China
| | - Shi Liu
- State Key Laboratory of Virology, Modern Virology Research Center, Frontier Science Center for Immunology and Metabolism, College of Life Sciences, Wuhan University, Wuhan, China
- Institute of Myocardial Injury and Repair, Wuhan University, Wuhan, China
- College of Bioscience and Biotechnology, Hunan Agricultural University, Hunan, Changsha, China
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China
| |
Collapse
|
8
|
González-Álamos M, Guerra P, Verdaguer N. Structure, Dynamics and Functional Implications of the Eukaryotic Vault Complex. Subcell Biochem 2024; 104:531-548. [PMID: 38963499 DOI: 10.1007/978-3-031-58843-3_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
Vault ribonucleoprotein particles are naturally designed nanocages, widely found in the eukaryotic kingdom. Vaults consist of 78 copies of the major vault protein (MVP) that are organized in 2 symmetrical cup-shaped halves, of an approximate size of 70x40x40 nm, leaving a huge internal cavity which accommodates the vault poly(ADP-ribose) polymerase (vPARP), the telomerase-associated protein-1 (TEP1) and some small untranslated RNAs. Diverse hypotheses have been developed on possible functions of vaults, based on their unique capsular structure, their rapid movements and the distinct subcellular localization of the particles, implicating transport of cargo, but they are all pending confirmation. Vault particles also possess many attributes that can be exploited in nanobiotechnology, particularly in the creation of vehicles for the delivery of multiple molecular cargoes. Here we review what is known about the structure and dynamics of the vault complex and discuss a possible mechanism for the vault opening process. The recent findings in the characterization of the vaults in cells and in its natural microenvironment will be also discussed.
Collapse
Affiliation(s)
- María González-Álamos
- Structural and Molecular Biology Department, Instituto de Biología Molecular de Barcelona (IBMB-CSIC), Barcelona, Spain
| | - Pablo Guerra
- Cryo-Electron Microscopy Platform - IBMB CSIC, Joint Electron Microscopy Center at ALBA (JEMCA), Barcelona, Spain
| | - Núria Verdaguer
- Structural and Molecular Biology Department, Instituto de Biología Molecular de Barcelona (IBMB-CSIC), Barcelona, Spain.
| |
Collapse
|
9
|
Kretz PF, Wagner C, Mikhaleva A, Montillot C, Hugel S, Morella I, Kannan M, Fischer MC, Milhau M, Yalcin I, Brambilla R, Selloum M, Herault Y, Reymond A, Collins SC, Yalcin B. Dissecting the autism-associated 16p11.2 locus identifies multiple drivers in neuroanatomical phenotypes and unveils a male-specific role for the major vault protein. Genome Biol 2023; 24:261. [PMID: 37968726 PMCID: PMC10647150 DOI: 10.1186/s13059-023-03092-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 10/18/2023] [Indexed: 11/17/2023] Open
Abstract
BACKGROUND Using mouse genetic studies and systematic assessments of brain neuroanatomical phenotypes, we set out to identify which of the 30 genes causes brain defects at the autism-associated 16p11.2 locus. RESULTS We show that multiple genes mapping to this region interact to regulate brain anatomy, with female mice exhibiting far fewer brain neuroanatomical phenotypes. In male mice, among the 13 genes associated with neuroanatomical defects (Mvp, Ppp4c, Zg16, Taok2, Slx1b, Maz, Fam57b, Bola2, Tbx6, Qprt, Spn, Hirip3, and Doc2a), Mvp is the top driver implicated in phenotypes pertaining to brain, cortex, hippocampus, ventricles, and corpus callosum sizes. The major vault protein (MVP), the main component of the vault organelle, is a conserved protein found in eukaryotic cells, yet its function is not understood. Here, we find MVP expression highly specific to the limbic system and show that Mvp regulates neuronal morphology, postnatally and specifically in males. We also recapitulate a previously reported genetic interaction and show that Mvp+/-;Mapk3+/- mice exhibit behavioral deficits, notably decreased anxiety-like traits detected in the elevated plus maze and open field paradigms. CONCLUSIONS Our study highlights multiple gene drivers in neuroanatomical phenotypes, interacting with each other through complex relationships. It also provides the first evidence for the involvement of the major vault protein in the regulation of brain size and neuroanatomy, specifically in male mice.
Collapse
Affiliation(s)
- Perrine F Kretz
- Institute of Genetics and Molecular and Cellular Biology, UMR7104, University of Strasbourg, CNRS, INSERM, IGBMC, U964, 67400, Illkirch, France
| | - Christel Wagner
- Institute of Genetics and Molecular and Cellular Biology, UMR7104, University of Strasbourg, CNRS, INSERM, IGBMC, U964, 67400, Illkirch, France
| | - Anna Mikhaleva
- Center for Integrative Genomics, University of Lausanne, CH-1015, Lausanne, Switzerland
| | | | - Sylvain Hugel
- Institute of Cellular and Integrative neuroscience, CNRS, UPR321267000, Strasbourg, France
| | - Ilaria Morella
- School of Biosciences, Neuroscience and Mental Health Innovation Institute, Cardiff University, Cardiff, CF24 4HQ, UK
| | - Meghna Kannan
- Institute of Genetics and Molecular and Cellular Biology, UMR7104, University of Strasbourg, CNRS, INSERM, IGBMC, U964, 67400, Illkirch, France
| | - Marie-Christine Fischer
- Institute of Genetics and Molecular and Cellular Biology, UMR7104, University of Strasbourg, CNRS, INSERM, IGBMC, U964, 67400, Illkirch, France
| | - Maxence Milhau
- Inserm UMR1231, Université de Bourgogne, 21000, Dijon, France
| | - Ipek Yalcin
- Institute of Cellular and Integrative neuroscience, CNRS, UPR321267000, Strasbourg, France
| | - Riccardo Brambilla
- School of Biosciences, Neuroscience and Mental Health Innovation Institute, Cardiff University, Cardiff, CF24 4HQ, UK
- Dipartimento di Biologia e Biotecnologie "Lazzaro Spallanzani", Università degli Studi di Pavia, Pavia, Italy
| | - Mohammed Selloum
- Institute of Genetics and Molecular and Cellular Biology, UMR7104, University of Strasbourg, CNRS, INSERM, IGBMC, U964, 67400, Illkirch, France
- University of Strasbourg, CNRS, INSERM, CELPHEDIA, PHENOMIN, ICS, 67400, Illkirch, France
| | - Yann Herault
- Institute of Genetics and Molecular and Cellular Biology, UMR7104, University of Strasbourg, CNRS, INSERM, IGBMC, U964, 67400, Illkirch, France
- University of Strasbourg, CNRS, INSERM, CELPHEDIA, PHENOMIN, ICS, 67400, Illkirch, France
| | - Alexandre Reymond
- Center for Integrative Genomics, University of Lausanne, CH-1015, Lausanne, Switzerland
| | - Stephan C Collins
- Institute of Genetics and Molecular and Cellular Biology, UMR7104, University of Strasbourg, CNRS, INSERM, IGBMC, U964, 67400, Illkirch, France
- Current address: Université de Bourgogne, Inserm UMR1231, 21000, Dijon, France
| | - Binnaz Yalcin
- Institute of Genetics and Molecular and Cellular Biology, UMR7104, University of Strasbourg, CNRS, INSERM, IGBMC, U964, 67400, Illkirch, France.
- Current address: Université de Bourgogne, Inserm UMR1231, 21000, Dijon, France.
| |
Collapse
|
10
|
João J, Prazeres DMF. Manufacturing of non-viral protein nanocages for biotechnological and biomedical applications. Front Bioeng Biotechnol 2023; 11:1200729. [PMID: 37520292 PMCID: PMC10374429 DOI: 10.3389/fbioe.2023.1200729] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 07/05/2023] [Indexed: 08/01/2023] Open
Abstract
Protein nanocages are highly ordered nanometer scale architectures, which are typically formed by homo- or hetero-self-assembly of multiple monomers into symmetric structures of different size and shape. The intrinsic characteristics of protein nanocages make them very attractive and promising as a biological nanomaterial. These include, among others, a high surface/volume ratio, multi-functionality, ease to modify or manipulate genetically or chemically, high stability, mono-dispersity, and biocompatibility. Since the beginning of the investigation into protein nanocages, several applications were conceived in a variety of areas such as drug delivery, vaccine development, bioimaging, biomineralization, nanomaterial synthesis and biocatalysis. The ability to generate large amounts of pure and well-folded protein assemblies is one of the keys to transform nanocages into clinically valuable products and move biomedical applications forward. This calls for the development of more efficient biomanufacturing processes and for the setting up of analytical techniques adequate for the quality control and characterization of the biological function and structure of nanocages. This review concisely covers and overviews the progress made since the emergence of protein nanocages as a new, next-generation class of biologics. A brief outline of non-viral protein nanocages is followed by a presentation of their main applications in the areas of bioengineering, biotechnology, and biomedicine. Afterwards, we focus on a description of the current processes used in the manufacturing of protein nanocages with particular emphasis on the most relevant aspects of production and purification. The state-of-the-art on current characterization techniques is then described and future alternative or complementary approaches in development are also discussed. Finally, a critical analysis of the limitations and drawbacks of the current manufacturing strategies is presented, alongside with the identification of the major challenges and bottlenecks.
Collapse
Affiliation(s)
- Jorge João
- iBB–Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB–Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Duarte Miguel F. Prazeres
- iBB–Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB–Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
11
|
Kurusu R, Fujimoto Y, Morishita H, Noshiro D, Takada S, Yamano K, Tanaka H, Arai R, Kageyama S, Funakoshi T, Komatsu-Hirota S, Taka H, Kazuno S, Miura Y, Koike M, Wakai T, Waguri S, Noda NN, Komatsu M. Integrated proteomics identifies p62-dependent selective autophagy of the supramolecular vault complex. Dev Cell 2023:S1534-5807(23)00191-0. [PMID: 37192622 DOI: 10.1016/j.devcel.2023.04.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 03/13/2023] [Accepted: 04/25/2023] [Indexed: 05/18/2023]
Abstract
In addition to membranous organelles, autophagy selectively degrades biomolecular condensates, in particular p62/SQSTM1 bodies, to prevent diseases including cancer. Evidence is growing regarding the mechanisms by which autophagy degrades p62 bodies, but little is known about their constituents. Here, we established a fluorescence-activated-particle-sorting-based purification method for p62 bodies using human cell lines and determined their constituents by mass spectrometry. Combined with mass spectrometry of selective-autophagy-defective mouse tissues, we identified vault, a large supramolecular complex, as a cargo within p62 bodies. Mechanistically, major vault protein directly interacts with NBR1, a p62-interacting protein, to recruit vault into p62 bodies for efficient degradation. This process, named vault-phagy, regulates homeostatic vault levels in vivo, and its impairment may be associated with non-alcoholic-steatohepatitis-derived hepatocellular carcinoma. Our study provides an approach to identifying phase-separation-mediated selective autophagy cargoes, expanding our understanding of the role of phase separation in proteostasis.
Collapse
Affiliation(s)
- Reo Kurusu
- Department of Physiology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Yuki Fujimoto
- Department of Physiology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Hideaki Morishita
- Department of Physiology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan.
| | - Daisuke Noshiro
- Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan
| | - Shuhei Takada
- Department of Physiology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Koji Yamano
- Department of Biomolecular Pathogenesis, Medical Research Institute, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Hideaki Tanaka
- Laboratory for Protein Crystallography, Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Ritsuko Arai
- Department of Anatomy and Histology, Fukushima Medical University School of Medicine, Hikarigaoka, Fukushima 960-1295, Japan
| | - Shun Kageyama
- Department of Physiology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Tomoko Funakoshi
- Department of Physiology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Satoko Komatsu-Hirota
- Department of Physiology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Hikari Taka
- Laboratory of Proteomics and Biomolecular Science, Biomedical Research Core Facilities, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Saiko Kazuno
- Laboratory of Proteomics and Biomolecular Science, Biomedical Research Core Facilities, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Yoshiki Miura
- Laboratory of Proteomics and Biomolecular Science, Biomedical Research Core Facilities, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Masato Koike
- Department of Cell Biology and Neuroscience, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Toshifumi Wakai
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata City, Niigata 951-8510, Japan
| | - Satoshi Waguri
- Department of Anatomy and Histology, Fukushima Medical University School of Medicine, Hikarigaoka, Fukushima 960-1295, Japan
| | - Nobuo N Noda
- Institute for Genetic Medicine, Hokkaido University, Sapporo 060-0815, Japan
| | - Masaaki Komatsu
- Department of Physiology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8421, Japan.
| |
Collapse
|
12
|
Addressing Critical Issues Related to Storage and Stability of the Vault Nanoparticle Expressed and Purified from Komagataella phaffi. Int J Mol Sci 2023; 24:ijms24044214. [PMID: 36835627 PMCID: PMC9959619 DOI: 10.3390/ijms24044214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/08/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
The vault nanoparticle is a eukaryotic assembly consisting of 78 copies of the 99-kDa major vault protein. They generate two cup-shaped symmetrical halves, which in vivo enclose protein and RNA molecules. Overall, this assembly is mainly involved in pro-survival and cytoprotective functions. It also holds a remarkable biotechnological potential for drug/gene delivery, thanks to its huge internal cavity and the absence of toxicity/immunogenicity. The available purification protocols are complex, partly because they use higher eukaryotes as expression systems. Here, we report a simplified procedure that combines human vault expression in the yeast Komagataella phaffii, as described in a recent report, and a purification process we have developed. This consists of RNase pretreatment followed by size-exclusion chromatography, which is far simpler than any other reported to date. Protein identity and purity was confirmed by SDS-PAGE, Western blot and transmission electron microscopy. We also found that the protein displayed a significant propensity to aggregate. We thus investigated this phenomenon and the related structural changes by Fourier-transform spectroscopy and dynamic light scattering, which led us to determine the most suitable storage conditions. In particular, the addition of either trehalose or Tween-20 ensured the best preservation of the protein in native, soluble form.
Collapse
|
13
|
Tailored Functionalized Protein Nanocarriers for Cancer Therapy: Recent Developments and Prospects. Pharmaceutics 2023; 15:pharmaceutics15010168. [PMID: 36678796 PMCID: PMC9861211 DOI: 10.3390/pharmaceutics15010168] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/28/2022] [Accepted: 12/29/2022] [Indexed: 01/06/2023] Open
Abstract
Recently, the potential use of nanoparticles for the targeted delivery of therapeutic and diagnostic agents has garnered increased interest. Several nanoparticle drug delivery systems have been developed for cancer treatment. Typically, protein-based nanocarriers offer several advantages, including biodegradability and biocompatibility. Using genetic engineering or chemical conjugation approaches, well-known naturally occurring protein nanoparticles can be further prepared, engineered, and functionalized in their self-assembly to meet the demands of clinical production efficiency. Accordingly, promising protein nanoparticles have been developed with outstanding tumor-targeting capabilities, ultimately overcoming multidrug resistance issues, in vivo delivery barriers, and mimicking the tumor microenvironment. Bioinspired by natural nanoparticles, advanced computational techniques have been harnessed for the programmable design of highly homogenous protein nanoparticles, which could open new routes for the rational design of vaccines and drug formulations. The current review aims to present several significant advancements made in protein nanoparticle technology, and their use in cancer therapy. Additionally, tailored construction methods and therapeutic applications of engineered protein-based nanoparticles are discussed.
Collapse
|
14
|
Fernández R, Carreño A, Mendoza R, Benito A, Ferrer-Miralles N, Céspedes MV, Corchero JL. Escherichia coli as a New Platform for the Fast Production of Vault-like Nanoparticles: An Optimized Protocol. Int J Mol Sci 2022; 23:ijms232415543. [PMID: 36555185 PMCID: PMC9778704 DOI: 10.3390/ijms232415543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/30/2022] [Accepted: 12/03/2022] [Indexed: 12/13/2022] Open
Abstract
Vaults are protein nanoparticles that are found in almost all eukaryotic cells but are absent in prokaryotic ones. Due to their properties (nanometric size, biodegradability, biocompatibility, and lack of immunogenicity), vaults show enormous potential as a bio-inspired, self-assembled drug-delivery system (DDS). Vault architecture is directed by self-assembly of the "major vault protein" (MVP), the main component of this nanoparticle. Recombinant expression (in different eukaryotic systems) of the MVP resulted in the formation of nanoparticles that were indistinguishable from native vaults. Nowadays, recombinant vaults for different applications are routinely produced in insect cells and purified by successive ultracentrifugations, which are both tedious and time-consuming strategies. To offer cost-efficient and faster protocols for nanoparticle production, we propose the production of vault-like nanoparticles in Escherichia coli cells, which are still one of the most widely used prokaryotic cell factories for recombinant protein production. The strategy proposed allowed for the spontaneous encapsulation of the engineered cargo protein within the self-assembled vault-like nanoparticles by simply mixing the clarified lysates of the producing cells. Combined with well-established affinity chromatography purification methods, our approach contains faster, cost-efficient procedures for biofabrication in a well-known microbial cell factory and the purification of "ready-to-use" loaded protein nanoparticles, thereby opening the way to faster and easier engineering and production of vault-based DDSs.
Collapse
Affiliation(s)
- Roger Fernández
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Aida Carreño
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - Rosa Mendoza
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Bellaterra, 08193 Barcelona, Spain
| | - Antoni Benito
- Laboratori d’Enginyeria de Proteïnes, Departament de Biologia, Universitat de Girona, 17003 Girona, Spain
- Institut d’Investigació Biomèdica de Girona Josep Trueta, (IdIBGi), 17190 Salt, Spain
| | - Neus Ferrer-Miralles
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Bellaterra, 08193 Barcelona, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
| | - María Virtudes Céspedes
- Grup d’Oncologia Ginecològica i Peritoneal, Institut d’Investigacions Biomédiques Sant Pau, Hospital de Santa Creu i Sant Pau, 08041 Barcelona, Spain
- Correspondence: (M.V.C.); (J.L.C.); Tel.: +34-93-2919000 (ext. 1427) (M.V.C.); +34-93-5812148 (J.L.C.)
| | - José Luis Corchero
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina, Instituto de Salud Carlos III, Bellaterra, 08193 Barcelona, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra, 08193 Barcelona, Spain
- Correspondence: (M.V.C.); (J.L.C.); Tel.: +34-93-2919000 (ext. 1427) (M.V.C.); +34-93-5812148 (J.L.C.)
| |
Collapse
|
15
|
Liu T, Li L, Cheng C, He B, Jiang T. Emerging prospects of protein/peptide-based nanoassemblies for drug delivery and vaccine development. NANO RESEARCH 2022; 15:7267-7285. [PMID: 35692441 PMCID: PMC9166156 DOI: 10.1007/s12274-022-4385-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 05/09/2023]
Abstract
Proteins have been widely used in the biomedical field because of their well-defined architecture, accurate molecular weight, excellent biocompatibility and biodegradability, and easy-to-functionalization. Inspired by the wisdom of nature, increasing proteins/peptides that possess self-assembling capabilities have been explored and designed to generate nanoassemblies with unique structure and function, including spatially organized conformation, passive and active targeting, stimuli-responsiveness, and high stability. These characteristics make protein/peptide-based nanoassembly an ideal platform for drug delivery and vaccine development. In this review, we focus on recent advances in subsistent protein/peptide-based nanoassemblies, including protein nanocages, virus-like particles, self-assemblable natural proteins, and self-assemblable artificial peptides. The origin and characteristics of various protein/peptide-based assemblies and their applications in drug delivery and vaccine development are summarized. In the end, the prospects and challenges are discussed for the further development of protein/peptide-based nanoassemblies.
Collapse
Affiliation(s)
- Taiyu Liu
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816 China
| | - Lu Li
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816 China
| | - Cheng Cheng
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816 China
| | - Bingfang He
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816 China
| | - Tianyue Jiang
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, 211816 China
| |
Collapse
|
16
|
Gao Y, Wang M, Shah K, Singh Kalra S, Rome LH, Mahendra S. Decolorization and detoxification of synthetic dye compounds by laccase immobilized in vault nanoparticles. BIORESOURCE TECHNOLOGY 2022; 351:127040. [PMID: 35318145 DOI: 10.1016/j.biortech.2022.127040] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 06/14/2023]
Abstract
This study presents an eco-friendly and efficient technology, using immobilized enzymes, vault-encapsulated laccases (vlaccase), for decolorization and detoxification of dyes. Vault encapsulation remarkably improved the performance of laccase at industrially relevant conditions, including neutral to alkaline pH and relatively high temperatures. Two representative anthraquinone and azo dyes, Reactive Blue 19 and Acid Orange 7, respectively, were rapidly decolorized (72% and 80%) by vlaccase treatment while natural laccase (nlaccase) achieved 40% and 32% decolorization. The toxicity of treated and untreated dyes was tested on model bacterial, algal, and insect cells. The inhibitory effects of dyes towards selected bacteria were reduced in vlaccase-treated samples. The chlorophyll synthesis in algae was less inhibited by dyes after vlaccase treatment. Furthermore, the toxicity of dye degradation products to insect cells was significantly mitigated in the vlaccase group. Collectively, these results indicate that vlaccase is a stable and strong enzymatic system for removing dyes from waters.
Collapse
Affiliation(s)
- Yifan Gao
- Department of Civil and Environmental Engineering, University of California, Los Angeles, CA 90095, United States
| | - Meng Wang
- Department of Civil and Environmental Engineering, University of California, Los Angeles, CA 90095, United States; Department of Civil and Environmental Engineering, University of Pittsburgh, Pittsburgh, PA 15260, United States
| | - Kshitija Shah
- Department of Civil and Environmental Engineering, University of California, Los Angeles, CA 90095, United States
| | - Shashank Singh Kalra
- Department of Civil and Environmental Engineering, University of California, Los Angeles, CA 90095, United States
| | - Leonard H Rome
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, United States; California NanoSystems Institute, University of California, Los Angeles, CA 90095, United States
| | - Shaily Mahendra
- Department of Civil and Environmental Engineering, University of California, Los Angeles, CA 90095, United States; California NanoSystems Institute, University of California, Los Angeles, CA 90095, United States.
| |
Collapse
|
17
|
Gao Y, Shah K, Kwok I, Wang M, Rome LH, Mahendra S. Immobilized fungal enzymes: Innovations and potential applications in biodegradation and biosynthesis. Biotechnol Adv 2022; 57:107936. [PMID: 35276253 DOI: 10.1016/j.biotechadv.2022.107936] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/28/2022] [Accepted: 03/03/2022] [Indexed: 01/10/2023]
Abstract
Microbial enzymes catalyze various reactions inside and outside living cells. Among the widely studied enzymes, fungal enzymes have been used for some of the most diverse purposes, especially in bioremediation, biosynthesis, and many nature-inspired commercial applications. To improve their stability and catalytic ability, fungal enzymes are often immobilized on assorted materials, conventional as well as nanoscale. Recent advances in fungal enzyme immobilization provide effective and sustainable approaches to achieve improved environmental and commercial outcomes. This review aims to provide a comprehensive overview of commonly studied fungal enzymes and immobilization technologies. It also summarizes recent advances involving immobilized fungal enzymes for the degradation or assembly of compounds used in the manufacture of products, such as detergents, food additives, and fossil fuel alternatives. Furthermore, challenges and future directions are highlighted to offer new perspectives on improving existing technologies and addressing unexplored fields of applications.
Collapse
Affiliation(s)
- Yifan Gao
- Department of Civil and Environmental Engineering, University of California, Los Angeles, CA 90095, United States
| | - Kshitjia Shah
- Department of Civil and Environmental Engineering, University of California, Los Angeles, CA 90095, United States
| | - Ivy Kwok
- Department of Civil and Environmental Engineering, University of California, Los Angeles, CA 90095, United States
| | - Meng Wang
- Department of Civil and Environmental Engineering, University of Pittsburgh, Pittsburgh, PA 15260, United States
| | - Leonard H Rome
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, United States; California NanoSystems Institute, University of California, Los Angeles, CA 90095, United States
| | - Shaily Mahendra
- Department of Civil and Environmental Engineering, University of California, Los Angeles, CA 90095, United States; California NanoSystems Institute, University of California, Los Angeles, CA 90095, United States.
| |
Collapse
|
18
|
Martín F, Carreño A, Mendoza R, Caruana P, Rodríguez F, Bravo M, Benito A, Ferrer-Miralles N, Céspedes MV, Corchero JL. All-in-one biofabrication and loading of recombinant vaults in human cells. Biofabrication 2022; 14. [PMID: 35203066 DOI: 10.1088/1758-5090/ac584d] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 02/24/2022] [Indexed: 11/12/2022]
Abstract
One of the most promising approaches in the drug delivery field is the use of naturally occurring self-assembling protein nanoparticles, such as virus-like particles, bacterial microcompartments or vault ribonucleoprotein particles as drug delivery systems (DDS). Among them, eukaryotic vaults show a promising future due to their structural features, in vitro stability and non-immunogenicity. Recombinant vaults are routinely produced in insect cells and purified through several ultracentrifugations, both tedious and time-consuming processes. As an alternative, this work proposes a new approach and protocols for the production of recombinant vaults in human cells by transient gene expression of a His-tagged version of the Major Vault Protein (MVP-H6), the development of new affinity-based purification processes for such recombinant vaults, and the all-in-one biofabrication and encapsulation of a cargo recombinant protein within such vaults by their co-expression in human cells. Protocols proposed here allow the easy and straightforward biofabrication and purification of engineered vaults loaded with virtually any INT-tagged cargo protein, in very short times, paving the way to faster and easier engineering and production of better and more efficient DDS.
Collapse
Affiliation(s)
- Fernando Martín
- Universitat Autonoma de Barcelona, Institut de Biotecnologia i de Biomedicina, Campus Universitari Bellaterra, Bellaterra, Bellaterra, Catalunya, 08193, SPAIN
| | - Aida Carreño
- Universitat Autonoma de Barcelona, Institut de Biotecnologia i de Biomedicina, Campus Universitari Bellaterra, Bellaterra, Bellaterra, Catalunya, 08193, SPAIN
| | - Rosa Mendoza
- CIBER-BBN, Institut de Biotecnologia i de Biomedicina, Campus Universitari Bellaterra, Bellaterra, Bellaterra, 08193, SPAIN
| | - Pablo Caruana
- Hospital de la Santa Creu i Sant Pau, Sant Pau Biomedical Research Institute (IIB Sant Pau) Carrer Sant Quintí, 77-79, Barcelona, Catalunya, 08041, SPAIN
| | - Francisco Rodríguez
- Hospital de la Santa Creu i Sant Pau, Sant Pau Biomedical Research Institute (IIB Sant Pau) Carrer Sant Quintí, 77-79 08041. Barcelona, Spain, Barcelona, Catalunya, 08041, SPAIN
| | - Marlon Bravo
- Universitat de Girona, Laboratori Enginyeria Proteines, Dept biologia, Universitat de Girona, Girona, Catalunya, 17003, SPAIN
| | - Antoni Benito
- Universitat de Girona, Facultat de Ciències, Universitat de Girona, Campus de Montilivi, Carrer Maria Aurèlia Capmany, 40,, Girona, Catalunya, 17003, SPAIN
| | - Neus Ferrer-Miralles
- Universitat Autonoma de Barcelona, Institut de Biotecnologia i de Biomedicina, Campus Universitari Bellaterra, Bellaterra, Bellaterra, Catalunya, 08193, SPAIN
| | - Mª Virtudes Céspedes
- Hospital de la Santa Creu i Sant Pau, Sant Pau Biomedical Research Institute (IIB Sant Pau) Carrer Sant Quintí, 77-79, Barcelona, Catalunya, 08041, SPAIN
| | - Jose Luis Corchero
- CIBER-BBN, Institut de Biotecnologia i de Biomedicina, Campus Universitari Bellaterra, Bellaterra, 08193, SPAIN
| |
Collapse
|
19
|
Guerra P, González-Alamos M, Llauró A, Casañas A, Querol-Audí J, de Pablo PJ, Verdaguer N. Symmetry disruption commits vault particles to disassembly. SCIENCE ADVANCES 2022; 8:eabj7795. [PMID: 35138889 PMCID: PMC8827651 DOI: 10.1126/sciadv.abj7795] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Vaults are ubiquitous ribonucleoprotein particles involved in a diversity of cellular processes, with promising applications as nanodevices for delivery of multiple cargos. The vault shell is assembled by the symmetrical association of multiple copies of the major vault protein that, initially, generates half vaults. The pairwise, anti-parallel association of two half vaults produces whole vaults. Here, using a combination of vault recombinant reconstitution and structural techniques, we characterized the molecular determinants for the vault opening process. This process commences with a relaxation of the vault waist, causing the expansion of the inner cavity. Then, local disengagement of amino-terminal domains at the vault midsection seeds a conformational change that leads to the aperture, facilitating access to the inner cavity where cargo is hosted. These results inform a hitherto uncharacterized step of the vault cycle and will aid current engineering efforts leveraging vault for tailored cargo delivery.
Collapse
Affiliation(s)
- Pablo Guerra
- Structural Biology Department, Instituto de Biología Molecular de Barcelona (IBMB-CSIC), Parc Científic de Barcelona, Baldiri i Reixac 15, E-08028 Barcelona, Spain
| | - María González-Alamos
- Structural Biology Department, Instituto de Biología Molecular de Barcelona (IBMB-CSIC), Parc Científic de Barcelona, Baldiri i Reixac 15, E-08028 Barcelona, Spain
| | - Aida Llauró
- Department of Condensed Matter Physics, Autonomous University of Madrid, Madrid 28049, Spain
| | - Arnau Casañas
- Structural Biology Department, Instituto de Biología Molecular de Barcelona (IBMB-CSIC), Parc Científic de Barcelona, Baldiri i Reixac 15, E-08028 Barcelona, Spain
| | - Jordi Querol-Audí
- Structural Biology Department, Instituto de Biología Molecular de Barcelona (IBMB-CSIC), Parc Científic de Barcelona, Baldiri i Reixac 15, E-08028 Barcelona, Spain
| | - Pedro J. de Pablo
- Department of Condensed Matter Physics, Autonomous University of Madrid, Madrid 28049, Spain
| | - Núria Verdaguer
- Structural Biology Department, Instituto de Biología Molecular de Barcelona (IBMB-CSIC), Parc Científic de Barcelona, Baldiri i Reixac 15, E-08028 Barcelona, Spain
- Corresponding author.
| |
Collapse
|
20
|
Major Vault Protein Inhibits Porcine Reproductive and Respiratory Syndrome Virus Infection in CRL2843 CD163 Cell Lines and Primary Porcine Alveolar Macrophages. Viruses 2021; 13:v13112267. [PMID: 34835073 PMCID: PMC8618244 DOI: 10.3390/v13112267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/02/2021] [Accepted: 11/09/2021] [Indexed: 11/16/2022] Open
Abstract
Porcine reproductive and respiratory syndrome (PRRS), a significant viral infectious disease that commonly occurs among farmed pigs, leads to considerable economic losses to the swine industry worldwide. Major vault protein (MVP) is a host factor that induces type Ⅰ interferon (IFN) production. In this study, we evaluated the effect of MVP on PRRSV infection in CRL2843CD163 cell lines and porcine alveolar macrophages (PAMs). Our results showed that MVP expression was downregulated by PRRSV infection. Adenoviral overexpression of MVP inhibited PRRSV replication, whereas the siRNA knockdown of MVP promoted PRRSV replication. In addition, MVP knockdown has an adverse effect on the inhibitive role of MVP overexpression on PRRSV replication. Moreover, MVP could induce the expression of type Ⅰ IFNs and IFN-stimulated gene 15 (ISG15) in PRRSV-infected PAMs. Based on these results, MVP may be a potential molecular target of drugs for the effective prevention and treatment of PRRSV infection.
Collapse
|
21
|
Goel D, Sinha S. Naturally occurring protein nano compartments: basic structure, function, and genetic engineering. NANO EXPRESS 2021. [DOI: 10.1088/2632-959x/ac2c93] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
22
|
Immunoediting role for major vault protein in apoptotic signaling induced by bacterial N-acyl homoserine lactones. Proc Natl Acad Sci U S A 2021; 118:2012529118. [PMID: 33723037 PMCID: PMC8000436 DOI: 10.1073/pnas.2012529118] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The major vault protein (MVP) mediates diverse cellular responses, including cancer cell resistance to chemotherapy and protection against inflammatory responses to Pseudomonas aeruginosa Here, we report the use of photoactive probes to identify MVP as a target of the N-(3-oxo-dodecanoyl) homoserine lactone (C12), a quorum sensing signal of certain proteobacteria including P. aeruginosa. A treatment of normal and cancer cells with C12 or other N-acyl homoserine lactones (AHLs) results in rapid translocation of MVP into lipid raft (LR) membrane fractions. Like AHLs, inflammatory stimuli also induce LR-localization of MVP, but the C12 stimulation reprograms (functionalizes) bioactivity of the plasma membrane by recruiting death receptors, their apoptotic adaptors, and caspase-8 into LR. These functionalized membranes control AHL-induced signaling processes, in that MVP adjusts the protein kinase p38 pathway to attenuate programmed cell death. Since MVP is the structural core of large particles termed vaults, our findings suggest a mechanism in which MVP vaults act as sentinels that fine-tune inflammation-activated processes such as apoptotic signaling mediated by immunosurveillance cytokines including tumor necrosis factor-related apoptosis inducing ligand (TRAIL).
Collapse
|
23
|
Frascotti G, Galbiati E, Mazzucchelli M, Pozzi M, Salvioni L, Vertemara J, Tortora P. The Vault Nanoparticle: A Gigantic Ribonucleoprotein Assembly Involved in Diverse Physiological and Pathological Phenomena and an Ideal Nanovector for Drug Delivery and Therapy. Cancers (Basel) 2021; 13:cancers13040707. [PMID: 33572350 PMCID: PMC7916137 DOI: 10.3390/cancers13040707] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/03/2021] [Accepted: 02/03/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary In recent decades, a molecular complex referred to as vault nanoparticle has attracted much attention by the scientific community, due to its unique properties. At the molecular scale, it is a huge assembly consisting of 78 97-kDa polypeptide chains enclosing an internal cavity, wherein enzymes involved in DNA integrity maintenance and some small noncoding RNAs are accommodated. Basically, two reasons justify this interest. On the one hand, this complex represents an ideal tool for the targeted delivery of drugs, provided it is suitably engineered, either chemically or genetically; on the other hand, it has been shown to be involved in several cellular pathways and mechanisms that most often result in multidrug resistance. It is therefore expected that a better understanding of the physiological roles of this ribonucleoproteic complex may help develop new therapeutic strategies capable of coping with cancer progression. Here, we provide a comprehensive review of the current knowledge. Abstract The vault nanoparticle is a eukaryotic ribonucleoprotein complex consisting of 78 individual 97 kDa-“major vault protein” (MVP) molecules that form two symmetrical, cup-shaped, hollow halves. It has a huge size (72.5 × 41 × 41 nm) and an internal cavity, wherein the vault poly(ADP-ribose) polymerase (vPARP), telomerase-associated protein-1 (TEP1), and some small untranslated RNAs are accommodated. Plenty of literature reports on the biological role(s) of this nanocomplex, as well as its involvement in diseases, mostly oncological ones. Nevertheless, much has still to be understood as to how vault participates in normal and pathological mechanisms. In this comprehensive review, current understanding of its biological roles is discussed. By different mechanisms, vault’s individual components are involved in major cellular phenomena, which result in protection against cellular stresses, such as DNA-damaging agents, irradiation, hypoxia, hyperosmotic, and oxidative conditions. These diverse cellular functions are accomplished by different mechanisms, mainly gene expression reprogramming, activation of proliferative/prosurvival signaling pathways, export from the nucleus of DNA-damaging drugs, and import of specific proteins. The cellular functions of this nanocomplex may also result in the onset of pathological conditions, mainly (but not exclusively) tumor proliferation and multidrug resistance. The current understanding of its biological roles in physiological and pathological processes should also provide new hints to extend the scope of its exploitation as a nanocarrier for drug delivery.
Collapse
|
24
|
Hahne JC, Lampis A, Valeri N. Vault RNAs: hidden gems in RNA and protein regulation. Cell Mol Life Sci 2021; 78:1487-1499. [PMID: 33063126 PMCID: PMC7904556 DOI: 10.1007/s00018-020-03675-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 09/27/2020] [Accepted: 10/05/2020] [Indexed: 12/11/2022]
Abstract
Non-coding RNAs are important regulators of differentiation during embryogenesis as well as key players in the fine-tuning of transcription and furthermore, they control the post-transcriptional regulation of mRNAs under physiological conditions. Deregulated expression of non-coding RNAs is often identified as one major contribution in a number of pathological conditions. Non-coding RNAs are a heterogenous group of RNAs and they represent the majority of nuclear transcripts in eukaryotes. An evolutionary highly conserved sub-group of non-coding RNAs is represented by vault RNAs, named since firstly discovered as component of the largest known ribonucleoprotein complexes called "vault". Although they have been initially described 30 years ago, vault RNAs are largely unknown and their molecular role is still under investigation. In this review we will summarize the known functions of vault RNAs and their involvement in cellular mechanisms.
Collapse
Affiliation(s)
- Jens Claus Hahne
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK.
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK.
| | - Andrea Lampis
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Nicola Valeri
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
- Department of Medicine, The Royal Marsden NHS Foundation Trust, London, UK
| |
Collapse
|
25
|
Zhang X, Yang Y, Bu X, Wei Y, Lou X. The major vault protein is dispensable for zebrafish organ regeneration. Heliyon 2020; 6:e05422. [PMID: 33195847 PMCID: PMC7644919 DOI: 10.1016/j.heliyon.2020.e05422] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 10/08/2020] [Accepted: 10/29/2020] [Indexed: 01/11/2023] Open
Abstract
As the main constituent of the largest cellular ribonucleoprotein complex, the evolutionary highly conserved major vault protein (MVP) has been proposed play vital roles in the regeneration of multiple organs. In current study, we use a mvp knockout zebrafish line recently generated to characterize the function of MVP during organ regeneration. We found the regenerative capacity of heart, spinal cord and fin is preserved in mvp knockout zebrafish. Further experiments demonstrated in injured mvp knockout zebrafish, the cell death is enhanced while the transcriptome landscape is largely unchanged. These data showed MVP acts as an anti-apoptotic factor at early phase of injury response while plays a dispensable role in the regenerative programs in zebrafish.
Collapse
Affiliation(s)
- Xue Zhang
- Medical School, Nanjing University, China
| | - Yuxi Yang
- Medical School, Nanjing University, China
| | - Xiaoxue Bu
- Medical School, Nanjing University, China
| | | | - Xin Lou
- Medical School, Nanjing University, China
| |
Collapse
|
26
|
Bioengineered recombinant vault nanoparticles coupled with NY-ESO-1 glioma-associated antigens induce maturation of native dendritic cells. J Neurooncol 2020; 148:1-7. [DOI: 10.1007/s11060-020-03472-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 03/23/2020] [Indexed: 11/27/2022]
|
27
|
Voth BL, Pelargos PE, Barnette NE, Bhatt NS, Chen CHJ, Lagman C, Chung LK, Nguyen T, Sheppard JP, Romiyo P, Mareninov S, Kickhoefer VA, Yong WH, Rome LH, Yang I. Intratumor injection of CCL21-coupled vault nanoparticles is associated with reduction in tumor volume in an in vivo model of glioma. J Neurooncol 2020; 147:599-605. [PMID: 32274629 DOI: 10.1007/s11060-020-03479-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 03/26/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE Glioblastoma (GBM) is the most common and malignant primary adult brain tumor. Current care includes surgical resection, radiation, and chemotherapy. Recent clinical trials for GBM have demonstrated extended survival using interventions such as tumor vaccines or tumor-treating fields. However, prognosis generally remains poor, with expected survival of 20 months after randomization. Chemokine-based immunotherapy utilizing CCL21 locally recruits lymphocytes and dendritic cells to enhance host antitumor response. Here, we report a preliminary study utilizing CPZ-vault nanoparticles as a vehicle to package, protect, and steadily deliver therapy to optimize CCL21 therapy in a murine flank model of GBM. METHODS GL261 cells were subcutaneously injected into the left flank of eight-week-old female C57BL/6 mice. Mice were treated with intratumoral injections of either: (1) CCL21-packaged vault nanoparticles (CPZ-CCL21), (2) free recombinant CCL21 chemokine empty vault nanoparticles, (3) empty vault nanoparticles, or 4) PBS. RESULTS The results of this study showed that CCL21-packaged vault nanoparticle injections can decrease the tumor volume in vivo. Additionally, this study showed mice injected with CCL21-packaged vault nanoparticle had the smallest average tumor volume and remained the only treatment group with a negative percent change in tumor volume. CONCLUSIONS This preliminary study establishes vault nanoparticles as a feasible vehicle to increase drug delivery and immune response in a flank murine model of GBM. Future animal studies involving an intracranial orthotopic tumor model are required to fully evaluate the potential for CCL21-packaged vault nanoparticles as a strategy to bypass the blood brain barrier, enhance intracranial immune activity, and improve intracranial tumor control and survival.
Collapse
Affiliation(s)
- Brittany L Voth
- Departments of Neurosurgery, University of California, Los Angeles, CA, USA
| | | | - Natalie E Barnette
- Departments of Neurosurgery, University of California, Los Angeles, CA, USA
| | - Nikhilesh S Bhatt
- Departments of Neurosurgery, University of California, Los Angeles, CA, USA
| | | | - Carlito Lagman
- Departments of Neurosurgery, University of California, Los Angeles, CA, USA
| | - Lawrance K Chung
- Departments of Neurosurgery, University of California, Los Angeles, CA, USA
| | - Thien Nguyen
- Departments of Neurosurgery, University of California, Los Angeles, CA, USA
| | - John P Sheppard
- Departments of Neurosurgery, University of California, Los Angeles, CA, USA
| | - Prasanth Romiyo
- Departments of Neurosurgery, University of California, Los Angeles, CA, USA
| | - Sergey Mareninov
- Departments of Biological Chemistry, Jonsson Comprehensive Cancer Center, University of California, 300 Stein Plaza, Suite 562, 5th Floor Wasserman Building, Los Angeles, CA, 90095-6901, USA
| | - Valerie A Kickhoefer
- Departments of Biological Chemistry, Jonsson Comprehensive Cancer Center, University of California, 300 Stein Plaza, Suite 562, 5th Floor Wasserman Building, Los Angeles, CA, 90095-6901, USA
| | - William H Yong
- Departments of Pathology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Leonard H Rome
- Departments of Neurosurgery, University of California, Los Angeles, CA, USA
| | - Isaac Yang
- Departments of Neurosurgery, University of California, Los Angeles, CA, USA. .,Departments of Radiation Oncology, University of California, Los Angeles, CA, USA. .,Departments of Biological Chemistry, Jonsson Comprehensive Cancer Center, University of California, 300 Stein Plaza, Suite 562, 5th Floor Wasserman Building, Los Angeles, CA, 90095-6901, USA.
| |
Collapse
|
28
|
Abstract
RNA-binding proteins typically change the fate of RNA, such as stability, translation or processing. Conversely, we recently uncovered that the small non-coding vault RNA 1-1 (vtRNA1-1) directly binds to the autophagic receptor p62/SQSTM1 and changes the protein's function. We refer to this process as 'riboregulation'. Here, we discuss this newly uncovered vault RNA function against the background of three decades of vault RNA research. We highlight the vtRNA1-1-p62 interaction as an example of riboregulation of a key cellular process.
Collapse
Affiliation(s)
- Magdalena Büscher
- European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany.,Collaboration for joint PhD degree between EMBL and Heidelberg University, Faculty of Biosciences, Heidelberg, Germany
| | - Rastislav Horos
- European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Matthias W Hentze
- European Molecular Biology Laboratory, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| |
Collapse
|
29
|
Fulcher JA, Tamshen K, Wollenberg AL, Kickhoefer VA, Mrazek J, Elliott J, Ibarrondo FJ, Anton PA, Rome LH, Maynard HD, Deming T, Yang OO. Human Vault Nanoparticle Targeted Delivery of Antiretroviral Drugs to Inhibit Human Immunodeficiency Virus Type 1 Infection. Bioconjug Chem 2019; 30:2216-2227. [PMID: 31265254 DOI: 10.1021/acs.bioconjchem.9b00451] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
"Vaults" are ubiquitously expressed endogenous ribonucleoprotein nanoparticles with potential utility for targeted drug delivery. Here, we show that recombinant human vault nanoparticles are readily engulfed by certain key human peripheral blood mononuclear cells (PBMC), predominately dendritic cells, monocytes/macrophages, and activated T cells. As these cell types are the primary targets for human immunodeficiency virus type 1 (HIV-1) infection, we examined the utility of recombinant human vaults for targeted delivery of antiretroviral drugs. We chemically modified three different antiretroviral drugs, zidovudine, tenofovir, and elvitegravir, for direct conjugation to vaults. Tested in infection assays, drug-conjugated vaults inhibited HIV-1 infection of PBMC with equivalent activity to free drugs, indicating vault delivery and drug release in the cytoplasm of HIV-1-susceptible cells. The ability to deliver functional drugs via vault nanoparticle conjugates suggests their potential utility for targeted drug delivery against HIV-1.
Collapse
Affiliation(s)
- Jennifer A Fulcher
- Division of Infectious Diseases, Department of Medicine , David Geffen School of Medicine at UCLA , Los Angeles , California , United States
| | - Kyle Tamshen
- Department of Chemistry and Biochemistry , University of California , Los Angeles , California , United States
| | - Alexander L Wollenberg
- Department of Chemistry and Biochemistry , University of California , Los Angeles , California , United States
| | - Valerie A Kickhoefer
- Department of Biological Chemistry , David Geffen School of Medicine at UCLA , Los Angeles , California , United States
| | - Jan Mrazek
- Division of Infectious Diseases, Department of Medicine , David Geffen School of Medicine at UCLA , Los Angeles , California , United States
| | - Julie Elliott
- Vatche and Tamar Manoukian Division of Digestive Diseases , David Geffen School of Medicine at UCLA , Los Angeles , California , United States
| | - F Javier Ibarrondo
- Division of Infectious Diseases, Department of Medicine , David Geffen School of Medicine at UCLA , Los Angeles , California , United States
| | - Peter A Anton
- Vatche and Tamar Manoukian Division of Digestive Diseases , David Geffen School of Medicine at UCLA , Los Angeles , California , United States.,AIDS Healthcare Foundation , Los Angeles , California , United States
| | - Leonard H Rome
- Department of Biological Chemistry , David Geffen School of Medicine at UCLA , Los Angeles , California , United States.,California NanoSystems Institute , University of California , Los Angeles , California , United States
| | - Heather D Maynard
- Department of Chemistry and Biochemistry , University of California , Los Angeles , California , United States.,California NanoSystems Institute , University of California , Los Angeles , California , United States.,Department of Bioengineering , University of California , Los Angeles , California , United States
| | - Timothy Deming
- Department of Chemistry and Biochemistry , University of California , Los Angeles , California , United States.,California NanoSystems Institute , University of California , Los Angeles , California , United States.,Department of Bioengineering , University of California , Los Angeles , California , United States
| | - Otto O Yang
- Division of Infectious Diseases, Department of Medicine , David Geffen School of Medicine at UCLA , Los Angeles , California , United States.,AIDS Healthcare Foundation , Los Angeles , California , United States
| |
Collapse
|
30
|
Muñoz-Juan A, Carreño A, Mendoza R, Corchero JL. Latest Advances in the Development of Eukaryotic Vaults as Targeted Drug Delivery Systems. Pharmaceutics 2019; 11:E300. [PMID: 31261673 PMCID: PMC6680493 DOI: 10.3390/pharmaceutics11070300] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 06/21/2019] [Accepted: 06/26/2019] [Indexed: 12/04/2022] Open
Abstract
The use of smart drug delivery systems (DDSs) is one of the most promising approaches to overcome some of the drawbacks of drug-based therapies, such as improper biodistribution and lack of specific targeting. Some of the most attractive candidates as DDSs are naturally occurring, self-assembling protein nanoparticles, such as viruses, virus-like particles, ferritin cages, bacterial microcompartments, or eukaryotic vaults. Vaults are large ribonucleoprotein nanoparticles present in almost all eukaryotic cells. Expression in different cell factories of recombinant versions of the "major vault protein" (MVP) results in the production of recombinant vaults indistinguishable from native counterparts. Such recombinant vaults can encapsulate virtually any cargo protein, and they can be specifically targeted by engineering the C-terminus of MVP monomer. These properties, together with nanometric size, a lumen large enough to accommodate cargo molecules, biodegradability, biocompatibility and no immunogenicity, has raised the interest in vaults as smart DDSs. In this work we provide an overview of eukaryotic vaults as a new, self-assembling protein-based DDS, focusing in the latest advances in the production and purification of this platform, its application in nanomedicine, and the current preclinical and clinical assays going on based on this nanovehicle.
Collapse
Affiliation(s)
- Amanda Muñoz-Juan
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Aida Carreño
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Rosa Mendoza
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
- Networking Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
| | - José L Corchero
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.
- Networking Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain.
- Department of Genetics and Microbiology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain.
| |
Collapse
|
31
|
Wang M, Chen Y, Kickhoefer VA, Rome LH, Allard P, Mahendra S. A Vault-Encapsulated Enzyme Approach for Efficient Degradation and Detoxification of Bisphenol A and Its Analogues. ACS SUSTAINABLE CHEMISTRY & ENGINEERING 2019; 7:5808-5817. [PMID: 36419408 PMCID: PMC9681160 DOI: 10.1021/acssuschemeng.8b05432] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
We report an effective and environmentally sustainable water treatment approach using enzymes encapsulated in biogenic vault nanoparticles. Manganese peroxidase (MnP), whose stability was remarkably extended by encapsulating into vaults, rapidly catalyzed the biotransformation of endocrine-disrupting compounds, including bisphenol A (BPA), bisphenol F (BPF), and bisphenol AP (BPAP). The vault-encapsulated MnP (vMnP) treatment removed 80-95% of each of the tested bisphenols (BPs) at lower enzyme dosage, while free native MnP (nMnP) only resulted in a 19-36% removal, over a 24-h period. Treatment by vMnP and nMnP resulted in considerable disparities in product species and abundance, which were consistent with the observed changes in the estrogenic activities of BPs. To test if vMnP-catalyzed transformations generated toxic intermediates, we assessed biological hallmarks of BP toxicity, namely, the ability to disrupt reproductive processes. The toxicity of vMnP-treated samples, as measured in the model organism, Caenorhabditis elegans, was dramatically reduced for all three BPs, including the reproductive indicators of BPA exposure such as reduced fertility and increased germ cell death. Collectively, our results indicate that the vMnP system represents an efficient and safe approach for the removal of BPs and promise the development of vault-encapsulated customized enzymes for treating other targeted organic compounds in contaminated waters.
Collapse
Affiliation(s)
- Meng Wang
- Department of Civil and Environmental Engineering, University of California, Los Angeles, 420 Westwood Plaza, Los Angeles, California 90095, United States
| | - Yichang Chen
- Molecular Toxicology Interdepartmental Program, University of California, Los Angeles, 650 Charles E Young Drive South, Los Angeles, California 90095, United States
| | - Valerie A. Kickhoefer
- Department of Biological Chemistry, University of California, Los Angeles, 615 Charles E Young Drive South, Los Angeles, California 90095, United States
- Vault Nano Inc., 570 Westwood Plaza, Los Angeles, California 90095, United States
| | - Leonard H. Rome
- Department of Biological Chemistry, University of California, Los Angeles, 615 Charles E Young Drive South, Los Angeles, California 90095, United States
- California Nanosystems Institute, University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095, United States
| | - Patrick Allard
- Molecular Toxicology Interdepartmental Program, University of California, Los Angeles, 650 Charles E Young Drive South, Los Angeles, California 90095, United States
- Institute for Society & Genetics, University of California, Los Angeles, 621 Charles E Young Drive South, Los Angeles, California 90095, United States
| | - Shaily Mahendra
- Department of Civil and Environmental Engineering, University of California, Los Angeles, 420 Westwood Plaza, Los Angeles, California 90095, United States
- Molecular Toxicology Interdepartmental Program, University of California, Los Angeles, 650 Charles E Young Drive South, Los Angeles, California 90095, United States
- California Nanosystems Institute, University of California, Los Angeles, 570 Westwood Plaza, Los Angeles, California 90095, United States
| |
Collapse
|
32
|
Synthesis and assembly of human vault particles in yeast. Biotechnol Bioeng 2018; 115:2941-2950. [DOI: 10.1002/bit.26825] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 08/04/2018] [Accepted: 08/30/2018] [Indexed: 01/04/2023]
|
33
|
Diaz D, Care A, Sunna A. Bioengineering Strategies for Protein-Based Nanoparticles. Genes (Basel) 2018; 9:E370. [PMID: 30041491 PMCID: PMC6071185 DOI: 10.3390/genes9070370] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 07/16/2018] [Accepted: 07/17/2018] [Indexed: 12/16/2022] Open
Abstract
In recent years, the practical application of protein-based nanoparticles (PNPs) has expanded rapidly into areas like drug delivery, vaccine development, and biocatalysis. PNPs possess unique features that make them attractive as potential platforms for a variety of nanobiotechnological applications. They self-assemble from multiple protein subunits into hollow monodisperse structures; they are highly stable, biocompatible, and biodegradable; and their external components and encapsulation properties can be readily manipulated by chemical or genetic strategies. Moreover, their complex and perfect symmetry have motivated researchers to mimic their properties in order to create de novo protein assemblies. This review focuses on recent advances in the bioengineering and bioconjugation of PNPs and the implementation of synthetic biology concepts to exploit and enhance PNP's intrinsic properties and to impart them with novel functionalities.
Collapse
Affiliation(s)
- Dennis Diaz
- Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia.
| | - Andrew Care
- Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia.
- Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, Macquarie University, Sydney, NSW 2109, Australia.
| | - Anwar Sunna
- Department of Molecular Sciences, Macquarie University, Sydney, NSW 2109, Australia.
- Australian Research Council Centre of Excellence for Nanoscale BioPhotonics, Macquarie University, Sydney, NSW 2109, Australia.
- Biomolecular Discovery and Design Research Centre, Macquarie University, Sydney, NSW 2109, Australia.
| |
Collapse
|
34
|
Galbiati E, Avvakumova S, La Rocca A, Pozzi M, Messali S, Magnaghi P, Colombo M, Prosperi D, Tortora P. A fast and straightforward procedure for vault nanoparticle purification and the characterization of its endocytic uptake. Biochim Biophys Acta Gen Subj 2018; 1862:2254-2260. [PMID: 30036602 DOI: 10.1016/j.bbagen.2018.07.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 07/12/2018] [Accepted: 07/17/2018] [Indexed: 02/03/2023]
Abstract
BACKGROUND Vaults are eukaryotic ribonucleoprotein particles composed of up 78 copies of the 97 kDa major vault protein that assembles into a barrel-like, "nanocapsule" enclosing poly(ADP-ribose) polymerase, telomerase-associated protein-1 and small untranslated RNAs. Overall, the molecular mass of vault particles amounts to about 13 MDa. Although it has been implicated in several cellular functions, its physiological roles remain poorly understood. Also, the possibility to exploit it as a nanovector for drug delivery is currently being explored in several laboratories. METHODS Using the baculovirus expression system, vaults were expressed and purified by a dialysis step using a 1 MDa molecular weight cutoff membrane and a subsequent size exclusion chromatography. Purity was assessed by SDS-PAGE, transmission electron microscopy and dynamic light scattering. Particle's endocytic uptake was monitored by flow cytometry and confocal microscopy. RESULTS The purification protocol here reported is far simpler and faster than those currently available and lead to the production of authentic vault. We then demonstrated its clathrin-mediated endocytic uptake by normal fibroblast and glioblastoma, but not carcinoma cell lines. In contrast, no significant caveolin-mediated endocytosis was detected. CONCLUSIONS These results provide the first evidence for an intrinsic propensity of the vault complex to undergo endocytic uptake cultured eukaryotic cells. GENERAL SIGNIFICANCE The newly developed purification procedure will greatly facilitate any investigation based on the use of the vault particle as a natural nanocarrier. Its clathrin-mediated endocytic uptake observed in normal and in some tumor cell lines sheds light on its physiological role.
Collapse
Affiliation(s)
- Elisabetta Galbiati
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, I-20126 Milano, Italy
| | - Svetlana Avvakumova
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, I-20126 Milano, Italy
| | - Alessandra La Rocca
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, I-20126 Milano, Italy
| | - Maria Pozzi
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, I-20126 Milano, Italy
| | - Silvia Messali
- Oncology, Nerviano Medical Sciences, Viale Pasteur 10, Milano, 20014, Nerviano, Italy
| | - Paola Magnaghi
- Oncology, Nerviano Medical Sciences, Viale Pasteur 10, Milano, 20014, Nerviano, Italy
| | - Miriam Colombo
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, I-20126 Milano, Italy
| | - Davide Prosperi
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, I-20126 Milano, Italy
| | - Paolo Tortora
- Department of Biotechnologies and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, I-20126 Milano, Italy.
| |
Collapse
|
35
|
Ding K, Zhang X, Mrazek J, Kickhoefer VA, Lai M, Ng HL, Yang OO, Rome LH, Zhou ZH. Solution Structures of Engineered Vault Particles. Structure 2018; 26:619-626.e3. [PMID: 29551289 DOI: 10.1016/j.str.2018.02.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 10/31/2017] [Accepted: 02/12/2018] [Indexed: 12/16/2022]
Abstract
Prior crystal structures of the vault have provided clues of its structural variability but are non-conclusive due to crystal packing. Here, we obtained vaults by engineering at the N terminus of rat major vault protein (MVP) an HIV-1 Gag protein segment and determined their near-atomic resolution (∼4.8 Å) structures in a solution/non-crystalline environment. The barrel-shaped vaults in solution adopt two conformations, 1 and 2, both with D39 symmetry. From the N to C termini, each MVP monomer has three regions: body, shoulder, and cap. While conformation 1 is identical to one of the crystal structures, the shoulder in conformation 2 is translocated longitudinally up to 10 Å, resulting in an outward-projected cap. Our structures clarify the structural discrepancies in the body region in the prior crystallography models. The vault's drug-delivery potential is highlighted by the internal disposition and structural flexibility of its Gag-loaded N-terminal extension at the barrel waist of the engineered vault.
Collapse
Affiliation(s)
- Ke Ding
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Xing Zhang
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jan Mrazek
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Valerie A Kickhoefer
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Mason Lai
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Hwee L Ng
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Otto O Yang
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; AIDS Healthcare Foundation, Los Angeles, CA 90028, USA
| | - Leonard H Rome
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Z Hong Zhou
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA; California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
36
|
Wang M, Abad D, Kickhoefer VA, Rome LH, Mahendra S. Encapsulation of Exogenous Proteins in Vault Nanoparticles. Methods Mol Biol 2018; 1798:25-37. [PMID: 29868949 DOI: 10.1007/978-1-4939-7893-9_3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Natural vault nanoparticles are ribonucleoprotein particles with a mass of 13 MDa that have been found in a wide variety of eukaryotes. Empty recombinant vaults are assembled from heterologously expressed Major Vault Protein (MVP), forming the barrel-shaped vault shell. These structures are morphologically indistinguishable from natural vault particles. Here, we describe the packaging and purification of exogenous proteins into these recombinant vault particles by mixing with proteins attached to the INT domain that binds to MVP.
Collapse
Affiliation(s)
- Meng Wang
- Department of Civil and Environmental Engineering, University of California, Los Angeles, CA, USA
| | - Danny Abad
- Department of Biological Chemistry, University of California, Los Angeles, CA, USA
| | - Valerie A Kickhoefer
- Department of Biological Chemistry, University of California, Los Angeles, CA, USA
| | - Leonard H Rome
- Department of Biological Chemistry, University of California, Los Angeles, CA, USA
| | - Shaily Mahendra
- Department of Civil and Environmental Engineering, University of California, Los Angeles, CA, USA.
| |
Collapse
|
37
|
Noncoding RNAs in Retrovirus Replication. RETROVIRUS-CELL INTERACTIONS 2018. [PMCID: PMC7173536 DOI: 10.1016/b978-0-12-811185-7.00012-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Although a limited percentage of the genome produces proteins, approximately 90% is transcribed, indicating important roles for noncoding RNA (ncRNA). It is now known that these ncRNAs have a multitude of cellular functions ranging from the regulation of gene expression to roles as structural elements in ribonucleoprotein complexes. ncRNA is also represented at nearly every step of viral life cycles. This chapter will focus on ncRNAs of both host and viral origin and their roles in retroviral life cycles. Cellular ncRNA represents a significant portion of material packaged into retroviral virions and includes transfer RNAs, 7SL RNA, U RNA, and vault RNA. Initially thought to be random packaging events, these host RNAs are now proposed to contribute to viral assembly and infectivity. Within the cell, long ncRNA and endogenous retroviruses have been found to regulate aspects of the retroviral life cycle in diverse ways. Additionally, the HIV-1 transactivating response element RNA is thought to impact viral infection beyond the well-characterized role as a transcription activator. RNA interference, thought to be an early version of the innate immune response to viral infection, can still be observed in plants and invertebrates today. The ability of retroviral infection to manipulate the host RNAi pathway is described here. Finally, RNA-based therapies, including gene editing approaches, are being explored as antiretroviral treatments and are discussed.
Collapse
|
38
|
Yu K, Yau YH, Sinha A, Tan T, Kickhoefer VA, Rome LH, Lee H, Shochat SG, Lim S. Modulation of the Vault Protein-Protein Interaction for Tuning of Molecular Release. Sci Rep 2017; 7:14816. [PMID: 29093465 PMCID: PMC5665922 DOI: 10.1038/s41598-017-12870-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 09/15/2017] [Indexed: 11/23/2022] Open
Abstract
Vaults are naturally occurring ovoid nanoparticles constructed from a protein shell that is composed of multiple copies of major vault protein (MVP). The vault-interacting domain of vault poly(ADP-ribose)-polymerase (INT) has been used as a shuttle to pack biomolecular cargo in the vault lumen. However, the interaction between INT and MVP is poorly understood. It is hypothesized that the release rate of biomolecular cargo from the vault lumen is related to the interaction between MVP and INT. To tune the release of molecular cargos from the vault nanoparticles, we determined the interactions between the isolated INT-interacting MVP domains (iMVP) and wild-type INT and compared them to two structurally modified INT: 15-amino acid deletion at the C terminus (INTΔC15) and histidine substituted at the interaction surface (INT/DSA/3 H) to impart a pH-sensitive response. The apparent affinity constants determined using surface plasmon resonance (SPR) biosensor technology are 262 ± 4 nM for iMVP/INT, 1800 ± 160 nM for iMVP/INTΔC15 at pH 7.4. The INT/DSA/3 H exhibits stronger affinity to iMVP (KDapp = 24 nM) and dissociates at a slower rate than wild-type INT at pH 6.0.
Collapse
Affiliation(s)
- Kang Yu
- Bioengineering Division, School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Yin Hoe Yau
- Structural Biology and Biochemistry Division, School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Ameya Sinha
- Bioengineering Division, School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore
| | - Tabitha Tan
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Valerie A Kickhoefer
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Leonard H Rome
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA.,California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Hwankyu Lee
- Department of Chemical Engineering, Dankook University, Jukjeon, Yongin, 448-701, South Korea
| | - Susana G Shochat
- Structural Biology and Biochemistry Division, School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore, 637551, Singapore
| | - Sierin Lim
- Bioengineering Division, School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, Singapore, 637457, Singapore. .,NTU-Northwestern Institute for Nanomedicine, Nanyang Technological University, 50 Nanyang Drive, Singapore, 637553, Singapore.
| |
Collapse
|
39
|
Benner NL, Zang X, Buehler DC, Kickhoefer VA, Rome ME, Rome LH, Wender PA. Vault Nanoparticles: Chemical Modifications for Imaging and Enhanced Delivery. ACS NANO 2017; 11:872-881. [PMID: 28029784 PMCID: PMC5372831 DOI: 10.1021/acsnano.6b07440] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Vault nanoparticles represent promising vehicles for drug and probe delivery. Innately found within human cells, vaults are stable, biocompatible nanocapsules possessing an internal volume that can encapsulate hundreds to thousands of molecules. They can also be targeted. Unlike most nanoparticles, vaults are nonimmunogenic and monodispersed and can be rapidly produced in insect cells. Efforts to create vaults with modified properties have been, to date, almost entirely limited to recombinant bioengineering approaches. Here we report a systematic chemical study of covalent vault modifications, directed at tuning vault properties for research and clinical applications, such as imaging, targeted delivery, and enhanced cellular uptake. As supra-macromolecular structures, vaults contain thousands of derivatizable amino acid side chains. This study is focused on establishing the comparative selectivity and efficiency of chemically modifying vault lysine and cysteine residues, using Michael additions, nucleophilic substitutions, and disulfide exchange reactions. We also report a strategy that converts the more abundant vault lysine residues to readily functionalizable thiol terminated side chains through treatment with 2-iminothiolane (Traut's reagent). These studies provide a method to doubly modify vaults with cell penetrating peptides and imaging agents, allowing for in vitro studies on their enhanced uptake into cells.
Collapse
Affiliation(s)
- Nancy L. Benner
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Xiaoyu Zang
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Daniel C. Buehler
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Valerie A. Kickhoefer
- Department of Biological Chemistry, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California 90095, United States
| | - Michael E. Rome
- Department of Biological Chemistry, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California 90095, United States
| | - Leonard H. Rome
- Department of Biological Chemistry, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California 90095, United States
| | - Paul A. Wender
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
- Department of Chemical and Systems Biology, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
40
|
Jiang J, Liu G, Kickhoefer VA, Rome LH, Li LX, McSorley SJ, Kelly KA. A Protective Vaccine against Chlamydia Genital Infection Using Vault Nanoparticles without an Added Adjuvant. Vaccines (Basel) 2017; 5:vaccines5010003. [PMID: 28106821 PMCID: PMC5371739 DOI: 10.3390/vaccines5010003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 12/23/2016] [Accepted: 01/06/2017] [Indexed: 12/01/2022] Open
Abstract
Chlamydia trachomatis genital infection is the most common sexually transmitted bacterial disease, causing a significant burden to females due to reproductive dysfunction. Intensive screening and antibiotic treatment are unable to completely prevent female reproductive dysfunction, thus, efforts have become focused on developing a vaccine. A major impediment is identifying a safe and effective adjuvant which induces cluster of differentiation 4 (CD4) cells with attributes capable of halting genital infection and inflammation. Previously, we described a natural nanocapsule called the vault which was engineered to contain major outer membrane protein (MOMP) and was an effective vaccine which significantly reduced early infection and favored development of a cellular immune response in a mouse model. In the current study, we used another chlamydial antigen, a polymorphic membrane protein G-1 (PmpG) peptide, to track antigen-specific cells and evaluate, in depth, the vault vaccine for its protective capacity in the absence of an added adjuvant. We found PmpG-vault immunized mice significantly reduced the genital bacterial burden and histopathologic parameters of inflammation following a C. muridarum challenge. Immunization boosted antigen-specific CD4 cells with a multiple cytokine secretion pattern and reduced the number of inflammatory cells in the genital tract making the vault vaccine platform safe and effective for chlamydial genital infection. We conclude that vaccination with a Chlamydia-vault vaccine boosts antigen-specific immunities that are effective at eradicating infection and preventing reproductive tract inflammation.
Collapse
Affiliation(s)
- Janina Jiang
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave. CHS 1P-177, Los Angeles, CA 90095, USA.
| | - Guangchao Liu
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave. CHS 1P-177, Los Angeles, CA 90095, USA.
| | - Valerie A Kickhoefer
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.
| | - Leonard H Rome
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.
| | - Lin-Xi Li
- Center for Comparative Medicine, Department of Anatomy, Physiology & Cell Biology, School of Veterinary Medicine, University of California-Davis, Davis, CA 95616, USA.
| | - Stephen J McSorley
- Center for Comparative Medicine, Department of Anatomy, Physiology & Cell Biology, School of Veterinary Medicine, University of California-Davis, Davis, CA 95616, USA.
| | - Kathleen A Kelly
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave. CHS 1P-177, Los Angeles, CA 90095, USA.
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
41
|
Bioengineered protein-based nanocage for drug delivery. Adv Drug Deliv Rev 2016; 106:157-171. [PMID: 26994591 DOI: 10.1016/j.addr.2016.03.002] [Citation(s) in RCA: 163] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 03/01/2016] [Accepted: 03/08/2016] [Indexed: 01/01/2023]
Abstract
Nature, in its wonders, presents and assembles the most intricate and delicate protein structures and this remarkable phenomenon occurs in all kingdom and phyla of life. Of these proteins, cage-like multimeric proteins provide spatial control to biological processes and also compartmentalizes compounds that may be toxic or unstable and avoids their contact with the environment. Protein-based nanocages are of particular interest because of their potential applicability as drug delivery carriers and their perfect and complex symmetry and ideal physical properties, which have stimulated researchers to engineer, modify or mimic these qualities. This article reviews various existing types of protein-based nanocages that are used for therapeutic purposes, and outlines their drug-loading mechanisms and bioengineering strategies via genetic and chemical functionalization. Through a critical evaluation of recent advances in protein nanocage-based drug delivery in vitro and in vivo, an outlook for de novo and in silico nanocage design, and also protein-based nanocage preclinical and future clinical applications will be presented.
Collapse
|
42
|
Llauró A, Guerra P, Kant R, Bothner B, Verdaguer N, de Pablo PJ. Decrease in pH destabilizes individual vault nanocages by weakening the inter-protein lateral interaction. Sci Rep 2016; 6:34143. [PMID: 27739422 PMCID: PMC5064368 DOI: 10.1038/srep34143] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 09/08/2016] [Indexed: 12/16/2022] Open
Abstract
Vault particles are naturally occurring proteinaceous cages with promising application as molecular containers. The use of vaults as functional transporters requires a profound understanding of their structural stability to guarantee the protection and controlled payload delivery. Previous results performed with bulk techniques or at non-physiological conditions have suggested pH as a parameter to control vault dynamics. Here we use Atomic Force Microscopy (AFM) to monitor the structural evolution of individual vault particles while changing the pH in real time. Our experiments show that decreasing the pH of the solution destabilize the barrel region, the central part of vault particles, and leads to the aggregation of the cages. Additional analyses using Quartz-Crystal Microbalance (QCM) and Differential Scanning Fluorimetry (DSF) are consistent with our single molecule AFM experiments. The observed topographical defects suggest that low pH weakens the bonds between adjacent proteins. We hypothesize that the observed effects are related to the strong polar character of the protein-protein lateral interactions. Overall, our study unveils the mechanism for the influence of a biologically relevant range of pHs on the stability and dynamics of vault particles.
Collapse
Affiliation(s)
- Aida Llauró
- Departamento de Física de la Materia Condensada, UAM, Francisco Tomás y Valiente 7, 28049-Madrid, Spain
| | - Pablo Guerra
- Structural Biology Unit, Institut de Biologia Molecular de Barcelona, CSIC. Baldiri I Reixac 10, 08028-Barcelona, Spain
| | - Ravi Kant
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, Montana, USA
| | - Brian Bothner
- Department of Chemistry and Biochemistry, Montana State University, Bozeman, Montana, USA
| | - Núria Verdaguer
- Structural Biology Unit, Institut de Biologia Molecular de Barcelona, CSIC. Baldiri I Reixac 10, 08028-Barcelona, Spain
| | - Pedro J de Pablo
- Departamento de Física de la Materia Condensada, UAM, Francisco Tomás y Valiente 7, 28049-Madrid, Spain.,Condensed Matter Physics Center IFIMAC UAM, Francisco Tomás y Valiente 7, 28049-Madrid, Spain
| |
Collapse
|
43
|
Polka JK, Hays SG, Silver PA. Building Spatial Synthetic Biology with Compartments, Scaffolds, and Communities. Cold Spring Harb Perspect Biol 2016; 8:cshperspect.a024018. [PMID: 27270297 DOI: 10.1101/cshperspect.a024018] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Traditional views of synthetic biology often treat the cell as an unstructured container in which biological reactions proceed uniformly. In reality, the organization of biological molecules has profound effects on cellular function: not only metabolic, but also physical and mechanical. Here, we discuss a variety of perturbations available to biologists in controlling protein, nucleotide, and membrane localization. These range from simple tags, fusions, and scaffolds to heterologous expression of compartments and other structures that confer unique physical properties to cells. Next, we relate these principles to those guiding the spatial environments outside of cells such as the extracellular matrix. Finally, we discuss new directions in building intercellular organizations to create novel symbioses.
Collapse
Affiliation(s)
- Jessica K Polka
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115 Wyss Institute for Biologically Inspired Engineering, Boston, Massachusetts 02115
| | - Stephanie G Hays
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115 Wyss Institute for Biologically Inspired Engineering, Boston, Massachusetts 02115
| | - Pamela A Silver
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115 Wyss Institute for Biologically Inspired Engineering, Boston, Massachusetts 02115
| |
Collapse
|
44
|
Wang M, Abad D, Kickhoefer VA, Rome LH, Mahendra S. Vault Nanoparticles Packaged with Enzymes as an Efficient Pollutant Biodegradation Technology. ACS NANO 2015; 9:10931-10940. [PMID: 26493711 DOI: 10.1021/acsnano.5b04073] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Vault nanoparticles packaged with enzymes were synthesized as agents for efficiently degrading environmental contaminants. Enzymatic biodegradation is an attractive technology for in situ cleanup of contaminated environments because enzyme-catalyzed reactions are not constrained by nutrient requirements for microbial growth and often have higher biodegradation rates. However, the limited stability of extracellular enzymes remains a major challenge for practical applications. Encapsulation is a recognized method to enhance enzymatic stability, but it can increase substrate diffusion resistance, lower catalytic rates, and increase the apparent half-saturation constants. Here, we report an effective approach for boosting enzymatic stability by single-step packaging into vault nanoparticles. With hollow core structures, assembled vault nanoparticles can simultaneously contain multiple enzymes. Manganese peroxidase (MnP), which is widely used in biodegradation of organic contaminants, was chosen as a model enzyme in the present study. MnP was incorporated into vaults via fusion to a packaging domain called INT, which strongly interacts with vaults' interior surface. MnP fused to INT and vaults packaged with the MnP-INT fusion protein maintained peroxidase activity. Furthermore, MnP-INT packaged in vaults displayed stability significantly higher than that of free MnP-INT, with slightly increased Km value. Additionally, vault-packaged MnP-INT exhibited 3 times higher phenol biodegradation in 24 h than did unpackaged MnP-INT. These results indicate that the packaging of MnP enzymes in vault nanoparticles extends their stability without compromising catalytic activity. This research will serve as the foundation for the development of efficient and sustainable vault-based bioremediation approaches for removing multiple contaminants from drinking water and groundwater.
Collapse
Affiliation(s)
- Meng Wang
- Department of Civil and Environmental Engineering, ‡Department of Biological Chemistry, and §California NanoSystems Institute, University of California , Los Angeles, California 90095, United States
| | - Danny Abad
- Department of Civil and Environmental Engineering, ‡Department of Biological Chemistry, and §California NanoSystems Institute, University of California , Los Angeles, California 90095, United States
| | - Valerie A Kickhoefer
- Department of Civil and Environmental Engineering, ‡Department of Biological Chemistry, and §California NanoSystems Institute, University of California , Los Angeles, California 90095, United States
| | - Leonard H Rome
- Department of Civil and Environmental Engineering, ‡Department of Biological Chemistry, and §California NanoSystems Institute, University of California , Los Angeles, California 90095, United States
| | - Shaily Mahendra
- Department of Civil and Environmental Engineering, ‡Department of Biological Chemistry, and §California NanoSystems Institute, University of California , Los Angeles, California 90095, United States
| |
Collapse
|
45
|
Amort M, Nachbauer B, Tuzlak S, Kieser A, Schepers A, Villunger A, Polacek N. Expression of the vault RNA protects cells from undergoing apoptosis. Nat Commun 2015; 6:7030. [PMID: 25952297 PMCID: PMC4430821 DOI: 10.1038/ncomms8030] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Accepted: 03/26/2015] [Indexed: 02/07/2023] Open
Abstract
Non-protein-coding RNAs are a functionally versatile class of transcripts exerting their biological roles on the RNA level. Recently, we demonstrated that the vault complex-associated RNAs (vtRNAs) are significantly upregulated in Epstein-Barr virus (EBV)-infected human B cells. Very little is known about the function(s) of the vtRNAs or the vault complex. Here, we individually express latent EBV-encoded proteins in B cells and identify the latent membrane protein 1 (LMP1) as trigger for vtRNA upregulation. Ectopic expression of vtRNA1-1, but not of the other vtRNA paralogues, results in an improved viral establishment and reduced apoptosis, a function located in the central domain of vtRNA1-1. Knockdown of the major vault protein has no effect on these phenotypes revealing that vtRNA1-1 and not the vault complex contributes to general cell death resistance. This study describes a NF-κB-mediated role of the non-coding vtRNA1-1 in inhibiting both the extrinsic and intrinsic apoptotic pathways.
Collapse
Affiliation(s)
- Melanie Amort
- Division of Genomics and RNomics, Medical University Innsbruck, Innsbruck A-6020, Austria
| | - Birgit Nachbauer
- Division of Genomics and RNomics, Medical University Innsbruck, Innsbruck A-6020, Austria
- Department of Chemistry and Biochemistry, University of Bern, Bern CH-3012, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern Bern 3012, Switzerland
| | - Selma Tuzlak
- Division of Developmental Immunology, Medical University Innsbruck, Innsbruck A-6020, Austria
| | - Arnd Kieser
- Research Unit Gene Vectors, Helmholtz Zentrum München, München D-81377, Germany
- German Center for Infection Research (DZIF), Partner site Munich, München D-81377, Germany
| | - Aloys Schepers
- Research Unit Gene Vectors, Helmholtz Zentrum München, München D-81377, Germany
- German Center for Infection Research (DZIF), Partner site Munich, München D-81377, Germany
| | - Andreas Villunger
- Division of Developmental Immunology, Medical University Innsbruck, Innsbruck A-6020, Austria
| | - Norbert Polacek
- Division of Genomics and RNomics, Medical University Innsbruck, Innsbruck A-6020, Austria
- Department of Chemistry and Biochemistry, University of Bern, Bern CH-3012, Switzerland
| |
Collapse
|
46
|
Woodward CL, Mendonça LM, Jensen GJ. Direct visualization of vaults within intact cells by electron cryo-tomography. Cell Mol Life Sci 2015; 72:3401-9. [PMID: 25864047 DOI: 10.1007/s00018-015-1898-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 03/13/2015] [Accepted: 04/02/2015] [Indexed: 02/04/2023]
Abstract
The vault complex is the largest cellular ribonucleoprotein complex ever characterized and is present across diverse Eukarya. Despite significant information regarding the structure, composition and evolutionary conservation of the vault, little is know about the complex's actual biological function. To determine if intracellular vaults are morphologically similar to previously studied purified and recombinant vaults, we have used electron cryo-tomography to characterize the vault complexes found in the thin edges of primary human cells growing in tissue culture. Our studies confirm that intracellular vaults are similar in overall size and shape to purified and recombinant vaults previously analyzed. Results from subtomogram averaging indicate that densities within the vault lumen are not ordered, but randomly distributed. We also observe that vaults located in the extreme periphery of the cytoplasm predominately associate with granule-like structures and actin. Our ultrastructure studies augment existing biochemical, structural and genetic information on the vault, and provide important intracellular context for the ongoing efforts to understand the biological function of the native cytoplasmic vault.
Collapse
Affiliation(s)
- Cora L Woodward
- Division of Biology, California Institute of Technology, 1200 E California Blvd, Pasadena, CA, 91125, USA
| | | | | |
Collapse
|
47
|
Mrazek J, Toso D, Ryazantsev S, Zhang X, Zhou ZH, Fernandez BC, Kickhoefer VA, Rome LH. Polyribosomes are molecular 3D nanoprinters that orchestrate the assembly of vault particles. ACS NANO 2014; 8:11552-9. [PMID: 25354757 PMCID: PMC4245718 DOI: 10.1021/nn504778h] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Ribosomes are molecular machines that function in polyribosome complexes to translate genetic information, guide the synthesis of polypeptides, and modulate the folding of nascent proteins. Here, we report a surprising function for polyribosomes as a result of a systematic examination of the assembly of a large ribonucleoprotein complex, the vault particle. Structural and functional evidence points to a model of vault assembly whereby the polyribosome acts like a 3D nanoprinter to direct the ordered translation and assembly of the multi-subunit vault homopolymer, a process which we refer to as polyribosome templating. Structure-based mutagenesis and cell-free in vitro expression studies further demonstrated the critical importance of the polyribosome in vault assembly. Polyribosome templating prevents chaos by ensuring efficiency and order in the production of large homopolymeric protein structures in the crowded cellular environment and might explain the origin of many polyribosome-associated molecular assemblies inside the cell.
Collapse
Affiliation(s)
- Jan Mrazek
- Department of Biological Chemistry, David Geffen School of Medicine, Department of Microbiology, Immunology & Molecular Genetics, and California Nanosystems Institute, University of California at Los Angeles, Los Angeles, California 90095, United States
- Address correspondence to ,
| | - Daniel Toso
- Department of Biological Chemistry, David Geffen School of Medicine, Department of Microbiology, Immunology & Molecular Genetics, and California Nanosystems Institute, University of California at Los Angeles, Los Angeles, California 90095, United States
| | - Sergey Ryazantsev
- Department of Biological Chemistry, David Geffen School of Medicine, Department of Microbiology, Immunology & Molecular Genetics, and California Nanosystems Institute, University of California at Los Angeles, Los Angeles, California 90095, United States
| | - Xing Zhang
- Department of Biological Chemistry, David Geffen School of Medicine, Department of Microbiology, Immunology & Molecular Genetics, and California Nanosystems Institute, University of California at Los Angeles, Los Angeles, California 90095, United States
| | - Z. Hong Zhou
- Department of Biological Chemistry, David Geffen School of Medicine, Department of Microbiology, Immunology & Molecular Genetics, and California Nanosystems Institute, University of California at Los Angeles, Los Angeles, California 90095, United States
| | - Beatriz Campo Fernandez
- Department of Biological Chemistry, David Geffen School of Medicine, Department of Microbiology, Immunology & Molecular Genetics, and California Nanosystems Institute, University of California at Los Angeles, Los Angeles, California 90095, United States
| | - Valerie A. Kickhoefer
- Department of Biological Chemistry, David Geffen School of Medicine, Department of Microbiology, Immunology & Molecular Genetics, and California Nanosystems Institute, University of California at Los Angeles, Los Angeles, California 90095, United States
| | - Leonard H. Rome
- Department of Biological Chemistry, David Geffen School of Medicine, Department of Microbiology, Immunology & Molecular Genetics, and California Nanosystems Institute, University of California at Los Angeles, Los Angeles, California 90095, United States
- Address correspondence to ,
| |
Collapse
|
48
|
Zhu Y, Jiang J, Said-Sadier N, Boxx G, Champion C, Tetlow A, Kickhoefer VA, Rome LH, Ojcius DM, Kelly KA. Activation of the NLRP3 inflammasome by vault nanoparticles expressing a chlamydial epitope. Vaccine 2014; 33:298-306. [PMID: 25448112 DOI: 10.1016/j.vaccine.2014.11.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 11/04/2014] [Accepted: 11/15/2014] [Indexed: 11/28/2022]
Abstract
The full potential of vaccines relies on development of effective delivery systems and adjuvants and is critical for development of successful vaccine candidates. We have shown that recombinant vaults engineered to encapsulate microbial epitopes are highly stable structures and are an ideal vaccine vehicle for epitope delivery which does not require the inclusion of an adjuvant. We studied the ability of vaults which were engineered for use as a vaccine containing an immunogenic epitope of Chlamydia trachomatis, polymorphic membrane protein G (PmpG), to be internalized into human monocytes and behave as a "natural adjuvant". We here show that incubation of monocytes with the PmpG-1-vaults activates caspase-1 and stimulates IL-1β secretion through a process requiring the NLRP3 inflammasome and that cathepsin B and Syk are involved in the inflammasome activation. We also observed that the PmpG-1-vaults are internalized through a pathway that is transiently acidic and leads to destabilization of lysosomes. In addition, immunization of mice with PmpG-1-vaults induced PmpG-1 responsive CD4(+) cells upon re-stimulation with PmpG peptide in vitro, suggesting that vault vaccines can be engineered for specific adaptive immune responses. We conclude that PmpG-1-vault vaccines can stimulate NLRP3 inflammasomes and induce PmpG-specific T cell responses.
Collapse
Affiliation(s)
- Ye Zhu
- Department of Molecular Cell Biology, and Health Sciences Research Institute, University of California, Merced, CA 95343, USA
| | - Janina Jiang
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA 90095, USA
| | - Najwane Said-Sadier
- Department of Molecular Cell Biology, and Health Sciences Research Institute, University of California, Merced, CA 95343, USA
| | - Gale Boxx
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA 90095, USA
| | - Cheryl Champion
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA 90095, USA
| | - Ashley Tetlow
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA 90095, USA
| | - Valerie A Kickhoefer
- Department of Biological Chemistry, University of California, Los Angeles, CA 90095, USA
| | - Leonard H Rome
- Department of Biological Chemistry, University of California, Los Angeles, CA 90095, USA
| | - David M Ojcius
- Department of Molecular Cell Biology, and Health Sciences Research Institute, University of California, Merced, CA 95343, USA.
| | - Kathleen A Kelly
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
49
|
Llauró A, Guerra P, Irigoyen N, Rodríguez JF, Verdaguer N, de Pablo PJ. Mechanical stability and reversible fracture of vault particles. Biophys J 2014; 106:687-95. [PMID: 24507609 DOI: 10.1016/j.bpj.2013.12.035] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 11/18/2013] [Accepted: 12/23/2013] [Indexed: 11/16/2022] Open
Abstract
Vaults are the largest ribonucleoprotein particles found in eukaryotic cells, with an unclear cellular function and promising applications as vehicles for drug delivery. In this article, we examine the local stiffness of individual vaults and probe their structural stability with atomic force microscopy under physiological conditions. Our data show that the barrel, the central part of the vault, governs both the stiffness and mechanical strength of these particles. In addition, we induce single-protein fractures in the barrel shell and monitor their temporal evolution. Our high-resolution atomic force microscopy topographies show that these fractures occur along the contacts between two major vault proteins and disappear over time. This unprecedented systematic self-healing mechanism, which enables these particles to reversibly adapt to certain geometric constraints, might help vaults safely pass through the nuclear pore complex and potentiate their role as self-reparable nanocontainers.
Collapse
Affiliation(s)
- Aida Llauró
- Departamento de Física de la Materia Condensada, Universidad Autónoma de Madrid, Madrid, Spain
| | - Pablo Guerra
- Institut de Biologia Molecular de Barcelona, Consejo Superior de Investigaciones Científicas, Barcelona, Spain
| | - Nerea Irigoyen
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, United Kingdom
| | - José F Rodríguez
- Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Núria Verdaguer
- Institut de Biologia Molecular de Barcelona, Consejo Superior de Investigaciones Científicas, Barcelona, Spain
| | - Pedro J de Pablo
- Departamento de Física de la Materia Condensada, Universidad Autónoma de Madrid, Madrid, Spain.
| |
Collapse
|
50
|
Buehler DC, Marsden MD, Shen S, Toso DB, Wu X, Loo JA, Zhou ZH, Kickhoefer VA, Wender PA, Zack JA, Rome LH. Bioengineered vaults: self-assembling protein shell-lipophilic core nanoparticles for drug delivery. ACS NANO 2014; 8:7723-32. [PMID: 25061969 PMCID: PMC4148163 DOI: 10.1021/nn5002694] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 07/25/2014] [Indexed: 05/22/2023]
Abstract
We report a novel approach to a new class of bioengineered, monodispersed, self-assembling vault nanoparticles consisting of a protein shell exterior with a lipophilic core interior designed for drug and probe delivery. Recombinant vaults were engineered to contain a small amphipathic α-helix derived from the nonstructural protein 5A of hepatitis C virus, thereby creating within the vault lumen a lipophilic microenvironment into which lipophilic compounds could be reversibly encapsulated. Multiple types of electron microscopy showed that attachment of this peptide resulted in larger than expected additional mass internalized within the vault lumen attributable to incorporation of host lipid membrane constituents spanning the vault waist (>35 nm). These bioengineered lipophilic vaults reversibly associate with a sample set of therapeutic compounds, including all-trans retinoic acid, amphotericin B, and bryostatin 1, incorporating hundreds to thousands of drug molecules per vault nanoparticle. Bryostatin 1 is of particular therapeutic interest because of its ability to potently induce expression of latent HIV, thus representing a preclinical lead in efforts to eradicate HIV/AIDS. Vaults loaded with bryostatin 1 released free drug, resulting in activation of HIV from provirus latency in vitro and induction of CD69 biomarker expression following intravenous injection into mice. The ability to preferentially and reversibly encapsulate lipophilic compounds into these novel bioengineered vault nanoparticles greatly advances their potential use as drug delivery systems.
Collapse
Affiliation(s)
- Daniel C. Buehler
- Department of Biological Chemistry, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California 90095, United States
- Department of Chemistry, Department of Chemical and Systems Biology, Stanford University, Stanford, California 94305, United States
| | - Matthew D. Marsden
- Department of Medicine, Division of Hematology and Oncology, University of California Los Angeles, Los Angeles, California 90095, United States
| | - Sean Shen
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, California 90095, United States
| | - Daniel B. Toso
- Department of Microbiology, Immunology, & Molecular Genetics, University of California Los Angeles, Los Angeles, California 90095, United States
| | - Xiaomeng Wu
- Department of Microbiology, Immunology, & Molecular Genetics, University of California Los Angeles, Los Angeles, California 90095, United States
| | - Joseph A. Loo
- Department of Biological Chemistry, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California 90095, United States
- Department of Chemistry and Biochemistry, University of California Los Angeles, Los Angeles, California 90095, United States
- UCLA−DOE Institute for Genomics and Proteomics, University of California Los Angeles, Los Angeles, California 90095, United States
| | - Z. Hong Zhou
- Department of Microbiology, Immunology, & Molecular Genetics, University of California Los Angeles, Los Angeles, California 90095, United States
- California NanoSystems Institute at University of California Los Angeles, Los Angeles, California 90095, United States
| | - Valerie A. Kickhoefer
- Department of Biological Chemistry, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California 90095, United States
| | - Paul A. Wender
- Department of Chemistry, Department of Chemical and Systems Biology, Stanford University, Stanford, California 94305, United States
| | - Jerome A. Zack
- Department of Microbiology, Immunology, & Molecular Genetics, University of California Los Angeles, Los Angeles, California 90095, United States
- California NanoSystems Institute at University of California Los Angeles, Los Angeles, California 90095, United States
- Address correspondence to ;
| | - Leonard H. Rome
- Department of Biological Chemistry, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, California 90095, United States
- California NanoSystems Institute at University of California Los Angeles, Los Angeles, California 90095, United States
- Address correspondence to ;
| |
Collapse
|