1
|
Liang Y, Wei J, Shen J, Liang Z, Ma X, Du Y, Qian W, Dong H, Huang P, Chen A, Yi C. Immunological pathogenesis and treatment progress of adenovirus pneumonia in children. Ital J Pediatr 2025; 51:4. [PMID: 39789604 PMCID: PMC11715079 DOI: 10.1186/s13052-024-01836-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025] Open
Abstract
Human adenovirus is an infectious agent that causes respiratory infections in adults and children. It has been found that immunocompromised children are highly susceptible to this pathogen, as it can swiftly evolve into severe pneumonia with multiple sequelae. Due to the lack of immunity in children, the body's response mechanisms to innate and acquired immunity are specialized. We first examined the infection classification and clinical characteristics associated with adenovirus in children. Subsequently, we explored the in-depth understanding of the pathogenic mechanism of adenovirus pneumonia in children, focusing on immunological and cellular biological aspects. Adenovirus infection in children can disrupt the balance of the innate immune response, inducing immune cells to secrete an abundance of pro-inflammatory cytokines. This cascade results in a cytokine storm, which triggers an inflammatory response and causes lung tissue damage. As a result, the infection may progress to a severe state, potentially leading to multi-organ failure. Immunocompromised children exhibit impaired immune cell numbers and functions, which affects both the secretion of antibodies to humoral immunity and the immune response of cellular immunity to adenovirus. Lastly, we reviewed the progress in treating adenovirus pneumonia in children. There are many treatments for adenovirus pneumonia in children, which must be personalized based on a thorough assessment to optimize treatment outcomes. Recent advancements in pharmaceutical development have provided new treatment options for children. Immunomodulatory therapy can reduce inflammation in children, while adjuvant therapy can improve respiratory function; however, it can also lead to complications. Further, co-infections increased the complexity of diagnosis and treatment, necessitating dynamic adjustments to treatment regimens. This review could serve as the basis for identifying potential therapeutic approaches to alleviate the symptoms associated with adenovirus infections in children.
Collapse
Affiliation(s)
- Yaowen Liang
- The Second Hospital of Nanjing, Affiliated Hospital to Nanjing University of Chinese Medicine, Nanjing, China
| | - Jie Wei
- The Second Hospital of Nanjing, Affiliated Hospital to Nanjing University of Chinese Medicine, Nanjing, China
| | - Jianjun Shen
- Department of Chinese Medicine, The Second Hospital of Nanjing, Affiliated Hospital to Nanjing University of Chinese Medicine, Nanjing, China
| | - Zihao Liang
- The Second Hospital of Nanjing, Affiliated Hospital to Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiuchang Ma
- The Second Hospital of Nanjing, Affiliated Hospital to Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuchen Du
- The Second Hospital of Nanjing, Affiliated Hospital to Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenxian Qian
- The Second Hospital of Nanjing, Affiliated Hospital to Nanjing University of Chinese Medicine, Nanjing, China
| | - Hui Dong
- The Second Hospital of Nanjing, Affiliated Hospital to Nanjing University of Chinese Medicine, Nanjing, China
| | - Ping Huang
- Department of Hepatology, The Second Hospital of Nanjing, Affiliated Hospital to Nanjing University of Chinese Medicine, Nanjing, China.
| | - Apeng Chen
- Children's Hospital of Nanjing Medical University, Nanjing, China.
| | - Changhua Yi
- The Second Hospital of Nanjing, Affiliated Hospital to Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
2
|
Halbi G, Fayer I, Aranovich D, Gat S, Pavan MJ, Nachmias D, Sanchez DS, Brik A, Granek R, Bernheim-Groswasser A. Smart design of universally decorated nanoparticles for drug delivery applications driven by active transport. THE EUROPEAN PHYSICAL JOURNAL. E, SOFT MATTER 2023; 46:74. [PMID: 37653248 DOI: 10.1140/epje/s10189-023-00331-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 07/25/2023] [Indexed: 09/02/2023]
Abstract
Targeting the cell nucleus remains a challenge for drug delivery. Here, we present a universal platform for the smart design of nanoparticle (NP) decoration that is based on: (i) a spacer polymer, commonly biotin-polyethylene-glycol-thiol, whose grafting density and molecular weight can be tuned for optimized performance, and (ii) protein binding peptides, such as cell penetrating peptides (CPPs), cancer-targeting peptides, or nuclear localization signal (NLS) peptides, that are linked to the PEG free-end by universal chemistry. We manifested our platform with two different bromo-acetamide (Br-Ac) modified NLSs. We used cell extract-based and live cell assays to demonstrate the recruitment of dynein motor proteins, which drive the NP active transport toward the nucleus, and the enhancement of cellular and nuclear entry, manifesting the properties of NLS as a CPP. Our control of the NP decoration scheme, and the modularity of our platform, carry great advantages for nano-carrier design for drug delivery applications.
Collapse
Affiliation(s)
- Gal Halbi
- The Department of Chemical Engineering, Ben-Gurion University of the Negev, 84105, Beer Sheva, Israel
| | - Itay Fayer
- The Stella and Avram Goren-Goldstein Department of Biotechnology Engineering Ben-Gurion University of the Negev, 84105, Beer Sheva, Israel
| | - Dina Aranovich
- The Department of Chemical Engineering, Ben-Gurion University of the Negev, 84105, Beer Sheva, Israel
| | - Shachar Gat
- The Department of Chemical Engineering, Ben-Gurion University of the Negev, 84105, Beer Sheva, Israel
| | - Mariela J Pavan
- The Ilse Katz Institute for Meso and Nanoscale Science and Technology, Ben-Gurion University of the Negev, 84105, Beer Sheva, Israel
| | - Dikla Nachmias
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Daniel Sevilla Sanchez
- The Ilse Katz Institute for Meso and Nanoscale Science and Technology, Ben-Gurion University of the Negev, 84105, Beer Sheva, Israel
| | - Ashraf Brik
- Schulich Faculty of Chemistry, Technion-Israel Institute of Technology, 3200008, Haifa, Israel
| | - Rony Granek
- The Stella and Avram Goren-Goldstein Department of Biotechnology Engineering Ben-Gurion University of the Negev, 84105, Beer Sheva, Israel
- The Ilse Katz Institute for Meso and Nanoscale Science and Technology, Ben-Gurion University of the Negev, 84105, Beer Sheva, Israel
| | - Anne Bernheim-Groswasser
- The Department of Chemical Engineering, Ben-Gurion University of the Negev, 84105, Beer Sheva, Israel.
- The Ilse Katz Institute for Meso and Nanoscale Science and Technology, Ben-Gurion University of the Negev, 84105, Beer Sheva, Israel.
| |
Collapse
|
3
|
Benhaghnazar RL, Medina-Kauwe L. Adenovirus-Derived Nano-Capsid Platforms for Targeted Delivery and Penetration of Macromolecules into Resistant and Metastatic Tumors. Cancers (Basel) 2023; 15:3240. [PMID: 37370850 PMCID: PMC10296971 DOI: 10.3390/cancers15123240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/31/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Macromolecular therapeutics such as nucleic acids, peptides, and proteins have the potential to overcome treatment barriers for cancer. For example, nucleic acid or peptide biologics may offer an alternative strategy for attacking otherwise undruggable therapeutic targets such as transcription factors and similar oncologic drivers. Delivery of biological therapeutics into tumor cells requires a robust system of cell penetration to access therapeutic targets within the cell interior. A highly effective means of accomplishing this may be borrowed from cell-penetrating pathogens such as viruses. In particular, the cell entry function of the adenovirus penton base capsid protein has been effective at penetrating tumor cells for the intracellular deposition of macromolecular therapies and membrane-impermeable drugs. Here, we provide an overview describing the evolution of tumor-targeted penton-base-derived nano-capsids as a framework for discussing the requirements for overcoming key barriers to macromolecular delivery. The development and pre-clinical testing of these proteins for therapeutic delivery has begun to also uncover the elusive mechanism underlying the membrane-penetrating function of the penton base. An understanding of this mechanism may unlock the potential for macromolecular therapeutics to be effectively delivered into cancer cells and to provide a treatment option for tumors resisting current clinical therapies.
Collapse
Affiliation(s)
| | - Lali Medina-Kauwe
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA;
- Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
4
|
Martín-González N, Gómez-González A, Hernando-Pérez M, Bauer M, Greber UF, San Martín C, de Pablo PJ. Adenovirus core protein V reinforces the capsid and enhances genome release from disrupted particles. SCIENCE ADVANCES 2023; 9:eade9910. [PMID: 37027464 PMCID: PMC10081844 DOI: 10.1126/sciadv.ade9910] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 03/02/2023] [Indexed: 06/19/2023]
Abstract
Out of the three core proteins in human adenovirus, protein V is believed to connect the inner capsid surface to the outer genome layer. Here, we explored mechanical properties and in vitro disassembly of particles lacking protein V (Ad5-ΔV). Ad5-ΔV particles were softer and less brittle than the wild-type ones (Ad5-wt), but they were more prone to release pentons under mechanical fatigue. In Ad5-ΔV, core components did not readily diffuse out of partially disrupted capsids, and the core appeared more condensed than in Ad5-wt. These observations suggest that instead of condensing the genome, protein V antagonizes the condensing action of the other core proteins. Protein V provides mechanical reinforcement and facilitates genome release by keeping DNA connected to capsid fragments that detach during disruption. This scenario is in line with the location of protein V in the virion and its role in Ad5 cell entry.
Collapse
Affiliation(s)
- Natalia Martín-González
- Departament of Condensed Matter Physics, Universidad Autónoma de Madrid and Institute of Condensed Matter Physics (IFIMAC), 28049 Madrid, Spain
| | - Alfonso Gómez-González
- Department of Molecular Life Sciences, University of Zurich, CH-8057 Zurich, Switzerland
| | - Mercedes Hernando-Pérez
- Department of Macromolecular Structures, Centro Nacional de Biotecnología (CNB-CSIC), 28049 Madrid, Spain
| | - Michael Bauer
- Department of Molecular Life Sciences, University of Zurich, CH-8057 Zurich, Switzerland
| | - Urs F. Greber
- Department of Molecular Life Sciences, University of Zurich, CH-8057 Zurich, Switzerland
| | - Carmen San Martín
- Department of Macromolecular Structures, Centro Nacional de Biotecnología (CNB-CSIC), 28049 Madrid, Spain
| | - Pedro J. de Pablo
- Departament of Condensed Matter Physics, Universidad Autónoma de Madrid and Institute of Condensed Matter Physics (IFIMAC), 28049 Madrid, Spain
| |
Collapse
|
5
|
Río-Bergé C, Cong Y, Reggiori F. Getting on the right track: Interactions between viruses and the cytoskeletal motor proteins. Traffic 2023; 24:114-130. [PMID: 35146839 DOI: 10.1111/tra.12835] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 02/06/2022] [Accepted: 02/07/2022] [Indexed: 11/29/2022]
Abstract
The cytoskeleton is an essential component of the cell and it is involved in multiple physiological functions, including intracellular organization and transport. It is composed of three main families of proteinaceous filaments; microtubules, actin filaments and intermediate filaments and their accessory proteins. Motor proteins, which comprise the dynein, kinesin and myosin superfamilies, are a remarkable group of accessory proteins that mainly mediate the intracellular transport of cargoes along with the cytoskeleton. Like other cellular structures and pathways, viruses can exploit the cytoskeleton to promote different steps of their life cycle through associations with motor proteins. The complexity of the cytoskeleton and the differences among viruses, however, has led to a wide diversity of interactions, which in most cases remain poorly understood. Unveiling the details of these interactions is necessary not only for a better comprehension of specific infections, but may also reveal new potential drug targets to fight dreadful diseases such as rabies disease and acquired immunodeficiency syndrome (AIDS). In this review, we describe a few examples of the mechanisms that some human viruses, that is, rabies virus, adenovirus, herpes simplex virus, human immunodeficiency virus, influenza A virus and papillomavirus, have developed to hijack dyneins, kinesins and myosins.
Collapse
Affiliation(s)
- Clàudia Río-Bergé
- Department of Biomedical Sciences of Cells & Systems, Molecular Cell Biology Section, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Yingying Cong
- Department of Biomedical Sciences of Cells & Systems, Molecular Cell Biology Section, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Fulvio Reggiori
- Department of Biomedical Sciences of Cells & Systems, Molecular Cell Biology Section, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
6
|
Hasanzadeh A, Hamblin MR, Kiani J, Noori H, Hardie JM, Karimi M, Shafiee H. Could artificial intelligence revolutionize the development of nanovectors for gene therapy and mRNA vaccines? NANO TODAY 2022; 47:101665. [PMID: 37034382 PMCID: PMC10081506 DOI: 10.1016/j.nantod.2022.101665] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Gene therapy enables the introduction of nucleic acids like DNA and RNA into host cells, and is expected to revolutionize the treatment of a wide range of diseases. This growth has been further accelerated by the discovery of CRISPR/Cas technology, which allows accurate genomic editing in a broad range of cells and organisms in vitro and in vivo. Despite many advances in gene delivery and the development of various viral and non-viral gene delivery vectors, the lack of highly efficient non-viral systems with low cellular toxicity remains a challenge. The application of cutting-edge technologies such as artificial intelligence (AI) has great potential to find new paradigms to solve this issue. Herein, we review AI and its major subfields including machine learning (ML), neural networks (NNs), expert systems, deep learning (DL), computer vision and robotics. We discuss the potential of AI-based models and algorithms in the design of targeted gene delivery vehicles capable of crossing extracellular and intracellular barriers by viral mimicry strategies. We finally discuss the role of AI in improving the function of CRISPR/Cas systems, developing novel nanobots, and mRNA vaccine carriers.
Collapse
Affiliation(s)
- Akbar Hasanzadeh
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa
- Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Jafar Kiani
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamid Noori
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
| | - Joseph M. Hardie
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02139 USA
| | - Mahdi Karimi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran 1449614535, Iran
- Research Center for Science and Technology in Medicine, Tehran University of Medical Sciences, Tehran 141556559, Iran
- Applied Biotechnology Research Centre, Tehran Medical Science, Islamic Azad University, Tehran 1584743311, Iran
| | - Hadi Shafiee
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, 02139 USA
| |
Collapse
|
7
|
Halbi G, Fayer I, Aranovich D, Gat S, Bar S, Erukhimovitch V, Granek R, Bernheim-Groswasser A. Nano-Particles Carried by Multiple Dynein Motors Self-Regulate Their Number of Actively Participating Motors. Int J Mol Sci 2021; 22:ijms22168893. [PMID: 34445598 PMCID: PMC8396316 DOI: 10.3390/ijms22168893] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/28/2021] [Accepted: 08/04/2021] [Indexed: 12/13/2022] Open
Abstract
Intra-cellular active transport by native cargos is ubiquitous. We investigate the motion of spherical nano-particles (NPs) grafted with flexible polymers that end with a nuclear localization signal peptide. This peptide allows the recruitment of several mammalian dynein motors from cytoplasmic extracts. To determine how motor–motor interactions influenced motility on the single microtubule level, we conducted bead-motility assays incorporating surface adsorbed microtubules and combined them with model simulations that were based on the properties of a single dynein. The experimental and simulation results revealed long time trajectories: when the number of NP-ligated motors Nm increased, run-times and run-lengths were enhanced and mean velocities were somewhat decreased. Moreover, the dependence of the velocity on run-time followed a universal curve, regardless of the system composition. Model simulations also demonstrated left- and right-handed helical motion and revealed self-regulation of the number of microtubule-bound, actively transporting dynein motors. This number was stochastic along trajectories and was distributed mainly between one, two, and three motors, regardless of Nm. We propose that this self-regulation allows our synthetic NPs to achieve persistent motion that is associated with major helicity. Such a helical motion might affect obstacle bypassing, which can influence active transport efficiency when facing the crowded environment of the cell.
Collapse
Affiliation(s)
- Gal Halbi
- The Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (G.H.); (D.A.); (S.G.); (S.B.); (V.E.)
| | - Itay Fayer
- The Stella and Avram Goren-Goldstein Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel;
| | - Dina Aranovich
- The Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (G.H.); (D.A.); (S.G.); (S.B.); (V.E.)
| | - Shachar Gat
- The Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (G.H.); (D.A.); (S.G.); (S.B.); (V.E.)
| | - Shay Bar
- The Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (G.H.); (D.A.); (S.G.); (S.B.); (V.E.)
| | - Vitaly Erukhimovitch
- The Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (G.H.); (D.A.); (S.G.); (S.B.); (V.E.)
| | - Rony Granek
- The Stella and Avram Goren-Goldstein Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel;
- The Ilse Katz Institute for Meso and Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
- Correspondence: (R.G.); (A.B.-G.)
| | - Anne Bernheim-Groswasser
- The Department of Chemical Engineering, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (G.H.); (D.A.); (S.G.); (S.B.); (V.E.)
- The Ilse Katz Institute for Meso and Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
- Correspondence: (R.G.); (A.B.-G.)
| |
Collapse
|
8
|
Daussy CF, Pied N, Wodrich H. Understanding Post Entry Sorting of Adenovirus Capsids; A Chance to Change Vaccine Vector Properties. Viruses 2021; 13:1221. [PMID: 34202573 PMCID: PMC8310329 DOI: 10.3390/v13071221] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 12/25/2022] Open
Abstract
Adenovirus vector-based genetic vaccines have emerged as a powerful strategy against the SARS-CoV-2 health crisis. This success is not unexpected because adenoviruses combine many desirable features of a genetic vaccine. They are highly immunogenic and have a low and well characterized pathogenic profile paired with technological approachability. Ongoing efforts to improve adenovirus-vaccine vectors include the use of rare serotypes and non-human adenoviruses. In this review, we focus on the viral capsid and how the choice of genotypes influences the uptake and subsequent subcellular sorting. We describe how understanding capsid properties, such as stability during the entry process, can change the fate of the entering particles and how this translates into differences in immunity outcomes. We discuss in detail how mutating the membrane lytic capsid protein VI affects species C viruses' post-entry sorting and briefly discuss if such approaches could have a wider implication in vaccine and/or vector development.
Collapse
Affiliation(s)
| | | | - Harald Wodrich
- Microbiologie Fondamentale et Pathogénicité, MFP CNRS UMR 5234, University of Bordeaux, 146 rue Leo Saignat, CEDEX, 33076 Bordeaux, France; (C.F.D.); (N.P.)
| |
Collapse
|
9
|
Tosolini AP, Sleigh JN. Intramuscular Delivery of Gene Therapy for Targeting the Nervous System. Front Mol Neurosci 2020; 13:129. [PMID: 32765219 PMCID: PMC7379875 DOI: 10.3389/fnmol.2020.00129] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 06/26/2020] [Indexed: 12/11/2022] Open
Abstract
Virus-mediated gene therapy has the potential to deliver exogenous genetic material into specific cell types to promote survival and counteract disease. This is particularly enticing for neuronal conditions, as the nervous system is renowned for its intransigence to therapeutic targeting. Administration of gene therapy viruses into skeletal muscle, where distal terminals of motor and sensory neurons reside, has been shown to result in extensive transduction of cells within the spinal cord, brainstem, and sensory ganglia. This route is minimally invasive and therefore clinically relevant for gene therapy targeting to peripheral nerve soma. For successful transgene expression, viruses administered into muscle must undergo a series of processes, including host cell interaction and internalization, intracellular sorting, long-range retrograde axonal transport, endosomal liberation, and nuclear import. In this review article, we outline key characteristics of major gene therapy viruses—adenovirus, adeno-associated virus (AAV), and lentivirus—and summarize the mechanisms regulating important steps in the virus journey from binding at peripheral nerve terminals to nuclear delivery. Additionally, we describe how neuropathology can negatively influence these pathways, and conclude by discussing opportunities to optimize the intramuscular administration route to maximize gene delivery and thus therapeutic potential.
Collapse
Affiliation(s)
- Andrew P Tosolini
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - James N Sleigh
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, United Kingdom.,UK Dementia Research Institute, University College London, London, United Kingdom
| |
Collapse
|
10
|
Abstract
Both well-known and emerging viruses increasingly affect humans and cause disease, sometimes with devastating impact on society. The viruses present in the biosphere are the top predators in the life chain, virtually without enemies, except perhaps the immune system, and harsh environmental physicochemical conditions restricting their dissemination. We know a lot about viruses, but do we know enough? This series of reviews is dedicated to adenoviruses (AdVs), a family of nonenveloped DNA viruses occurring in vertebrates, including humans. AdVs have been the focus of intense research for more than 67 years. Besides causing disease, they have immensely contributed to the advance of life sciences and medicine over the past decades. Recently, AdVs have been widely used as vehicles in gene therapy and vaccination. They continue to provide fundamental insights into virus-host interactions in cells, tissues and organisms, as well as systems and metabolic networks. This special issue of FEBS Letters presents a unique collection of 23 state-of-the-art review articles by leading adenovirologists. In this prelude, I present the chapters, which provide a solid basis for further exploring the rich heritage in adenovirus molecular cell biology, structural biology, genetics, immunology, gene therapy and epidemiology. I conclude with an essential discussion of six blind spots in adenovirology.
Collapse
Affiliation(s)
- Urs F Greber
- Department of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| |
Collapse
|
11
|
Hossain E, Habiba U, Yanagawa-Matsuda A, Alam A, Ahmed I, Towfik Alam M, Yasuda M, Higashino F. Advantages of Using Paclitaxel in Combination with Oncolytic Adenovirus Utilizing RNA Destabilization Mechanism. Cancers (Basel) 2020; 12:cancers12051210. [PMID: 32408515 PMCID: PMC7281177 DOI: 10.3390/cancers12051210] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 04/29/2020] [Accepted: 05/07/2020] [Indexed: 12/17/2022] Open
Abstract
Oncolytic virotherapy is a novel approach to cancer therapy. Ad-fosARE is a conditionally replicative adenovirus engineered by inserting AU-rich elements (ARE) in the 3'-untranslated region of the E1A gene. In this study, we examined the oncolytic activity of Ad-fosARE and used it in a synergistic combination with the chemotherapeutic agent paclitaxel (PTX) for treating cancer cells. The expression of E1A was high in cancer cells due to stabilized E1A-ARE mRNA. As a result, the efficiency of its replication and cytolytic activity in cancer cells was higher than in normal cells. PTX treatment increased the cytoplasmic HuR relocalization in cancer cells, enhanced viral replication through elevated E1A expression, and upregulated CAR (Coxsackie-adenovirus receptor) required for viral uptake. Furthermore, PTX altered the instability of microtubules by acetylation and detyrosination, which is essential for viral internalization and trafficking to the nucleus. These results indicate that PTX can provide multiple advantages to the efficacy of Ad-fosARE both in vitro and in vivo, and provides a basis for designing novel clinical trials. Thus, this virus has a lot of benefits that are not found in other oncolytic viruses. The virus also has the potential for treating PXT-resistant cancers.
Collapse
Affiliation(s)
- Elora Hossain
- Department of Molecular Oncology, Faculty of Dental Medicine and Graduate School of Biomedical Science and Engineering, Hokkaido University, Sapporo 060-8638, Japan; (E.H.); (I.A.)
| | - Umma Habiba
- Department of Cancer Pathology, Faculty of Medicine, Hokkaido University, Sapporo 060-8638, Japan;
| | - Aya Yanagawa-Matsuda
- Department of Vascular Biology and Molecular Pathology, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Sapporo 060-8586, Japan; (A.Y.-M.); (M.T.A.)
| | - Arefin Alam
- Department of Restorative Dentistry, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Sapporo 060-8586, Japan;
| | - Ishraque Ahmed
- Department of Molecular Oncology, Faculty of Dental Medicine and Graduate School of Biomedical Science and Engineering, Hokkaido University, Sapporo 060-8638, Japan; (E.H.); (I.A.)
| | - Mohammad Towfik Alam
- Department of Vascular Biology and Molecular Pathology, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Sapporo 060-8586, Japan; (A.Y.-M.); (M.T.A.)
| | - Motoaki Yasuda
- Department of Oral Molecular Microbiology, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Sapporo 060-8586, Japan;
| | - Fumihiro Higashino
- Department of Molecular Oncology, Faculty of Dental Medicine and Graduate School of Biomedical Science and Engineering, Hokkaido University, Sapporo 060-8638, Japan; (E.H.); (I.A.)
- Department of Vascular Biology and Molecular Pathology, Faculty of Dental Medicine and Graduate School of Dental Medicine, Hokkaido University, Sapporo 060-8586, Japan; (A.Y.-M.); (M.T.A.)
- Correspondence: ; Tel.: +81-(0)11-706-4237
| |
Collapse
|
12
|
Scherer J, Yi J, Vallee RB. Role of cytoplasmic dynein and kinesins in adenovirus transport. FEBS Lett 2020; 594:1838-1847. [PMID: 32215924 DOI: 10.1002/1873-3468.13777] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 03/12/2020] [Accepted: 03/15/2020] [Indexed: 12/30/2022]
Abstract
Following receptor-mediated uptake into endocytic vesicles and subsequent escape, adenovirus particles are transported along microtubules. The microtubule motor proteins dynein and one or more kinesins are involved in this behavior. Dynein is implicated in adenovirus transport toward the nucleus. The kinesin Kif5B has now been found to move the adenovirus (AdV) toward microtubule plus ends, though a kinesin role in adenovirus-induced nuclear pore disruption has also been reported. In undifferentiated cells, dynein-mediated transport predominates early in infection, but motility becomes bidirectional with time. The latter behavior can be modeled as a novel assisted diffusion mechanism, which may allow virus particles to explore the cytoplasm more efficiently. Cytoplasmic dynein and Kif5B have both been found to bind AdV through direct interactions with the capsid proteins hexon and penton base, respectively. We review here the roles of the microtubule motor proteins in AdV infection, the relationship between motor protein recruitment to pathogenic vs. physiological cargoes, the evolutionary origins of microtubule-mediated AdV transport, and a role for the motor proteins in a novel host-defense mechanism.
Collapse
Affiliation(s)
- Julian Scherer
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Julie Yi
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Richard B Vallee
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| |
Collapse
|
13
|
Abstract
Viruses are obligatory parasites that take advantage of intracellular niches to replicate. During infection, their genomes are carried in capsids across the membranes of host cells to sites of virion production by exploiting cellular behaviour and resources to guide and achieve all aspects of delivery and the downstream virus manufacturing process. Successful entry hinges on execution of a precisely tuned viral uncoating program where incoming capsids disassemble in consecutive steps to ensure that genomes are released at the right time, and in the right place for replication to occur. Each step of disassembly is cell-assisted, involving individual pathways that transmit signals to regulate discrete functions, but at the same time, these signalling pathways are organized into larger networks, which communicate back and forth in complex ways in response to the presence of virus. In this review, we consider the elegant strategy by which adenoviruses (AdVs) target and navigate cellular networks to initiate the production of progeny virions. There are many remarkable aspects about the AdV entry program; for example, the virus gains targeted control of a large well-defined local network neighbourhood by coupling several interacting processes (including endocytosis, autophagy and microtubule trafficking) around a collective reference state centred on the interactional topology and multifunctional nature of protein VI. Understanding the network targeting activity of protein VI, as well as other built-in mechanisms that allow AdV particles to be efficient at navigating the subsystems of the cell, can be used to improve viral vectors, but also has potential to be incorporated for use in entirely novel delivery systems.
Collapse
Affiliation(s)
- Justin W Flatt
- Faculty of Biological and Environmental Sciences and HiLIFE-Institute of Biotechnology, University of Helsinki , 00790 Helsinki , Finland
| | - Sarah J Butcher
- Faculty of Biological and Environmental Sciences and HiLIFE-Institute of Biotechnology, University of Helsinki , 00790 Helsinki , Finland
| |
Collapse
|
14
|
Pied N, Wodrich H. Imaging the adenovirus infection cycle. FEBS Lett 2019; 593:3419-3448. [PMID: 31758703 DOI: 10.1002/1873-3468.13690] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/18/2019] [Accepted: 11/20/2019] [Indexed: 12/11/2022]
Abstract
Incoming adenoviruses seize control of cytosolic transport mechanisms to relocate their genome from the cell periphery to specialized sites in the nucleoplasm. The nucleus is the site for viral gene expression, genome replication, and the production of progeny for the next round of infection. By taking control of the cell, adenoviruses also suppress cell-autonomous immunity responses. To succeed in their production cycle, adenoviruses rely on well-coordinated steps, facilitated by interactions between viral proteins and cellular factors. Interactions between virus and host can impose remarkable morphological changes in the infected cell. Imaging adenoviruses has tremendously influenced how we delineate individual steps in the viral life cycle, because it allowed the development of specific optical markers to label these morphological changes in space and time. As technology advances, innovative imaging techniques and novel tools for specimen labeling keep uncovering previously unseen facets of adenovirus biology emphasizing why imaging adenoviruses is as attractive today as it was in the past. This review will summarize past achievements and present developments in adenovirus imaging centered on fluorescence microscopy approaches.
Collapse
Affiliation(s)
- Noémie Pied
- CNRS UMR 5234, Microbiologie Fondamentale et Pathogénicité, Université de Bordeaux, France
| | - Harald Wodrich
- CNRS UMR 5234, Microbiologie Fondamentale et Pathogénicité, Université de Bordeaux, France
| |
Collapse
|
15
|
Alonso-Valenteen F, Pacheco S, Srinivas D, Rentsendorj A, Chu D, Lubow J, Sims J, Miao T, Mikhael S, Hwang JY, Abrol R, Medina Kauwe LK. HER3-targeted protein chimera forms endosomolytic capsomeres and self-assembles into stealth nucleocapsids for systemic tumor homing of RNA interference in vivo. Nucleic Acids Res 2019; 47:11020-11043. [PMID: 31617560 PMCID: PMC6868389 DOI: 10.1093/nar/gkz900] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 09/12/2019] [Accepted: 10/09/2019] [Indexed: 12/31/2022] Open
Abstract
RNA interference represents a potent intervention for cancer treatment but requires a robust delivery agent for transporting gene-modulating molecules, such as small interfering RNAs (siRNAs). Although numerous molecular approaches for siRNA delivery are adequate in vitro, delivery to therapeutic targets in vivo is limited by payload integrity, cell targeting, efficient cell uptake, and membrane penetration. We constructed nonviral biomaterials to transport small nucleic acids to cell targets, including tumor cells, on the basis of the self-assembling and cell-penetrating activities of the adenovirus capsid penton base. Our recombinant penton base chimera contains polypeptide domains designed for noncovalent assembly with anionic molecules and tumor homing. Here, structural modeling, molecular dynamics simulations, and functional assays suggest that it forms pentameric units resembling viral capsomeres that assemble into larger capsid-like structures when combined with siRNA cargo. Pentamerization forms a barrel lined with charged residues mediating pH-responsive dissociation and exposing masked domains, providing insight on the endosomolytic mechanism. The therapeutic impact was examined on tumors expressing high levels of HER3/ErbB3 that are resistant to clinical inhibitors. Our findings suggest that our construct may utilize ligand mimicry to avoid host attack and target the siRNA to HER3+ tumors by forming multivalent capsid-like structures.
Collapse
Affiliation(s)
- Felix Alonso-Valenteen
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Sayuri Pacheco
- Department of Chemistry and Biochemistry, California State University, Northridge, CA 91330, USA
| | - Dustin Srinivas
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Altan Rentsendorj
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - David Chu
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jay Lubow
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jessica Sims
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Tianxin Miao
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Simoun Mikhael
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jae Youn Hwang
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Information and Communication Engineering, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Korea
| | - Ravinder Abrol
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Chemistry and Biochemistry, California State University, Northridge, CA 91330, USA
| | - Lali K Medina Kauwe
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
16
|
Abstract
The integration of drugs into nanocarriers favorably altered their pharmacodynamics and pharmacokinetics compared to free drugs, and increased their therapeutic index. However, selective cellular internalization in diseased tissues rather than normal tissues still presents a formidable challenge. In this chapter I will cover solutions involving environment-responsive cell-penetrating peptides (CPPs). I will discuss properties of CPPs as universal cellular uptake enhancers, and the modifications imparted to CPP-modified nanocarriers to confine CPP activation to diseased tissues.
Collapse
|
17
|
Abstract
More than 80 different adenovirus (AdV) types infect humans through the respiratory, ocular, or gastrointestinal tracts. They cause acute clinical mani-festations or persist under humoral and cell-based immunity. Immuno-suppressed individuals are at risk of death from an AdV infection. Concepts about cell entry of AdV build on strong foundations from molecular and cellular biology-and increasingly physical virology. Here, we discuss how virions enter and deliver their genome into the nucleus of epithelial cells. This process breaks open the virion at distinct sites because the particle has nonisometric mechanical strength and reacts to specific host factors along the entry pathway. We further describe how macrophages and dendritic cells resist AdV infection yet enhance productive entry into polarized epithelial cells. A deep understanding of the viral mechanisms and cell biological and biophysical principles will continue to unravel how epithelial and antigen-presenting cells respond to AdVs and control inflammation and persistence in pathology and therapy.
Collapse
Affiliation(s)
- Urs F Greber
- Department of Molecular Life Sciences, University of Zurich, 8057 Zurich, Switzerland;
| | - Justin W Flatt
- Institute of Biotechnology and Department of Biosciences, University of Helsinki, 00790 Helsinki, Finland;
| |
Collapse
|
18
|
Badr KR, Parente‐Rocha JA, Baeza LC, Ficcadori FS, Souza M, Soares CM, Guissoni ACP, Almeida TN, Cardoso DD. Quantitative proteomic analysis of A549 cells infected with human adenovirus type 2. J Med Virol 2019; 91:1239-1249. [DOI: 10.1002/jmv.25439] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 01/30/2019] [Accepted: 02/19/2019] [Indexed: 12/31/2022]
Affiliation(s)
- Kareem R. Badr
- Department of Microbiology, Human Virology LaboratoryInstitute of Tropical Pathology and Public Health, Federal University of GoiásGoiânia Goiás Brazil
| | - Juliana A. Parente‐Rocha
- Department of Microbiology, Molecular Biology LaboratoryInstitute of Biological Sciences, Federal University of GoiásGoiânia Goiás Brazil
| | - Lilian C. Baeza
- Department of Microbiology, Molecular Biology LaboratoryInstitute of Biological Sciences, Federal University of GoiásGoiânia Goiás Brazil
| | - Fabiola S. Ficcadori
- Department of Microbiology, Human Virology LaboratoryInstitute of Tropical Pathology and Public Health, Federal University of GoiásGoiânia Goiás Brazil
| | - Menira Souza
- Department of Microbiology, Human Virology LaboratoryInstitute of Tropical Pathology and Public Health, Federal University of GoiásGoiânia Goiás Brazil
| | - Célia M. Soares
- Department of Microbiology, Molecular Biology LaboratoryInstitute of Biological Sciences, Federal University of GoiásGoiânia Goiás Brazil
| | - Ana Carla P. Guissoni
- Department of Microbiology, Human Virology LaboratoryInstitute of Tropical Pathology and Public Health, Federal University of GoiásGoiânia Goiás Brazil
| | - Tâmera N. Almeida
- Department of Microbiology, Human Virology LaboratoryInstitute of Tropical Pathology and Public Health, Federal University of GoiásGoiânia Goiás Brazil
| | - Divina D. Cardoso
- Department of Microbiology, Human Virology LaboratoryInstitute of Tropical Pathology and Public Health, Federal University of GoiásGoiânia Goiás Brazil
| |
Collapse
|
19
|
Abstract
The gap junctions (GJs), which form intercellular communicating channels between two apposing cells or form hemichannel with extracellular environment, perform crucial functions to maintain small molecule homeostasis. The central nervous system (CNS) GJs are important for maintenance of myelin sheath and neuronal activity. Connexin (Cx) proteins are building blocks of GJs. Recent cell-biological investigations show that amongst the CNS specific Cxs, the most abundant Cx protein, Cx43 and its oligodendrocytic coupling partner Cx47 primarily important for maintenance of CNS myelin. Recent investigations elucidate that the expression of Cx43 and Cx47 is very important to maintain K+ buffering and nutrient homeostasis in oligodendrocytes, CNS myelin and oligodendrocyte function. The investigations on Multiple Sclerosis (MS) patient samples and EAE hypothesized that the functional loss of Cx43/Cx47 could be associated with spread of chronic MS lesions. Exploring the mechanism of initial GJ alteration and its effect on demyelination in this model of MS might play a primary role to understand the basis of altered CNS homeostasis, observed during MS. In this review, we mainly discuss the role of CNS GJs, specifically the Cx43/Cx47 axis in the perspective of demyelination.
Collapse
|
20
|
Lee JS, Ismail AM, Lee JY, Zhou X, Materne EC, Chodosh J, Rajaiya J. Impact of dynamin 2 on adenovirus nuclear entry. Virology 2019; 529:43-56. [PMID: 30660774 DOI: 10.1016/j.virol.2019.01.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 01/06/2019] [Accepted: 01/07/2019] [Indexed: 01/28/2023]
Abstract
The large GTPase dynamin 2 controls both endosomal fission and microtubule acetylation. Here we report that dynamin 2 alters microtubules and regulates the trafficking of human adenovirus type 37. Dynamin 2 knockdown by siRNA in infected cells resulted in accumulation of acetylated tubulin, repositioning of microtubule organizing centers (MTOCs) closer to cell nuclei, increased virus in the cytosol (with a compensatory decrease in endosomal virus), reduced proinflammatory cytokine induction, and increased binding of virus to the nucleoporin, Nup358. These events led to increased viral DNA nuclear entry and viral replication. Overexpression of dynamin 2 generated opposite effects. Therefore, dynamin 2 inhibits adenovirus replication and promotes innate immune responses by the infected cell. MTOC transposition in dynamin 2 knockdown promotes a closer association with nuclear pore complexes to facilitate viral DNA delivery. Dynamin 2 plays a key role in adenoviral trafficking and influences host responses to infection.
Collapse
Affiliation(s)
- Ji Sun Lee
- Howe Laboratory, Mass Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Ashrafali M Ismail
- Howe Laboratory, Mass Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Jeong Yoon Lee
- Howe Laboratory, Mass Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Xiaohong Zhou
- Howe Laboratory, Mass Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Emma C Materne
- Howe Laboratory, Mass Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - James Chodosh
- Howe Laboratory, Mass Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Jaya Rajaiya
- Howe Laboratory, Mass Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
21
|
Basu R, Sarma JDAS. Connexin 43/47 channels are important for astrocyte/ oligodendrocyte cross-talk in myelination and demyelination. J Biosci 2018; 43:1055-1068. [PMID: 30541963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2022]
Abstract
The gap junctions (GJs), which form intercellular communicating channels between two apposing cells or form hemichannel with extracellular environment, perform crucial functions to maintain small molecule homeostasis. The central nervous system (CNS) GJs are important for maintenance of myelin sheath and neuronal activity. Connexin (Cx) proteins are building blocks of GJs. Recent cell-biological investigations show that amongst the CNS specific Cxs, the most abundant Cx protein, Cx43 and its oligodendrocytic coupling partner Cx47 primarily important for maintenance of CNS myelin. Recent investigations elucidate that the expression of Cx43 and Cx47 is very important to maintain K? buffering and nutrient homeostasis in oligodendrocytes, CNS myelin and oligodendrocyte function. The investigations on Multiple Sclerosis (MS) patient samples and EAE hypothesized that the functional loss of Cx43/Cx47 could be associated with spread of chronic MS lesions. Exploring the mechanism of initial GJ alteration and its effect on demyelination in this model of MS might play a primary role to understand the basis of altered CNS homeostasis, observed during MS. In this review, we mainly discuss the role of CNS GJs, specifically the Cx43/Cx47 axis in the perspective of demyelination.
Collapse
Affiliation(s)
- Rahul Basu
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur 741246, India
| | | |
Collapse
|
22
|
Marrugal-Lorenzo JA, Serna-Gallego A, González-González L, Buñuales M, Poutou J, Pachón J, Gonzalez-Aparicio M, Hernandez-Alcoceba R, Sánchez-Céspedes J. Inhibition of adenovirus infection by mifepristone. Antiviral Res 2018; 159:77-83. [PMID: 30268911 DOI: 10.1016/j.antiviral.2018.09.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 09/20/2018] [Accepted: 09/26/2018] [Indexed: 11/18/2022]
Abstract
The repurposing of drugs approved by the regulatory agencies for other indications is emerging as a valuable alternative for the development of new antimicrobial therapies, involving lower risks and costs than the de novo development of novel antimicrobial drugs. Adenovirus infections have showed a steady increment in recent years, with a high clinical impact in both immunosuppressed and immunocompetent patients. In this context, the lack of a specific drug to treat these infections supports the search for new therapeutic alternatives. In this study, we examined the anti-HAdV properties of mifepristone, a commercially available synthetic steroid drug. Mifepristone showed significant in vitro anti-HAdV activity at low micromolar concentrations with little cytotoxicity. Our mechanistic assays suggest that this drug could affect the microtubule transport, interfering with the entry of the virus into the nucleus and therefore inhibiting HAdV infection.
Collapse
Affiliation(s)
- José A Marrugal-Lorenzo
- Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, Clinical Unit of Infectious Diseases, Microbiology, and Preventive Medicine, Seville, Spain
| | - Ana Serna-Gallego
- Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, Clinical Unit of Infectious Diseases, Microbiology, and Preventive Medicine, Seville, Spain
| | - Loreto González-González
- Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, Clinical Unit of Infectious Diseases, Microbiology, and Preventive Medicine, Seville, Spain
| | - Maria Buñuales
- Gene Therapy Unit CIMA, Foundation for Applied Medical Research, University of Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Spain
| | - Joanna Poutou
- Gene Therapy Unit CIMA, Foundation for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Jerónimo Pachón
- Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, Clinical Unit of Infectious Diseases, Microbiology, and Preventive Medicine, Seville, Spain; Department of Medicine, University of Seville, Seville, Spain
| | - Manuela Gonzalez-Aparicio
- Gene Therapy Unit CIMA, Foundation for Applied Medical Research, University of Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Spain
| | - Ruben Hernandez-Alcoceba
- Gene Therapy Unit CIMA, Foundation for Applied Medical Research, University of Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Spain.
| | - Javier Sánchez-Céspedes
- Institute of Biomedicine of Seville (IBiS), University Hospital Virgen del Rocío/CSIC/University of Seville, Clinical Unit of Infectious Diseases, Microbiology, and Preventive Medicine, Seville, Spain; Department of Medicine, University of Seville, Seville, Spain.
| |
Collapse
|
23
|
Imaging, Tracking and Computational Analyses of Virus Entry and Egress with the Cytoskeleton. Viruses 2018; 10:v10040166. [PMID: 29614729 PMCID: PMC5923460 DOI: 10.3390/v10040166] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 03/27/2018] [Accepted: 03/28/2018] [Indexed: 12/27/2022] Open
Abstract
Viruses have a dual nature: particles are “passive substances” lacking chemical energy transformation, whereas infected cells are “active substances” turning-over energy. How passive viral substances convert to active substances, comprising viral replication and assembly compartments has been of intense interest to virologists, cell and molecular biologists and immunologists. Infection starts with virus entry into a susceptible cell and delivers the viral genome to the replication site. This is a multi-step process, and involves the cytoskeleton and associated motor proteins. Likewise, the egress of progeny virus particles from the replication site to the extracellular space is enhanced by the cytoskeleton and associated motor proteins. This overcomes the limitation of thermal diffusion, and transports virions and virion components, often in association with cellular organelles. This review explores how the analysis of viral trajectories informs about mechanisms of infection. We discuss the methodology enabling researchers to visualize single virions in cells by fluorescence imaging and tracking. Virus visualization and tracking are increasingly enhanced by computational analyses of virus trajectories as well as in silico modeling. Combined approaches reveal previously unrecognized features of virus-infected cells. Using select examples of complementary methodology, we highlight the role of actin filaments and microtubules, and their associated motors in virus infections. In-depth studies of single virion dynamics at high temporal and spatial resolutions thereby provide deep insight into virus infection processes, and are a basis for uncovering underlying mechanisms of how cells function.
Collapse
|
24
|
Dalmau-Mena I, Del Pino P, Pelaz B, Cuesta-Geijo MÁ, Galindo I, Moros M, de la Fuente JM, Alonso C. Nanoparticles engineered to bind cellular motors for efficient delivery. J Nanobiotechnology 2018; 16:33. [PMID: 29602307 PMCID: PMC5877387 DOI: 10.1186/s12951-018-0354-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 03/19/2018] [Indexed: 11/17/2022] Open
Abstract
Background Dynein is a cytoskeletal molecular motor protein that transports cellular cargoes along microtubules. Biomimetic synthetic peptides designed to bind dynein have been shown to acquire dynamic properties such as cell accumulation and active intra- and inter-cellular motion through cell-to-cell contacts and projections to distant cells. On the basis of these properties dynein-binding peptides could be used to functionalize nanoparticles for drug delivery applications. Results Here, we show that gold nanoparticles modified with dynein-binding delivery sequences become mobile, powered by molecular motor proteins. Modified nanoparticles showed dynamic properties, such as travelling the cytosol, crossing intracellular barriers and shuttling the nuclear membrane. Furthermore, nanoparticles were transported from one cell to another through cell-to-cell contacts and quickly spread to distant cells through cell projections. Conclusions The capacity of these motor-bound nanoparticles to spread to many cells and increasing cellular retention, thus avoiding losses and allowing lower dosage, could make them candidate carriers for drug delivery. Electronic supplementary material The online version of this article (10.1186/s12951-018-0354-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Inmaculada Dalmau-Mena
- Dpt. Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Carretera de la Coruña km 7.5, 28040, Madrid, Spain
| | - Pablo Del Pino
- Instituto de Nanociencia de Aragón, Universidad de Zaragoza, Mariano Esquillor, s/n, 50018, Zaragoza, Spain.,Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Física de Partículas, Universidad de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Beatriz Pelaz
- Instituto de Nanociencia de Aragón, Universidad de Zaragoza, Mariano Esquillor, s/n, 50018, Zaragoza, Spain.,Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS), Departamento de Física de Partículas, Universidad de Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Miguel Ángel Cuesta-Geijo
- Dpt. Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Carretera de la Coruña km 7.5, 28040, Madrid, Spain
| | - Inmaculada Galindo
- Dpt. Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Carretera de la Coruña km 7.5, 28040, Madrid, Spain
| | - María Moros
- Instituto de Nanociencia de Aragón, Universidad de Zaragoza, Mariano Esquillor, s/n, 50018, Zaragoza, Spain
| | - Jesús M de la Fuente
- Aragon Materials Science Institute (ICMA), CSIC-University of Zaragoza and CIBER-BBN, C/Pedro Cerbuna 12, 50009, Zaragoza, Spain
| | - Covadonga Alonso
- Dpt. Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Carretera de la Coruña km 7.5, 28040, Madrid, Spain.
| |
Collapse
|
25
|
Guissoni ACP, Soares CMA, Badr KR, Ficcadori FS, Parente AFA, Parente JA, Baeza LC, Souza M, Cardoso DDDDP. Proteomic analysis of A-549 cells infected with human adenovirus 40 by LC-MS. Virus Genes 2018; 54:351-360. [PMID: 29546667 DOI: 10.1007/s11262-018-1554-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 03/12/2018] [Indexed: 12/17/2022]
Abstract
Human Adenoviruses (HAdVs) are etiological agents of different syndromes such as gastroenteritis, cystitis, ocular, and respiratory diseases, and infection by these viruses may cause alterations in cellular homeostasis. The objective of the study was the proteomic analysis of A-549 cells infected with HAdV-40 using LC-MS. At 30 h of infection, the quantitative analysis revealed 336 differentially expressed proteins. From them, 206 were induced (up-regulated) and 130 were suppressed (down-regulated). The majority of up-regulated proteins were related to energy, cellular organization, stress response, and apoptosis pathways. It was observed alteration of cell metabolism with increase of the glycolytic pathway, β-oxidation, and respiratory chain. Also, the results suggest cytoskeleton reorganization and apoptosis induction. The data can improve knowledge about the replication of HAdV-40 in cell culture considering the proteins related to distinct metabolic pathways induced by viral infection in A-549 cells.
Collapse
Affiliation(s)
- Ana Carla Peixoto Guissoni
- Human Virology Laboratory, Institute of Tropical Pathology and Public Health, Federal University of Goias, Rua 235, S/N, Sala 418, Setor Universitário, Goiania, Goias, 74605050, Brazil
| | - Célia Maria Almeida Soares
- Molecular Biology Laboratory, Institute of Biological Sciences, Federal University of Goias, Goiania, Goias, Brazil
| | - Kareem R Badr
- Human Virology Laboratory, Institute of Tropical Pathology and Public Health, Federal University of Goias, Rua 235, S/N, Sala 418, Setor Universitário, Goiania, Goias, 74605050, Brazil
| | - Fabiola Sousa Ficcadori
- Human Virology Laboratory, Institute of Tropical Pathology and Public Health, Federal University of Goias, Rua 235, S/N, Sala 418, Setor Universitário, Goiania, Goias, 74605050, Brazil
| | - Ana Flávia Alves Parente
- Molecular Biology Laboratory, Institute of Biological Sciences, Federal University of Goias, Goiania, Goias, Brazil
| | - Juliana Alves Parente
- Molecular Biology Laboratory, Institute of Biological Sciences, Federal University of Goias, Goiania, Goias, Brazil
| | - Lilian Cristina Baeza
- Molecular Biology Laboratory, Institute of Biological Sciences, Federal University of Goias, Goiania, Goias, Brazil
| | - Menira Souza
- Human Virology Laboratory, Institute of Tropical Pathology and Public Health, Federal University of Goias, Rua 235, S/N, Sala 418, Setor Universitário, Goiania, Goias, 74605050, Brazil
| | - Divina das Dores de Paula Cardoso
- Human Virology Laboratory, Institute of Tropical Pathology and Public Health, Federal University of Goias, Rua 235, S/N, Sala 418, Setor Universitário, Goiania, Goias, 74605050, Brazil.
| |
Collapse
|
26
|
Milev MP, Yao X, Berthoux L, Mouland AJ. Impacts of virus-mediated manipulation of host Dynein. DYNEINS 2018. [PMCID: PMC7150161 DOI: 10.1016/b978-0-12-809470-9.00010-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In general viruses' modus operandi to propagate is achieved by the co-opting host cell components, membranes, proteins, and machineries to their advantage. This is true for virtually every aspect of a virus' replication cycle from virus entry to the budding or release of progeny virus particles. In this chapter, we will discuss new information on the impacts of virus-mediated manipulation of Dynein motor complexes and associated machineries and factors. We will highlight how these host cell components impact on pathogenicity and immune responses, as many of the virus-mediated hijacked components also play pivotal roles in immune responses to pathogen insult. There are several comprehensive reviews that define virus–Dynein interactions including the first edition of this book that describes how viruses manipulate the host cell machineries their advantage. An updated table is included to summarize these virus–host interactions. Notably, barriers to intracellular translocation represent major hurdles to viral components during de novo infection and during active replication and the generation of progeny virus particles. Clearly, the subversion of host cell molecular motor protein activities takes advantage of constitutive and regulated membrane trafficking events and will target virus components to intracytoplasmic locales and membrane assembly. Broadening our understanding of the interplay between viruses, Dynein and the cytoskeleton will likely inform on new types of therapies. Continual enhancement of the breadth of new information on how viruses manipulate host cell biology will inevitably aid in the identification of new targets that can be poisoned to block old, new, and emerging viruses alike in their tracks.
Collapse
|
27
|
Yang M, Yang CS, Guo W, Tang J, Huang Q, Feng S, Jiang A, Xu X, Jiang G, Liu YQ. A novel fiber chimeric conditionally replicative adenovirus-Ad5/F35 for tumor therapy. Cancer Biol Ther 2017; 18:833-840. [PMID: 29144842 DOI: 10.1080/15384047.2017.1395115] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Significant progress has been made in the diagnosis and treatment of cancer; however, significant challenges remain. Conditionally replicating adenoviruses (CRAds), which not only kill cancer cells, but also serve as vectors to express therapeutic genes, are a novel and effective method to treat cancer. However, most adenoviruses are Ad5, which infect cells through the coxsackie and adenovirus receptor (CAR). The transduction efficacy of Ad5 is restricted because of the absent or low expression of CAR on several cancer cells. Ad serotype 35 has a different tropism pattern to Ad5. Ad35 attaches to cells via a non-CAR receptor, CD46, which is expressed widely on most tumor cells. Thus, chimeric adenoviral vectors consisting of the knob and shaft of Ad35 combined with Ad5 have been constructed. The chimeric fiber adenoviral vectors can transduce CAR-positive and CAR-negative cell lines. In this review, we explore the application of the novel fiber chimeric conditionally replicative adenovirus-Ad5/F35 in tumor therapy in terms of safety, mechanism, transduction efficacy, and antitumor effect.
Collapse
Affiliation(s)
- Ming Yang
- a Department of Radiotherapy , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China.,b Department of Oncology , Affiliated Nanyang Second General Hospital , Nanyang , China
| | - Chun Sheng Yang
- c Department of Dermatology , Affiliated Huai'an Hospital of Xuzhou Medical University , the Second People's Hospital of Huai'an, Huai'an , China
| | - WenWen Guo
- a Department of Radiotherapy , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China
| | - JianQin Tang
- d Department of Dermatology , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China
| | - Qian Huang
- a Department of Radiotherapy , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China
| | - ShouXin Feng
- a Department of Radiotherapy , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China
| | - AiJun Jiang
- a Department of Radiotherapy , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China
| | - XiFeng Xu
- a Department of Radiotherapy , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China
| | - Guan Jiang
- d Department of Dermatology , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China
| | - Yan Qun Liu
- d Department of Dermatology , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China
| |
Collapse
|
28
|
Wang IH, Burckhardt CJ, Yakimovich A, Morf MK, Greber UF. The nuclear export factor CRM1 controls juxta-nuclear microtubule-dependent virus transport. J Cell Sci 2017; 130:2185-2195. [PMID: 28515232 DOI: 10.1242/jcs.203794] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 05/12/2017] [Indexed: 12/26/2022] Open
Abstract
Transport of large cargo through the cytoplasm requires motor proteins and polarized filaments. Viruses that replicate in the nucleus of post-mitotic cells use microtubules and the dynein-dynactin motor to traffic to the nuclear membrane and deliver their genome through nuclear pore complexes (NPCs) into the nucleus. How virus particles (virions) or cellular cargo are transferred from microtubules to the NPC is unknown. Here, we analyzed trafficking of incoming cytoplasmic adenoviruses by single-particle tracking and super-resolution microscopy. We provide evidence for a regulatory role of CRM1 (chromosome-region-maintenance-1; also known as XPO1, exportin-1) in juxta-nuclear microtubule-dependent adenovirus transport. Leptomycin B (LMB) abolishes nuclear targeting of adenovirus. It binds to CRM1, precludes CRM1-cargo binding and blocks signal-dependent nuclear export. LMB-inhibited CRM1 did not compete with adenovirus for binding to the nucleoporin Nup214 at the NPC. Instead, CRM1 inhibition selectively enhanced virion association with microtubules, and boosted virion motions on microtubules less than ∼2 µm from the nuclear membrane. The data show that the nucleus provides positional information for incoming virions to detach from microtubules, engage a slower microtubule-independent motility to the NPC and enhance infection.
Collapse
Affiliation(s)
- I-Hsuan Wang
- Department of Molecular Life Sciences, University of Zürich, 8057 Zurich, Switzerland
| | - Christoph J Burckhardt
- Department of Molecular Life Sciences, University of Zürich, 8057 Zurich, Switzerland
- Department of Bioinformatics, University of Texas Southwestern Medical Center at Dallas, Dallas, TX 75390, USA
| | - Artur Yakimovich
- Department of Molecular Life Sciences, University of Zürich, 8057 Zurich, Switzerland
| | - Matthias K Morf
- Department of Molecular Life Sciences, University of Zürich, 8057 Zurich, Switzerland
- Molecular Life Sciences Graduate School, ETH and University of Zürich, 8057 Zurich, Switzerland
| | - Urs F Greber
- Department of Molecular Life Sciences, University of Zürich, 8057 Zurich, Switzerland
| |
Collapse
|
29
|
Basu R, Bose A, Thomas D, Das Sarma J. Microtubule-assisted altered trafficking of astrocytic gap junction protein connexin 43 is associated with depletion of connexin 47 during mouse hepatitis virus infection. J Biol Chem 2017; 292:14747-14763. [PMID: 28566289 DOI: 10.1074/jbc.m117.786491] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 05/16/2017] [Indexed: 11/06/2022] Open
Abstract
Gap junctions (GJs) are important for maintenance of CNS homeostasis. GJ proteins, connexin 43 (Cx43) and connexin 47 (Cx47), play a crucial role in production and maintenance of CNS myelin. Cx43 is mainly expressed by astrocytes in the CNS and forms gap junction intercellular communications between astrocytes-astrocytes (Cx43-Cx43) and between astrocytes-oligodendrocytes (Cx43-Cx47). Mutations of these connexin (Cx) proteins cause dysmyelinating diseases in humans. Previously, it has been shown that Cx43 localization and expression is altered due to mouse hepatitis virus (MHV)-A59 infection both in vivo and in vitro; however, its mechanism and association with loss of myelin protein was not elaborated. Thus, we explored potential mechanisms by which MHV-A59 infection alters Cx43 localization and examined the effects of viral infection on Cx47 expression and its association with loss of the myelin marker proteolipid protein. Immunofluorescence and total internal reflection fluorescence microscopy confirmed that MHV-A59 used microtubules (MTs) as a conduit to reach the cell surface and restricted MT-mediated Cx43 delivery to the cell membrane. Co-immunoprecipitation experiments demonstrated that Cx43-β-tubulin molecular interaction was depleted due to protein-protein interaction between viral particles and MTs. During acute MHV-A59 infection, oligodendrocytic Cx47, which is mainly stabilized by Cx43 in vivo, was down-regulated, and its characteristic staining remained disrupted even at chronic phase. The loss of Cx47 was associated with loss of proteolipid protein at the chronic stage of MHV-A59 infection.
Collapse
Affiliation(s)
- Rahul Basu
- From the Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur-741246, India
| | - Abhishek Bose
- From the Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur-741246, India
| | - Deepthi Thomas
- From the Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur-741246, India
| | - Jayasri Das Sarma
- From the Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur-741246, India
| |
Collapse
|
30
|
Portilho DM, Persson R, Arhel N. Role of non-motile microtubule-associated proteins in virus trafficking. Biomol Concepts 2017; 7:283-292. [PMID: 27879481 DOI: 10.1515/bmc-2016-0018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 11/04/2016] [Indexed: 11/15/2022] Open
Abstract
Viruses are entirely dependent on their ability to infect a host cell in order to replicate. To reach their site of replication as rapidly and efficiently as possible following cell entry, many have evolved elaborate mechanisms to hijack the cellular transport machinery to propel themselves across the cytoplasm. Long-range movements have been shown to involve motor proteins along microtubules (MTs) and direct interactions between viral proteins and dynein and/or kinesin motors have been well described. Although less well-characterized, it is also becoming increasingly clear that non-motile microtubule-associated proteins (MAPs), including structural MAPs of the MAP1 and MAP2 families, and microtubule plus-end tracking proteins (+TIPs), can also promote viral trafficking in infected cells, by mediating interaction of viruses with filaments and/or motor proteins, and modulating filament stability. Here we review our current knowledge on non-motile MAPs, their role in the regulation of cytoskeletal dynamics and in viral trafficking during the early steps of infection.
Collapse
|
31
|
Ni R, Zhou J, Hossain N, Chau Y. Virus-inspired nucleic acid delivery system: Linking virus and viral mimicry. Adv Drug Deliv Rev 2016; 106:3-26. [PMID: 27473931 DOI: 10.1016/j.addr.2016.07.005] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 07/02/2016] [Accepted: 07/20/2016] [Indexed: 12/21/2022]
Abstract
Targeted delivery of nucleic acids into disease sites of human body has been attempted for decades, but both viral and non-viral vectors are yet to meet our expectations. Safety concerns and low delivery efficiency are the main limitations of viral and non-viral vectors, respectively. The structure of viruses is both ordered and dynamic, and is believed to be the key for effective transfection. Detailed understanding of the physical properties of viruses, their interaction with cellular components, and responses towards cellular environments leading to transfection would inspire the development of safe and effective non-viral vectors. To this goal, this review systematically summarizes distinctive features of viruses that are implied for efficient nucleic acid delivery but not yet fully explored in current non-viral vectors. The assembly and disassembly of viral structures, presentation of viral ligands, and the subcellular targeting of viruses are emphasized. Moreover, we describe the current development of cationic material-based viral mimicry (CVM) and structural viral mimicry (SVM) in these aspects. In light of the discrepancy, we identify future opportunities for rational design of viral mimics for the efficient delivery of DNA and RNA.
Collapse
Affiliation(s)
- Rong Ni
- Division of Biomedical Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; Institute for Advanced Study, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Junli Zhou
- Department of Chemical and Biomolecular Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Naushad Hossain
- Division of Biomedical Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Ying Chau
- Department of Chemical and Biomolecular Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China; Division of Biomedical Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.
| |
Collapse
|
32
|
Targeting Motor End Plates for Delivery of Adenoviruses: An Approach to Maximize Uptake and Transduction of Spinal Cord Motor Neurons. Sci Rep 2016; 6:33058. [PMID: 27619631 PMCID: PMC5020496 DOI: 10.1038/srep33058] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 08/08/2016] [Indexed: 02/07/2023] Open
Abstract
Gene therapy can take advantage of the skeletal muscles/motor neurons anatomical relationship to restrict gene expression to the spinal cord ventral horn. Furthermore, recombinant adenoviruses are attractive viral-vectors as they permit spatial and temporal modulation of transgene expression. In the literature, however, several inconsistencies exist with regard to the intramuscular delivery parameters of adenoviruses. The present study is an evaluation of the optimal injection sites on skeletal muscle, time course of expression and mice’s age for maximum transgene expression in motor neurons. Targeting motor end plates yielded a 2.5-fold increase in the number of transduced motor neurons compared to injections performed away from this region. Peak adenoviral transgene expression in motor neurons was detected after seven days. Further, greater numbers of transduced motor neurons were found in juvenile (3–7 week old) mice as compared with adults (8+ weeks old). Adenoviral injections produced robust transgene expression in motor neurons and skeletal myofibres. In addition, dendrites of transduced motor neurons were shown to extend well into the white matter where the descending motor pathways are located. These results also provide evidence that intramuscular delivery of adenovirus can be a suitable gene therapy approach to treat spinal cord injury.
Collapse
|
33
|
Rüdiger AT, Mayrhofer P, Ma-Lauer Y, Pohlentz G, Müthing J, von Brunn A, Schwegmann-Weßels C. Tubulins interact with porcine and human S proteins of the genus Alphacoronavirus and support successful assembly and release of infectious viral particles. Virology 2016; 497:185-197. [PMID: 27479465 PMCID: PMC7111311 DOI: 10.1016/j.virol.2016.07.022] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 07/16/2016] [Accepted: 07/18/2016] [Indexed: 01/05/2023]
Abstract
Coronavirus spike proteins mediate host-cell-attachment and virus entry. Virus replication takes place within the host cell cytosol, whereas assembly and budding occur at the endoplasmic reticulum-Golgi intermediate compartment. In this study we demonstrated that the last 39 amino acid stretches of Alphacoronavirus spike cytoplasmic domains of the human coronavirus 229E, NL63, and the porcine transmissible gastroenteritis virus TGEV interact with tubulin alpha and beta chains. In addition, a partial co-localization of TGEV spike proteins with authentic host cell β-tubulin was observed. Furthermore, drug-induced microtubule depolymerization led to changes in spike protein distribution, a reduction in the release of infectious virus particles and less amount of spike protein incorporated into virions. These data demonstrate that interaction of Alphacoronavirus spike proteins with tubulin supports S protein transport and incorporation into virus particles. The cytoplasmic domain of coronavirus S proteins interacts with tubulin. Microtubule depolymerization influences S protein distribution. Viral titers are reduced after microtubule depolymerization. S protein incorporation into virus particles depends on intact microtubule.
Collapse
Affiliation(s)
- Anna-Theresa Rüdiger
- Institute of Virology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hannover, Germany
| | - Peter Mayrhofer
- Virology Department, Max-von-Pettenkofer Institute, Ludwig-Maximilians University Munich, Pettenkoferstraße 9a, 80336 Munich, Germany
| | - Yue Ma-Lauer
- Virology Department, Max-von-Pettenkofer Institute, Ludwig-Maximilians University Munich, Pettenkoferstraße 9a, 80336 Munich, Germany
| | - Gottfried Pohlentz
- Institute for Hygiene, University of Münster, Robert-Koch-Straße 41, 48149 Münster, Germany
| | - Johannes Müthing
- Institute for Hygiene, University of Münster, Robert-Koch-Straße 41, 48149 Münster, Germany
| | - Albrecht von Brunn
- Virology Department, Max-von-Pettenkofer Institute, Ludwig-Maximilians University Munich, Pettenkoferstraße 9a, 80336 Munich, Germany; German Centers for Infection Research (DZIF), Ludwig-Maximilians-University Munich, Germany.
| | - Christel Schwegmann-Weßels
- Institute of Virology, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hannover, Germany.
| |
Collapse
|
34
|
Tosolini AP, Morris R. Viral-mediated gene therapy for spinal cord injury (SCI) from a translational neuroanatomical perspective. Neural Regen Res 2016; 11:743-4. [PMID: 27335556 PMCID: PMC4904463 DOI: 10.4103/1673-5374.182698] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Andrew P Tosolini
- Translational Neuroscience Facility, School of Medical Sciences, the University of New South Wales (UNSW Australia), Sydney, Australia; Current address for Tosolini AP: Sobell Department of Motor Neuroscience and Movement Disorders, Institute of Neurology, University College London, London, UK
| | - Renée Morris
- Translational Neuroscience Facility, School of Medical Sciences, the University of New South Wales (UNSW Australia), Sydney, Australia
| |
Collapse
|
35
|
Abstract
ABSTRACT Viruses are a diverse class of nanoparticles. However, they have evolved a few common mechanisms that enable successful infection of their host cells. The first stage of this process involves entry into the cell. For enveloped viruses this process has been well characterized. For nonenveloped viruses, the focus of this review, the entry mechanisms are less well understood. For these viruses, a typical pathway involves receptor attachment followed by internalization into cellular vesicles and subsequent viral escape to the cytosol and transport to the site of genome replication. Significantly, these viruses have evolved numerous mechanisms to fulfill this seemingly simple infection scheme. We focus on the latest observations for several families of nonenveloped viruses and highlight specific members for eukaryotic families: Adenoviridae, Papillomaviridae, Parvoviridae, Picornaviridae, Polyomaviridae and Reoviridae; and prokaryotic families: Microviridae, Myoviridae, Podoviridae and Siphoviridae.
Collapse
Affiliation(s)
- Bridget Lins
- Department of Biochemistry & Molecular Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Mavis Agbandje-McKenna
- Department of Biochemistry & Molecular Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
36
|
Lopez-Gordo E, Denby L, Nicklin SA, Baker AH. The importance of coagulation factors binding to adenovirus: historical perspectives and implications for gene delivery. Expert Opin Drug Deliv 2014; 11:1795-813. [DOI: 10.1517/17425247.2014.938637] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Estrella Lopez-Gordo
- University of Glasgow, Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, 126 University Place, Glasgow G12 8TA, UK
| | - Laura Denby
- University of Glasgow, Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, 126 University Place, Glasgow G12 8TA, UK
| | - Stuart A Nicklin
- University of Glasgow, Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, 126 University Place, Glasgow G12 8TA, UK
| | - Andrew H Baker
- University of Glasgow, Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, 126 University Place, Glasgow G12 8TA, UK ;
| |
Collapse
|
37
|
Cohen O, Granek R. Nucleus-targeted drug delivery: theoretical optimization of nanoparticles decoration for enhanced intracellular active transport. NANO LETTERS 2014; 14:2515-2521. [PMID: 24646130 DOI: 10.1021/nl500248q] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
A rational design for a nanoparticle is suggested, which will maximize its arrival efficiency from the plasma membrane to the nuclear surrounding. The design is based on grafting the particle surface with polymer spacers, each ending with a motor protein associating molecule, for example, nuclear localization signal peptide. It is theoretically shown that the spacer polymer molecular weight can be adjusted to significantly increase the effective particle processivity time. This should lead to appreciable enhancement of active transport of the nanocarrier, and consequently drug delivery, to the nucleus.
Collapse
Affiliation(s)
- Ohad Cohen
- The Stella and Avram Goren-Goldstein Department of Biotechnology Engineering and ‡The Ilse Katz Institute for Meso and Nanoscale Science and Technology, Ben-Gurion University of the Negev , Beer Sheva 84105, Israel
| | | |
Collapse
|
38
|
Lyi SM, Tan MJA, Parrish CR. Parvovirus particles and movement in the cellular cytoplasm and effects of the cytoskeleton. Virology 2014; 456-457:342-52. [PMID: 24889253 DOI: 10.1016/j.virol.2014.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2014] [Revised: 04/03/2014] [Accepted: 04/03/2014] [Indexed: 12/20/2022]
Abstract
Cell infection by parvoviruses requires that capsids be delivered from outside the cell to the cytoplasm, followed by genome trafficking to the nucleus. Here we microinject capsids into cells that lack receptors and followed their movements within the cell over time. In general the capsids remained close to the positions where they were injected, and most particles did not move to the vicinity of or enter the nucleus. When 70 kDa-dextran was injected along with the capsids that did not enter the nucleus in significant amounts. Capsids conjugated to peptides containing the SV40 large T-antigen nuclear localization signal remained in the cytoplasm, although bovine serum albumen conjugated to the same peptide entered the nucleus rapidly. No effects of disruption of microfilaments, intermediate filaments, or microtubules on the distribution of the capsids were observed. These results suggest that movement of intact capsids within cells is primarily associated with passive processes.
Collapse
Affiliation(s)
- Sangbom Michael Lyi
- Baker Institute for Animal Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, United States
| | - Min Jie Alvin Tan
- Baker Institute for Animal Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, United States.
| | - Colin R Parrish
- Baker Institute for Animal Health, Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, United States.
| |
Collapse
|
39
|
Pigeon L, Gonçalves C, Gosset D, Pichon C, Midoux P. An E3-14.7K peptide that promotes microtubules-mediated transport of plasmid DNA increases polyplexes transfection efficiency. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2013; 9:3845-3851. [PMID: 23661626 DOI: 10.1002/smll.201300217] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Indexed: 06/02/2023]
Abstract
Chemical vectors as cationic polymers and cationic lipids are promising alternatives to viral vectors for gene therapy. Beside endosome escape and nuclear import, plasmid DNA (pDNA) migration in the cytosol toward the nuclear envelope is also regarded as a limiting step for efficient DNA transfection with non-viral vectors. Here, the interaction between E3-14.7K and FIP-1 to favor migration of pDNA along microtubules is exploited. E3-14.7K is an early protein of human adenoviruses that interacts via FIP-1 (Fourteen.7K Interacting Protein 1) protein with the light-chain components of the human microtubule motor protein dynein (TCTEL1). This peptide is conjugated with pDNA and mediates interaction of pDNA in vitro with isolated microtubules as well as with microtubules in cellulo. Videomicroscopy and tracking treatment of images clearly demonstrate that P79-98/pDNA conjugate exhibits a linear transport with large amplitude along microtubules upon 2 h transfection with polyplexes whereas control pDNA conjugate exhibits small non-directional movements in the cytoplasm. Remarkably, P79-98/peGFP polyplexes enhance by a factor 2.5 (up to 76%) the number of transfected cells. The results demonstrate, for the first time, that the transfection efficiency of polyplexes can be drastically increased when the microtubules migration of pDNA is facilitated by a peptide allowing pDNA docking to TCTEL1. This is a real breakthrough in the non viral gene delivery field that opens hope to build artificial viruses.
Collapse
Affiliation(s)
- Lucie Pigeon
- Centre de Biophysique Moléculaire, CNRS UPR4301, Inserm and University of Orléans, 45071 Orléans cedex 02, France
| | | | | | | | | |
Collapse
|
40
|
Cianciola NL, Greene DJ, Morton RE, Carlin CR. Adenovirus RIDα uncovers a novel pathway requiring ORP1L for lipid droplet formation independent of NPC1. Mol Biol Cell 2013; 24:3309-25. [PMID: 24025716 PMCID: PMC3814149 DOI: 10.1091/mbc.e12-10-0760] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Expression of the adenovirus protein RIDα rescues the cholesterol storage phenotype in NPC1-deficient cells by inducing formation of lipid droplets. The function of RIDα is independent of NPC1 but dependent on NPC2 and the oxysterol-binding protein ORP1L. This study provides the first evidence that ORP1L plays a role in sterol transport and LD formation. Niemann–Pick disease type C (NPC) is caused by mutations in NPC1 or NPC2, which coordinate egress of low-density-lipoprotein (LDL)-cholesterol from late endosomes. We previously reported that the adenovirus-encoded protein RIDα rescues the cholesterol storage phenotype in NPC1-mutant fibroblasts. We show here that RIDα reconstitutes deficient endosome-to-endoplasmic reticulum (ER) transport, allowing excess LDL-cholesterol to be esterified by acyl-CoA:cholesterol acyltransferase and stored in lipid droplets (LDs) in NPC1-deficient cells. Furthermore, the RIDα pathway is regulated by the oxysterol-binding protein ORP1L. Studies have classified ORP1L as a sterol sensor involved in LE positioning downstream of GTP-Rab7. Our data, however, suggest that ORP1L may play a role in transport of LDL-cholesterol to a specific ER pool designated for LD formation. In contrast to NPC1, which is dispensable, the RIDα/ORP1L-dependent route requires functional NPC2. Although NPC1/NPC2 constitutes the major pathway, therapies that amplify minor egress routes for LDL-cholesterol could significantly improve clinical management of patients with loss-of-function NPC1 mutations. The molecular identity of putative alternative pathways, however, is poorly characterized. We propose RIDα as a model system for understanding physiological egress routes that use ORP1L to activate ER feedback responses involved in LD formation.
Collapse
Affiliation(s)
- Nicholas L Cianciola
- Department of Molecular Biology and Microbiology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106 Department of Cell Biology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195 Case Comprehensive Cancer Center, School of Medicine, Case Western Reserve University, Cleveland, OH 44106
| | | | | | | |
Collapse
|
41
|
Abstract
The outer shell of the adenovirus capsid comprises three major types of protein (hexon, penton base and fiber) that perform the majority of functions facilitating the early stages of adenovirus infection. These stages include initial cell-surface binding followed by receptor-mediated endocytosis, endosomal penetration and cytosolic entry, and intracellular trafficking toward the nucleus. Numerous studies have shown that the penton base contributes to several of these steps and have supported the development of this protein into a delivery agent for therapeutic molecules. Studies revealing that the fiber and hexon bear unexpected properties of cell entry and/or nuclear homing have supported the development of these capsid proteins, as well into potential delivery vehicles. This review summarizes the findings to date of the protein-cell activities of these capsid proteins in the absence of the whole virus and their potential for therapeutic application with regard to the delivery of foreign molecules.
Collapse
|
42
|
Eichwald C, Arnoldi F, Laimbacher AS, Schraner EM, Fraefel C, Wild P, Burrone OR, Ackermann M. Rotavirus viroplasm fusion and perinuclear localization are dynamic processes requiring stabilized microtubules. PLoS One 2012; 7:e47947. [PMID: 23110139 PMCID: PMC3479128 DOI: 10.1371/journal.pone.0047947] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Accepted: 09/17/2012] [Indexed: 12/21/2022] Open
Abstract
Rotavirus viroplasms are cytosolic, electron-dense inclusions corresponding to the viral machinery of replication responsible for viral template transcription, dsRNA genome segments replication and assembly of new viral cores. We have previously observed that, over time, those viroplasms increase in size and decrease in number. Therefore, we hypothesized that this process was dependent on the cellular microtubular network and its associated dynamic components. Here, we present evidence demonstrating that viroplasms are dynamic structures, which, in the course of an ongoing infection, move towards the perinuclear region of the cell, where they fuse among each other, thereby gaining considerably in size and, simultaneouly, explaining the decrease in numbers. On the viral side, this process seems to depend on VP2 for movement and on NSP2 for fusion. On the cellular side, both the temporal transition and the maintenance of the viroplasms are dependent on the microtubular network, its stabilization by acetylation, and, surprisingly, on a kinesin motor of the kinesin-5 family, Eg5. Thus, we provide for the first time deeper insights into the dynamics of rotavirus replication, which can explain the behavior of viroplasms in the infected cell.
Collapse
|
43
|
Reduced infectivity of adenovirus type 5 particles and degradation of entering viral genomes associated with incomplete processing of the preterminal protein. J Virol 2012; 86:13554-65. [PMID: 23035217 DOI: 10.1128/jvi.02337-12] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
To investigate further the contribution of the adenovirus type 5 (Ad5) E1B 55-kDa protein to genome replication, viral DNA accumulation was examined in primary human fibroblasts and epithelial cells infected with Ad5 or the E1B 55-kDa-null mutant Hr6. Unexpectedly, all cell types were observed to contain a significantly higher concentration of entering Hr6 than of Ad5 DNA, as did an infectious unit of Hr6. However, the great majority of the Hr6 genomes were degraded soon after entry. As this unusual phenotype cannot be ascribed to the Hr6 E1B frameshift mutation (J. S. Chahal and S. J. Flint, J. Virol. 86:3064-3072, 2012), the sequences of the Ad5 and Hr6 genomes were compared by using high-throughput sequencing. Seven previously unrecognized mutations were identified in the Hr6 genome, two of which result in substitutions in virion proteins, G315V in the preterminal protein (preTP) and A406V in fiber protein IV. Previous observations and the visualization by immunofluorescence of greater numbers of viral genomes entering the cytosol of Hr6-infected cells than of Ad5-infected cells indicated that the fiber mutation could not be responsible for the low-infectivity phenotype of Hr6. However, comparison of the forms of terminal protein present in purified virus particles indicated that the production of mature terminal protein from a processing intermediate is impaired in Hr6 particles. We therefore propose that complete processing of preTP within virus particles is necessary for the ability of viral genomes to become localized at appropriate sites and persist in infected cells.
Collapse
|
44
|
Functional characterisation of the WW minimal domain for delivering therapeutic proteins by adenovirus dodecahedron. PLoS One 2012; 7:e45416. [PMID: 23028993 PMCID: PMC3459938 DOI: 10.1371/journal.pone.0045416] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2012] [Accepted: 08/16/2012] [Indexed: 12/16/2022] Open
Abstract
Protein transduction offers a great therapeutic potential by efficient delivery of biologically active cargo into cells. The Adenovirus Dd (Dodecahedron) has recently been shown to deliver proteins fused to the tandem WW2-3-4 structural domains from the E3 ubiquitin ligase Nedd4. In this study, we conclusively show that Dd is able to efficiently deliver cargo inside living cells, which mainly localize in fast moving endocytic vesicles, supporting active transport along the cytoskeleton. We further improve this delivery system by expressing a panel of 13 WW-GFP mutant forms to characterize their binding properties towards Dd. We identified the domain WW3 and its mutant form WW3_10_13 to be sufficient for optimal binding to Dd. We greatly minimise the interacting WW modules from 20 to 6 kDa without compromising its efficient delivery by Dd. Using these minimal WW domains fused to the tumor suppressor p53 protein, we show efficient cellular uptake and distribution into cancer cells, leading to specific induction of apoptosis in these cells. Taken together, these findings represent a step further towards the development of a Dd-based delivery system for future therapeutic application.
Collapse
|
45
|
Badding MA, Dean DA. Highly acetylated tubulin permits enhanced interactions with and trafficking of plasmids along microtubules. Gene Ther 2012; 20:616-24. [PMID: 23013836 PMCID: PMC3587030 DOI: 10.1038/gt.2012.77] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Microtubule-based transport is required for plasmid translocation to the nucleus during transfections, and having stable structures could enhance this movement. In previous studies in which the cytoskeleton was disrupted, we found that populations of microtubules remain that are stable and highly acetylated. By increasing the levels of acetylated tubulin through inhibition of the tubulin deacetylase HDAC6, we observe more rapid plasmid nuclear localization of transfected plasmids and greater levels of gene transfer. In this study, we sought to understand plasmid movement in cells with enhanced tubulin acetylation. Using variations of a microtubule spin down assay, we found that plasmids bound to hyper-acetylated microtubules to a greater degree than they did to unmodified microtubules. To determine if microtubule acetylation also affects cytoplasmic trafficking, plasmid movement was evaluated in real time by particle tracking in cells with varying levels of acetylated microtubules. We found that plasmids display greater net rates of movement, spend more time in productive motion and display longer runs of continuous motion in cells with highly acetylated microtubules compared to those with fewer modifications. These results all suggest that plasmid movement is enhanced along highly acetylated microtubules, reducing the time spent in the cytoplasm prior to nuclear import. Taken together, these findings provide a foundation for determining how modulation of microtubule acetylation can be used as a means to increase intracellular trafficking of plasmids and enhance gene therapy.
Collapse
Affiliation(s)
- M A Badding
- Department of Pediatrics, Division of Neonatology, School of Medicine and Dentistry, University of Rochester, Rochester, NY 14642, USA
| | | |
Collapse
|
46
|
Zeng Q, Han J, Zhao D, Gong T, Zhang Z, Sun X. Protection of adenovirus from neutralizing antibody by cationic PEG derivative ionically linked to adenovirus. Int J Nanomedicine 2012; 7:985-97. [PMID: 22412299 PMCID: PMC3299205 DOI: 10.2147/ijn.s27526] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background The generation of anti-adenovirus neutralizing antibody (NAb) in humans severely restricts the utilization of recombinant adenovirus serotype 5 (Ad5) vectors in gene therapy for a wide range of clinical trials. To overcome this limitation, we ionically complexed Ad5 with a newly synthesized copolymer, which we called APC, making an adenovirus shielded from NAb. Methods APC, a cationic polyethylene glycol derivative, was synthesized via two steps of ring-opening copolymerization of ethylene oxide and allyl glycidyl ether, followed by the addition of 2-mercaptoethylamine. The copolymer or the control PEI-2k was ionically complexed to anionic Ad5 in 5% glucose, and in vitro transduction assays were carried out in coxsackievirus and adenovirus receptor-positive cells (A549) and coxsackievirus and adenovirus receptor-negative cells (B16 and SKOV3). The physical properties and morphology of adenovirus alone or the complexes were investigated respectively by zeta potential, size distribution, and transmission electron microscopy image. Then cytotoxicity of APC was examined using 3-[4, 5-dimethylthiazol-2-yl]-2, 5-diphenyltetrazolium bromide assays. Finally, the ability of APC to protect adenovirus from NAb was evaluated by transfection assays after a neutralizing effect. Results APC was successfully synthesized and showed a low cytotoxicity. Positively charged Ad5/APC exhibited slightly increased diameter (130.2 ± 0.60 nm) than naked Ad5 (115.6 ± 5.46 nm) while Ad5/PEI-2k showed severe aggregation (1382 ± 79.9 nm). Ad5/APC achieved a gene transfection level as high as Ad5/PEI-2k in A549 or B16 cells, and significantly higher than Ad5/PEI-2k in SKOV3 cells. Most importantly, after the exposure to the neutralizing antibody, naked Ad5 and Ad5/PEI-2k exhibited poor gene expression while Ad5/APC still showed significantly efficient gene expression. Conclusion Our results demonstrated that Ad5/APC complex offered good protection for Ad5 against NAb in vitro and suggested a potential strategy of resistance to NAb in vivo.
Collapse
Affiliation(s)
- Qin Zeng
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, People's Republic of China
| | | | | | | | | | | |
Collapse
|
47
|
Duffy MR, Parker AL, Bradshaw AC, Baker AH. Manipulation of adenovirus interactions with host factors for gene therapy applications. Nanomedicine (Lond) 2012; 7:271-88. [DOI: 10.2217/nnm.11.186] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Nanomedicine based on the use of adenovirus vectors for therapeutic gene delivery shows broad potential. Specific targeting for many gene therapy applications, such as metastatic cancers or cardiovascular diseases requires intravascular delivery of the vector. However, a major barrier to successful adenovirus vector targeting follows systemic delivery, as upon contact with the bloodstream the virus interacts with a variety of host proteins, in particular coagulation factor X, which mediates profound liver gene transfer. This inherent hepatic tropism combined with macrophage scavenging minimizes the efficacy of the virus at the desired sites and induces toxic side effects. Understanding the complex, multifaceted interactions of adenovirus with host factors is of vital importance to the design of safer vectors with improved efficacy and pharmacokinetic profiles. Increased knowledge of adenovirus biology provides the opportunity to develop innovative strategies to detarget the virus from the liver following intravascular delivery and redirect the vector to disease areas.
Collapse
Affiliation(s)
- Margaret R Duffy
- British Heart Foundation Glasgow Cardiovascular Research Center, College of Medical, Veterinary & Life Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Alan L Parker
- British Heart Foundation Glasgow Cardiovascular Research Center, College of Medical, Veterinary & Life Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Angela C Bradshaw
- British Heart Foundation Glasgow Cardiovascular Research Center, College of Medical, Veterinary & Life Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| | - Andrew H Baker
- British Heart Foundation Glasgow Cardiovascular Research Center, College of Medical, Veterinary & Life Sciences, University of Glasgow, 126 University Place, Glasgow G12 8TA, UK
| |
Collapse
|
48
|
Samson M, Jung D. Intracellular trafficking and fate of chimeric adenovirus 5/F35 in human B lymphocytes. J Gene Med 2012; 13:451-61. [PMID: 21766397 DOI: 10.1002/jgm.1588] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Investigation of the molecular processes that control the development and function of lymphocytes is essential for our understanding of humoral immunity, as well as lymphocyte-associated pathogenesis. Adenovirus-mediated gene transfer provides a powerful tool for investigating these processes. However, we observed variation in transgene expression among normal human peripheral blood B lymphocytes from different donors and at distinct stages of differentiation. It is recognized that efficient gene transfer is highly dependent on the intracellular route by which the viruses travel within the host cell. Thus, we aimed to examine this aspect in the present study. METHODS We analyzed the binding, uptake, intracellular trafficking and fate of CY3-labelled Ad5/F35 vectors in lymphoid cell lines and primary B cells. Furthermore, we decreased protein synthesis levels and rapid endocytosis in a plasma cell line exhibiting a high level of protein synthesis activity and activated transcription and endocytosis in primary B cells, which are less active than plasma cells. RESULTS Major differences in intracellular trafficking pattern between B cells and plasma cell line U266 were identified that explain the observed divergence in transgene expression efficiency. Importantly, modification of the transcriptional or translational activity of U266 cells reverted the Ad5/F35 endocytic trafficking to that seen in B cells, with a loss of transgene expression, whereas activation of B cells with phorbol 12-myristate 13-acetate had the opposite effects. CONCLUSIONS Taken together, these results suggest that Ad5/F35 is more efficiently transduced in cells with a strong transcriptional activity as a result of differences in intracellular trafficking. This finding extends our current knowledge of the mechanisms of adenovirus-mediated gene transfer.
Collapse
|
49
|
Feracci H, Gutierrez BS, Hempel W, Gil IS. Organic Nanoparticles. NANOBIOTECHNOLOGY - INORGANIC NANOPARTICLES VS ORGANIC NANOPARTICLES 2012. [DOI: 10.1016/b978-0-12-415769-9.00008-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
|
50
|
Kariithi HM, Ince IA, Boeren S, Abd-Alla AMM, Parker AG, Aksoy S, Vlak JM, van Oers MM. The salivary secretome of the tsetse fly Glossina pallidipes (Diptera: Glossinidae) infected by salivary gland hypertrophy virus. PLoS Negl Trop Dis 2011; 5:e1371. [PMID: 22132244 PMCID: PMC3222630 DOI: 10.1371/journal.pntd.0001371] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2011] [Accepted: 09/05/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The competence of the tsetse fly Glossina pallidipes (Diptera; Glossinidae) to acquire salivary gland hypertrophy virus (SGHV), to support virus replication and successfully transmit the virus depends on complex interactions between Glossina and SGHV macromolecules. Critical requisites to SGHV transmission are its replication and secretion of mature virions into the fly's salivary gland (SG) lumen. However, secretion of host proteins is of equal importance for successful transmission and requires cataloging of G. pallidipes secretome proteins from hypertrophied and non-hypertrophied SGs. METHODOLOGY/PRINCIPAL FINDINGS After electrophoretic profiling and in-gel trypsin digestion, saliva proteins were analyzed by nano-LC-MS/MS. MaxQuant/Andromeda search of the MS data against the non-redundant (nr) GenBank database and a G. morsitans morsitans SG EST database, yielded a total of 521 hits, 31 of which were SGHV-encoded. On a false discovery rate limit of 1% and detection threshold of least 2 unique peptides per protein, the analysis resulted in 292 Glossina and 25 SGHV MS-supported proteins. When annotated by the Blast2GO suite, at least one gene ontology (GO) term could be assigned to 89.9% (285/317) of the detected proteins. Five (∼1.8%) Glossina and three (∼12%) SGHV proteins remained without a predicted function after blast searches against the nr database. Sixty-five of the 292 detected Glossina proteins contained an N-terminal signal/secretion peptide sequence. Eight of the SGHV proteins were predicted to be non-structural (NS), and fourteen are known structural (VP) proteins. CONCLUSIONS/SIGNIFICANCE SGHV alters the protein expression pattern in Glossina. The G. pallidipes SG secretome encompasses a spectrum of proteins that may be required during the SGHV infection cycle. These detected proteins have putative interactions with at least 21 of the 25 SGHV-encoded proteins. Our findings opens venues for developing novel SGHV mitigation strategies to block SGHV infections in tsetse production facilities such as using SGHV-specific antibodies and phage display-selected gut epithelia-binding peptides.
Collapse
Affiliation(s)
- Henry M. Kariithi
- Laboratory of Virology, Wageningen University, Wageningen, The Netherlands
- Insect Pest Control Laboratory, Programme of Nuclear Techniques in Food and Agriculture, International Atomic Energy Agency, Vienna, Austria
| | - Ikbal A. Ince
- Laboratory of Virology, Wageningen University, Wageningen, The Netherlands
- Department of Genetics and Bioengineering, Yeditepe University, Istanbul, Turkey
| | - Sjef Boeren
- Laboratory of Biochemistry, Wageningen University, Wageningen, The Netherlands
| | - Adly M. M. Abd-Alla
- Insect Pest Control Laboratory, Programme of Nuclear Techniques in Food and Agriculture, International Atomic Energy Agency, Vienna, Austria
| | - Andrew G. Parker
- Insect Pest Control Laboratory, Programme of Nuclear Techniques in Food and Agriculture, International Atomic Energy Agency, Vienna, Austria
| | - Serap Aksoy
- Yale School of Public Health, New Haven, Connecticut, United States of America
| | - Just M. Vlak
- Laboratory of Virology, Wageningen University, Wageningen, The Netherlands
| | | |
Collapse
|