1
|
Baldini L, Keller B, Dewitte L, Passarelli C, Ginevrino M, Carli D, Montin D, Bossuyt X, Warnatz K, Licciardi F. BENTA disease or CARD11 gain-of-function? A novel variant with atypical features and a literature review. Immunol Lett 2025; 275:107005. [PMID: 40157432 DOI: 10.1016/j.imlet.2025.107005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 02/28/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
INTRODUCTION The CARD11 (Caspase Recruitment Domain Family Member 11) gene encodes a scaffold protein critical for NF-κB signaling, regulating B-cell differentiation and T-cell effector functions. Gain-of-function (GOF) mutations in CARD11 cause BENTA disease (B cell Expansion with NF-κB and T cell Anergy), an autosomal dominant disorder typically presenting with early-onset polyclonal B-cell lymphocytosis, splenomegaly, lymphadenopathy, and recurrent infections. METHODS We describe three related patients harboring a novel CARD11-GOF mutation (D357E), presenting with a BENTA phenotype with atypical features, including high IgM levels and a normal B-cell count, with life-threatening HLH in one case. Additionally, we conducted a systematic literature review using PubMed and EMBASE to identify previously reported cases of CARD11 GOF mutations. RESULTS In vitro functional analysis demonstrated that the D357E variant activates the NF-κB signaling pathway in primary lymphocytes and in HEK293T cells transfected with mutant CARD11. Our literature review identified 13 studies describing 29 patients. Notably, HLH emerged as a common complication of CARD11 GOF mutations (18.8 %), while B-lymphocytosis -though frequent- was not universally present. CONCLUSION We identified a novel pathogenic CARD11 variant and described its atypical phenotype, further expanding the clinical spectrum of CARD11 GOF disorders. These findings underscore the need for increased awareness of HLH risk in patients with CARD11 GOF mutations.
Collapse
Affiliation(s)
- Letizia Baldini
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy; Dipartimento di Scienza della Sanità Pubblica e Pediatriche, Università degli Studi di Torino, Torino, Italy.
| | - Bärbel Keller
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Lisa Dewitte
- Department of Microbiology, Immunology and Transplantation, Clinical and Diagnostic Immunology, KU Leuven, Leuven, Belgium
| | - Chiara Passarelli
- Laboratory of Medical Genetics, Translational Cytogenomics Research Unit, Bambino Gesù Children Hospital, IRCCS, 00146 Rome, Italy
| | - Monia Ginevrino
- Laboratory of Medical Genetics, Translational Cytogenomics Research Unit, Bambino Gesù Children Hospital, IRCCS, 00146 Rome, Italy
| | - Diana Carli
- Dipartimento di Scienze Mediche, Università degli Studi di Torino, Torino, Italy
| | - Davide Montin
- Dipartimento di Scienza della Sanità Pubblica e Pediatriche, Università degli Studi di Torino, Torino, Italy; Dipartimento di Patologia e Cura del Bambino "Regina Margherita", Ospedale Infantile Regina Margherita, Torino, Italy
| | - Xavier Bossuyt
- Department of Microbiology, Immunology and Transplantation, Clinical and Diagnostic Immunology, KU Leuven, Leuven, Belgium
| | - Klaus Warnatz
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Department of Immunology, University Hospital Zurich, Zurich, Switzerland
| | - Francesco Licciardi
- Dipartimento di Scienza della Sanità Pubblica e Pediatriche, Università degli Studi di Torino, Torino, Italy; Dipartimento di Patologia e Cura del Bambino "Regina Margherita", Ospedale Infantile Regina Margherita, Torino, Italy
| |
Collapse
|
2
|
Goolab S, Scholefield J. Making gene editing accessible in resource limited environments: recommendations to guide a first-time user. Front Genome Ed 2024; 6:1464531. [PMID: 39386178 PMCID: PMC11461239 DOI: 10.3389/fgeed.2024.1464531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 09/05/2024] [Indexed: 10/12/2024] Open
Abstract
The designer nuclease, CRISPR-Cas9 system has advanced the field of genome engineering owing to its programmability and ease of use. The application of these molecular scissors for genome engineering earned the developing researchers the Nobel prize in Chemistry in the year 2020. At present, the potential of this technology to improve global challenges continues to grow exponentially. CRISPR-Cas9 shows promise in the recent advances made in the Global North such as the FDA-approved gene therapy for the treatment of sickle cell anaemia and β-thalassemia and the gene editing of porcine kidney for xenotransplantation into humans affected by end-stage kidney failure. Limited resources, low government investment with an allocation of 1% of gross domestic production to research and development including a shortage of skilled professionals and lack of knowledge may preclude the use of this revolutionary technology in the Global South where the countries involved have reduced science and technology budgets. Focusing on the practical application of genome engineering, successful genetic manipulation is not easily accomplishable and is influenced by the chromatin landscape of the target locus, guide RNA selection, the experimental design including the profiling of the gene edited cells, which impacts the overall outcome achieved. Our assessment primarily delves into economical approaches of performing efficient genome engineering to support the first-time user restricted by limited resources with the aim of democratizing the use of the technology across low- and middle-income countries. Here we provide a comprehensive overview on existing experimental techniques, the significance for target locus analysis and current pitfalls such as the underrepresentation of global genetic diversity. Several perspectives of genome engineering approaches are outlined, which can be adopted in a resource limited setting to enable a higher success rate of genome editing-based innovations in low- and middle-income countries.
Collapse
Affiliation(s)
- Shivani Goolab
- Bioengineering and Integrated Genomics Group, Future Production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa
| | - Janine Scholefield
- Bioengineering and Integrated Genomics Group, Future Production Chemicals Cluster, Council for Scientific and Industrial Research, Pretoria, South Africa
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Division of Human Genetics, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
3
|
Sokolinskaya EL, Ivanova ON, Fedyakina IT, Ivanov AV, Lukyanov KA. Natural-Target-Mimicking Translocation-Based Fluorescent Sensor for Detection of SARS-CoV-2 PLpro Protease Activity and Virus Infection in Living Cells. Int J Mol Sci 2024; 25:6635. [PMID: 38928340 PMCID: PMC11203561 DOI: 10.3390/ijms25126635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 06/13/2024] [Accepted: 06/15/2024] [Indexed: 06/28/2024] Open
Abstract
Papain-like protease PLpro, a domain within a large polyfunctional protein, nsp3, plays key roles in the life cycle of SARS-CoV-2, being responsible for the first events of cleavage of a polyprotein into individual proteins (nsp1-4) as well as for the suppression of cellular immunity. Here, we developed a new genetically encoded fluorescent sensor, named PLpro-ERNuc, for detection of PLpro activity in living cells using a translocation-based readout. The sensor was designed as follows. A fragment of nsp3 protein was used to direct the sensor on the cytoplasmic surface of the endoplasmic reticulum (ER) membrane, thus closely mimicking the natural target of PLpro. The fluorescent part included two bright fluorescent proteins-red mScarlet I and green mNeonGreen-separated by a linker with the PLpro cleavage site. A nuclear localization signal (NLS) was attached to ensure accumulation of mNeonGreen into the nucleus upon cleavage. We tested PLpro-ERNuc in a model of recombinant PLpro expressed in HeLa cells. The sensor demonstrated the expected cytoplasmic reticular network in the red and green channels in the absence of protease, and efficient translocation of the green signal into nuclei in the PLpro-expressing cells (14-fold increase in the nucleus/cytoplasm ratio). Then, we used PLpro-ERNuc in a model of Huh7.5 cells infected with the SARS-CoV-2 virus, where it showed robust ER-to-nucleus translocation of the green signal in the infected cells 24 h post infection. We believe that PLpro-ERNuc represents a useful tool for screening PLpro inhibitors as well as for monitoring virus spread in a culture.
Collapse
Affiliation(s)
- Elena L. Sokolinskaya
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia;
| | - Olga N. Ivanova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (O.N.I.); (A.V.I.)
| | - Irina T. Fedyakina
- Gamaleya National Research Centre for Epidemiology and Microbiology of the Ministry of Russia, 132098 Moscow, Russia;
| | - Alexander V. Ivanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (O.N.I.); (A.V.I.)
| | - Konstantin A. Lukyanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia;
| |
Collapse
|
4
|
Bulcaen M, Kortleven P, Liu RB, Maule G, Dreano E, Kelly M, Ensinck MM, Thierie S, Smits M, Ciciani M, Hatton A, Chevalier B, Ramalho AS, Casadevall I Solvas X, Debyser Z, Vermeulen F, Gijsbers R, Sermet-Gaudelus I, Cereseto A, Carlon MS. Prime editing functionally corrects cystic fibrosis-causing CFTR mutations in human organoids and airway epithelial cells. Cell Rep Med 2024; 5:101544. [PMID: 38697102 PMCID: PMC11148721 DOI: 10.1016/j.xcrm.2024.101544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 01/16/2024] [Accepted: 04/10/2024] [Indexed: 05/04/2024]
Abstract
Prime editing is a recent, CRISPR-derived genome editing technology capable of introducing precise nucleotide substitutions, insertions, and deletions. Here, we present prime editing approaches to correct L227R- and N1303K-CFTR, two mutations that cause cystic fibrosis and are not eligible for current market-approved modulator therapies. We show that, upon DNA correction of the CFTR gene, the complex glycosylation, localization, and, most importantly, function of the CFTR protein are restored in HEK293T and 16HBE cell lines. These findings were subsequently validated in patient-derived rectal organoids and human nasal epithelial cells. Through analysis of predicted and experimentally identified candidate off-target sites in primary stem cells, we confirm previous reports on the high prime editor (PE) specificity and its potential for a curative CF gene editing therapy. To facilitate future screening of genetic strategies in a translational CF model, a machine learning algorithm was developed for dynamic quantification of CFTR function in organoids (DETECTOR: "detection of targeted editing of CFTR in organoids").
Collapse
Affiliation(s)
- Mattijs Bulcaen
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium; Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium.
| | - Phéline Kortleven
- Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium
| | - Ronald B Liu
- Department of Biosystems, KU Leuven, 3000 Leuven, Belgium; School of Engineering, University of Edinburgh, EH9 3JL Edinburgh, UK
| | - Giulia Maule
- Department of CIBIO, University of Trento, 38123 Povo-Trento, Italy
| | - Elise Dreano
- INSERM, CNRS, Institut Necker Enfants Malades, 75015 Paris, France; Université Paris-Cité, 75015 Paris, France
| | - Mairead Kelly
- INSERM, CNRS, Institut Necker Enfants Malades, 75015 Paris, France; Université Paris-Cité, 75015 Paris, France
| | - Marjolein M Ensinck
- Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium
| | - Sam Thierie
- Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium
| | - Maxime Smits
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium; Leuven Viral Vector Core, KU Leuven, 3000 Leuven, Belgium
| | - Matteo Ciciani
- Department of CIBIO, University of Trento, 38123 Povo-Trento, Italy
| | - Aurelie Hatton
- INSERM, CNRS, Institut Necker Enfants Malades, 75015 Paris, France; Université Paris-Cité, 75015 Paris, France
| | - Benoit Chevalier
- INSERM, CNRS, Institut Necker Enfants Malades, 75015 Paris, France; Université Paris-Cité, 75015 Paris, France
| | - Anabela S Ramalho
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium
| | | | - Zeger Debyser
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium; Leuven Viral Vector Core, KU Leuven, 3000 Leuven, Belgium
| | - François Vermeulen
- Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium; Department of Pediatrics, UZ Leuven, 3000 Leuven, Belgium
| | - Rik Gijsbers
- Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium; Leuven Viral Vector Core, KU Leuven, 3000 Leuven, Belgium
| | - Isabelle Sermet-Gaudelus
- INSERM, CNRS, Institut Necker Enfants Malades, 75015 Paris, France; Université Paris-Cité, 75015 Paris, France; Cystic Fibrosis National Pediatric Reference Center, Pneumo-Allergologie Pédiatrique, Hôpital Necker Enfants Malades, Assistance Publique Hôpitaux de Paris (AP-HP), 75015 Paris, France; European Reference Network, ERN-Lung CF, 60596 Frankfurt am Mein, Germany
| | - Anna Cereseto
- Department of CIBIO, University of Trento, 38123 Povo-Trento, Italy
| | - Marianne S Carlon
- Department of Chronic Diseases and Metabolism, KU Leuven, 3000 Leuven, Belgium; Leuven Viral Vector Core, KU Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
5
|
Medaer L, David D, Smits M, Levtchenko E, Sampaolesi M, Gijsbers R. Residual Cystine Transport Activity for Specific Infantile and Juvenile CTNS Mutations in a PTEC-Based Addback Model. Cells 2024; 13:646. [PMID: 38607085 PMCID: PMC11011962 DOI: 10.3390/cells13070646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 04/03/2024] [Accepted: 04/04/2024] [Indexed: 04/13/2024] Open
Abstract
Cystinosis is a rare, autosomal recessive, lysosomal storage disease caused by mutations in the gene CTNS, leading to cystine accumulation in the lysosomes. While cysteamine lowers the cystine levels, it does not cure the disease, suggesting that CTNS exerts additional functions besides cystine transport. This study investigated the impact of infantile and juvenile CTNS mutations with discrepant genotype/phenotype correlations on CTNS expression, and subcellular localisation and function in clinically relevant cystinosis cell models to better understand the link between genotype and CTNS function. Using CTNS-depleted proximal tubule epithelial cells and patient-derived fibroblasts, we expressed a selection of CTNSmutants under various promoters. EF1a-driven expression led to substantial overexpression, resulting in CTNS protein levels that localised to the lysosomal compartment. All CTNSmutants tested also reversed cystine accumulation, indicating that CTNSmutants still exert transport activity, possibly due to the overexpression conditions. Surprisingly, even CTNSmutants expression driven by the less potent CTNS and EFS promoters reversed the cystine accumulation, contrary to the CTNSG339R missense mutant. Taken together, our findings shed new light on CTNS mutations, highlighting the need for robust assessment methodologies in clinically relevant cellular models and thus paving the way for better stratification of cystinosis patients, and advocating for the development of more personalized therapy.
Collapse
Affiliation(s)
- Louise Medaer
- Laboratory of Molecular Virology and Gene Therapy, Department of Pharmacological and Pharmaceutical Sciences, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium; (L.M.); (M.S.)
| | - Dries David
- Laboratory of Molecular Virology and Gene Therapy, Department of Pharmacological and Pharmaceutical Sciences, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium; (L.M.); (M.S.)
| | - Maxime Smits
- Laboratory of Molecular Virology and Gene Therapy, Department of Pharmacological and Pharmaceutical Sciences, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium; (L.M.); (M.S.)
- Leuven Viral Vector Core, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium
| | - Elena Levtchenko
- Department of Paediatric Nephrology & Development and Regeneration, University Hospitals Leuven & KU Leuven, 3000 Leuven, Belgium;
- Department of Paediatric Nephrology, Amsterdam University Medical Centre, 1081 Amsterdam, The Netherlands
| | - Maurilio Sampaolesi
- Translational Cardiology Laboratory, Department of Development and Regeneration, Stem Cell Institute, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium;
| | - Rik Gijsbers
- Laboratory of Molecular Virology and Gene Therapy, Department of Pharmacological and Pharmaceutical Sciences, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium; (L.M.); (M.S.)
- Leuven Viral Vector Core, Faculty of Medicine, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
6
|
Ortuste Quiroga HP, Fujimaki S, Ono Y. Pax7 reporter mouse models: a pocket guide for satellite cell research. Eur J Transl Myol 2023; 33:12174. [PMID: 38112596 PMCID: PMC10811643 DOI: 10.4081/ejtm.2023.12174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 12/21/2023] Open
Abstract
Since their discovery, satellite cells have showcased their need as primary contributors to skeletal muscle maintenance and repair. Satellite cells lay dormant, but when needed, activate, differentiate, fuse to fibres and self-renew, that has bestowed satellite cells with the title of muscle stem cells. The satellite cell specific transcription factor Pax7 has enabled researchers to develop animal models against the Pax7 locus in order to isolate and characterise satellite cell-mediated events. This review focuses specifically on describing Pax7 reporter mouse models. Here we describe how each model was generated and the key findings obtained. The strengths and limitations of each model are also discussed. The aim is to provide new and current satellite cell enthusiasts with a basic understanding of the available Pax7 reporter mice and hopefully guide selection of the most appropriate Pax7 model to answer a specific research question.
Collapse
Affiliation(s)
- Huascar Pedro Ortuste Quiroga
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Honjo, Chuo-ku, Kumamoto.
| | - Shin Fujimaki
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Honjo, Chuo-ku, Kumamoto.
| | - Yusuke Ono
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, Honjo, Chuo-ku, Kumamoto, Japan; Tokyo Metropolitan Institute for Geriatrics and Gerontology (TMIG), Sakae-cho, Itabashi, Tokyo.
| |
Collapse
|
7
|
VanDyke D, Xu L, Sargunas PR, Gilbreth RN, Baca M, Gao C, Hunt J, Spangler JB. Redirecting the specificity of tripartite motif containing-21 scaffolds using a novel discovery and design approach. J Biol Chem 2023; 299:105381. [PMID: 37866632 PMCID: PMC10694607 DOI: 10.1016/j.jbc.2023.105381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 09/30/2023] [Accepted: 10/16/2023] [Indexed: 10/24/2023] Open
Abstract
Hijacking the ubiquitin proteasome system to elicit targeted protein degradation (TPD) has emerged as a promising therapeutic strategy to target and destroy intracellular proteins at the post-translational level. Small molecule-based TPD approaches, such as proteolysis-targeting chimeras (PROTACs) and molecular glues, have shown potential, with several agents currently in clinical trials. Biological PROTACs (bioPROTACs), which are engineered fusion proteins comprised of a target-binding domain and an E3 ubiquitin ligase, have emerged as a complementary approach for TPD. Here, we describe a new method for the evolution and design of bioPROTACs. Specifically, engineered binding scaffolds based on the third fibronectin type III domain of human tenascin-C (Tn3) were installed into the E3 ligase tripartite motif containing-21 (TRIM21) to redirect its degradation specificity. This was achieved via selection of naïve yeast-displayed Tn3 libraries against two different oncogenic proteins associated with B-cell lymphomas, mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1) and embryonic ectoderm development protein (EED), and replacing the native substrate-binding domain of TRIM21 with our evolved Tn3 domains. The resulting TRIM21-Tn3 fusion proteins retained the binding properties of the Tn3 as well as the E3 ligase activity of TRIM21. Moreover, we demonstrated that TRIM21-Tn3 fusion proteins efficiently degraded their respective target proteins through the ubiquitin proteasome system in cellular models. We explored the effects of binding domain avidity and E3 ligase utilization to gain insight into the requirements for effective bioPROTAC design. Overall, this study presents a versatile engineering approach that could be used to design and engineer TRIM21-based bioPROTACs against therapeutic targets.
Collapse
Affiliation(s)
- Derek VanDyke
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Biologics Engineering, R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Linda Xu
- Biologics Engineering, R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Paul R Sargunas
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ryan N Gilbreth
- Biologics Engineering, R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Manuel Baca
- Biologics Engineering, R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Changshou Gao
- Biologics Engineering, R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - James Hunt
- Biologics Engineering, R&D, AstraZeneca, Cambridge, UK
| | - Jamie B Spangler
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, Maryland, USA; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
8
|
Kemmler CL, Moran HR, Murray BF, Scoresby A, Klem JR, Eckert RL, Lepovsky E, Bertho S, Nieuwenhuize S, Burger S, D'Agati G, Betz C, Puller AC, Felker A, Ditrychova K, Bötschi S, Affolter M, Rohner N, Lovely CB, Kwan KM, Burger A, Mosimann C. Next-generation plasmids for transgenesis in zebrafish and beyond. Development 2023; 150:dev201531. [PMID: 36975217 PMCID: PMC10263156 DOI: 10.1242/dev.201531] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 03/10/2023] [Indexed: 03/29/2023]
Abstract
Transgenesis is an essential technique for any genetic model. Tol2-based transgenesis paired with Gateway-compatible vector collections has transformed zebrafish transgenesis with an accessible modular system. Here, we establish several next-generation transgenesis tools for zebrafish and other species to expand and enhance transgenic applications. To facilitate gene regulatory element testing, we generated Gateway middle entry vectors harboring the small mouse beta-globin minimal promoter coupled to several fluorophores, CreERT2 and Gal4. To extend the color spectrum for transgenic applications, we established middle entry vectors encoding the bright, blue-fluorescent protein mCerulean and mApple as an alternative red fluorophore. We present a series of p2A peptide-based 3' vectors with different fluorophores and subcellular localizations to co-label cells expressing proteins of interest. Finally, we established Tol2 destination vectors carrying the zebrafish exorh promoter driving different fluorophores as a pineal gland-specific transgenesis marker that is active before hatching and through adulthood. exorh-based reporters and transgenesis markers also drive specific pineal gland expression in the eye-less cavefish (Astyanax). Together, our vectors provide versatile reagents for transgenesis applications in zebrafish, cavefish and other models.
Collapse
Affiliation(s)
- Cassie L. Kemmler
- University of Colorado, School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Hannah R. Moran
- University of Colorado, School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Brooke F. Murray
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Aaron Scoresby
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - John R. Klem
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Rachel L. Eckert
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Elizabeth Lepovsky
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Sylvain Bertho
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Susan Nieuwenhuize
- University of Colorado, School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Sibylle Burger
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Gianluca D'Agati
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Charles Betz
- Growth & Development, Biozentrum, Spitalstrasse 41, University of Basel, 4056 Basel, Switzerland
| | - Ann-Christin Puller
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Anastasia Felker
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Karolina Ditrychova
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Seraina Bötschi
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Markus Affolter
- Growth & Development, Biozentrum, Spitalstrasse 41, University of Basel, 4056 Basel, Switzerland
| | - Nicolas Rohner
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - C. Ben Lovely
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Kristen M. Kwan
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Alexa Burger
- University of Colorado, School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Christian Mosimann
- University of Colorado, School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA
| |
Collapse
|
9
|
Chang C, Music N, Cheung M, Rossignol E, Bedi S, Patel H, Safari M, Lee C, Otten GR, Settembre EC, Palladino G, Wen Y. Self-amplifying mRNA bicistronic influenza vaccines raise cross-reactive immune responses in mice and prevent infection in ferrets. Mol Ther Methods Clin Dev 2022; 27:195-205. [PMID: 36320414 PMCID: PMC9589142 DOI: 10.1016/j.omtm.2022.09.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 09/28/2022] [Indexed: 11/17/2022]
Abstract
Vaccines are the primary intervention against influenza. Currently licensed inactivated vaccines focus immunity on viral hemagglutinin (HA). Self-amplifying mRNA (sa-mRNA) vaccines offer an opportunity to generate immunity to multiple viral proteins, including additional neuraminidase (NA). This evaluation of a bicistronic approach for sa-mRNA vaccine development compared subgenomic promoter and internal ribosome entry site strategies and found consistent and balanced expression of both HA and NA proteins in transfected cells. In mice, sa-mRNA bicistronic A/H5N1 vaccines raised potent anti-HA and anti-NA neutralizing antibody responses and HA- or NA-specific CD4+ and CD8+ T cell responses. The addition of NA also boosted the cross-neutralizing response to heterologous A/H1N1. Similar immunogenicity results were obtained for bicistronic seasonal A/H3N2 and B/Yamagata vaccines. In ferrets, sa-mRNA bicistronic A/H1N1 vaccine fully protected lung from infection by homologous virus and showed significant reduction of viral load in upper respiratory tract, warranting further evaluation of sa-mRNA bicistronic vaccine in humans.
Collapse
Affiliation(s)
- Cheng Chang
- CSL, 50 Hampshire Street, Cambridge, MA 02139, USA
| | - Nedzad Music
- CSL, 50 Hampshire Street, Cambridge, MA 02139, USA
| | | | | | | | - Harsh Patel
- CSL, 50 Hampshire Street, Cambridge, MA 02139, USA
| | | | | | | | | | | | - Yingxia Wen
- CSL, 50 Hampshire Street, Cambridge, MA 02139, USA,Corresponding author Yingxia Wen, CSL, 50 Hampshire Street, Cambridge, MA 02139, USA.
| |
Collapse
|
10
|
Wüst R, Terrie L, Müntefering T, Ruck T, Thorrez L. Efficient co-isolation of microvascular endothelial cells and satellite cell-derived myoblasts from human skeletal muscle. Front Bioeng Biotechnol 2022; 10:964705. [PMID: 36213083 PMCID: PMC9534561 DOI: 10.3389/fbioe.2022.964705] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/18/2022] [Indexed: 12/03/2022] Open
Abstract
Vascularization of tissue-engineered constructs remains a key challenge in the field of skeletal muscle tissue engineering. One strategy for vascularizing organoids is in vitro pre-vascularization, relying on de novo assembly of undifferentiated endothelial cells into capillaries, a process termed vasculogenesis. In most endothelial cell research to date, human umbilical vein endothelial cells have been used primarily because of their availability. Nevertheless, this endothelial cell type is naturally not occurring in skeletal muscle tissue. Since endothelial cells display a tissue-specific phenotype, it is of interest to use muscle-specific microvascular endothelial cells to study pre-vascularization in skeletal muscle tissue engineering research. Thus far, tissue biopsies had to be processed in two separate protocols to obtain cells from the myogenic and the endothelial compartment. Here, we describe a novel, detailed protocol for the co-isolation of human skeletal muscle microvascular endothelial cells and satellite cell-derived myoblasts. It incorporates an automated mechanical and enzymatic tissue dissociation followed by magnetically activated cell sorting based on a combination of endothelial and skeletal muscle cell markers. Qualitative, quantitative, and functional characterization of the obtained cells is described and demonstrated by representative results. The simultaneous isolation of both cell types from the same donor is advantageous in terms of time efficiency. In addition, it may be the only possible method to isolate both cell types as the amount of tissue biopsy is often limited. The isolation of the two cell types is crucial for further studies to elucidate cell crosstalk in health and disease. Furthermore, the use of muscle-specific microvascular endothelial cells allows a shift towards engineering more physiologically relevant functional tissue, with downstream applications including drug screening and regenerative medicine.
Collapse
Affiliation(s)
- Rebecca Wüst
- Tissue Engineering Lab, Dep. Development and Regeneration, KU Leuven Kulak, Kortrijk, Belgium
| | - Lisanne Terrie
- Tissue Engineering Lab, Dep. Development and Regeneration, KU Leuven Kulak, Kortrijk, Belgium
| | - Thomas Müntefering
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Tobias Ruck
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Lieven Thorrez
- Tissue Engineering Lab, Dep. Development and Regeneration, KU Leuven Kulak, Kortrijk, Belgium
- *Correspondence: Lieven Thorrez,
| |
Collapse
|
11
|
Li Y, Cui ZJ. Photodynamic Activation of the Cholecystokinin 1 Receptor with Tagged Genetically Encoded Protein Photosensitizers: Optimizing the Tagging Patterns. Photochem Photobiol 2022; 98:1215-1228. [PMID: 35211987 DOI: 10.1111/php.13611] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 02/20/2022] [Indexed: 02/05/2023]
Abstract
Cholecystokinin 1 receptor (CCK1R) is activated photodynamically. For this to happen in situ, genetically encoded protein photosensitizers (GEPP) may be tagged to natively expressed CCK1R, but how to best tag GEPP has not been examined. Therefore, GEPP (miniSOG or KillerRed) was tagged to CCK1R and light-driven photodynamic CCK1R activation was monitored by Fura-2 fluorescent calcium imaging, to screen for optimized tagging patterns. Blue light-emitting diode irradiation of CHO-K1 cells expressing miniSOG fused to N- or C-terminus of CCK1R was found to both trigger persistent calcium oscillations-a hallmark of permanent photodynamic CCK1R activation. Photodynamic CCK1R activation was accomplished also with miniSOG fused to N-terminus of CCK1R via linker (GlySerGly)4 or 8 , but not linker (GSG)12 or an internal ribosomal entry site insert. KillerRed fused to N- or C-terminus of CCK1R after white light irradiation resulted in similar activation of in-frame CCK1R. Photodynamic CCK1R activation in miniSOG-CCK1R-CHO-K1 cells was blocked by singlet oxygen (1 O2 ) quencher uric acid or Trolox C, corroborating the role of 1 O2 as the reactive intermediate. It is concluded that photodynamic CCK1R activation can be achieved either with direct GEPP fusion to CCK1R or fusion via a short linker, fusion via long linkers might serve as the internal control.
Collapse
Affiliation(s)
- Yuan Li
- Institute of Cell Biology, Beijing Normal University, Beijing, China
| | - Zong Jie Cui
- Institute of Cell Biology, Beijing Normal University, Beijing, China
| |
Collapse
|
12
|
Sokolinskaya EL, Putlyaeva LV, Polinovskaya VS, Lukyanov KA. Genetically Encoded Fluorescent Sensors for SARS-CoV-2 Papain-like Protease PLpro. Int J Mol Sci 2022; 23:ijms23147826. [PMID: 35887174 PMCID: PMC9318946 DOI: 10.3390/ijms23147826] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/07/2022] [Accepted: 07/14/2022] [Indexed: 02/04/2023] Open
Abstract
In the SARS-CoV-2 lifecycle, papain-like protease PLpro cuts off the non-structural proteins nsp1, nsp2, and nsp3 from a large polyprotein. This is the earliest viral enzymatic activity, which is crucial for all downstream steps. Here, we designed two genetically encoded fluorescent sensors for the real-time detection of PLpro activity in live cells. The first sensor was based on the Förster resonance energy transfer (FRET) between the red fluorescent protein mScarlet as a donor and the biliverdin-binding near-infrared fluorescent protein miRFP670 as an acceptor. A linker with the PLpro recognition site LKGG in between made this FRET pair sensitive to PLpro cleavage. Upon the co-expression of mScarlet-LKGG-miRFP670 and PLpro in HeLa cells, we observed a gradual increase in the donor fluorescence intensity of about 1.5-fold. In the second sensor, both PLpro and its target—green mNeonGreen and red mScarletI fluorescent proteins separated by an LKGG-containing linker—were attached to the endoplasmic reticulum (ER) membrane. Upon cleavage by PLpro, mScarletI diffused from the ER throughout the cell. About a two-fold increase in the nucleus/cytoplasm ratio was observed as a result of the PLpro action. We believe that the new PLpro sensors can potentially be used to detect the earliest stages of SARS-CoV-2 propagation in live cells as well as for the screening of PLpro inhibitors.
Collapse
|
13
|
Masilamani AP, Schulzki R, Yuan S, Haase IV, Kling E, Dewes F, Andrieux G, Börries M, Schnell O, Heiland DH, Schilling O, Ferrarese R, Carro MS. Calpain-mediated cleavage generates a ZBTB18 N-terminal product that regulates HIF1A signaling and glioblastoma metabolism. iScience 2022; 25:104625. [PMID: 35800763 PMCID: PMC9253709 DOI: 10.1016/j.isci.2022.104625] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 05/13/2022] [Accepted: 06/10/2022] [Indexed: 11/28/2022] Open
Abstract
Proteolytic cleavage is an important post-translational mechanism to increase protein variability and functionality. In cancer, this process can be deregulated to shut off tumor-suppressive functions. Here, we report that in glioblastoma (GBM), the tumor suppressor ZBTB18 is targeted for protein cleavage by the intracellular protease calpain. The N-terminal (Nte) ZBTB18 cleaved fragment localizes to the cytoplasm and thus, is unable to exert the gene expression repressive function of the uncleaved protein. Mass spectrometry (MS) analysis indicates that the Nte ZBTB18 short form (SF) interacts with C-terminal (Cte) binding proteins 1 and 2 (CTBP1/2), which appear to be involved in HIF1A signaling activation. In fact, we show that the new ZBTB18 product activates HIF1A-regulated genes, which in turn lead to increased lipid uptake, lipid droplets (LD) accumulation, and enhanced metabolic activity. We propose that calpain-mediated ZBTB18 cleavage represents a new mechanism to counteract ZBTB18 tumor suppression and increase tumor-promoting functions in GBM cells.
Collapse
Affiliation(s)
- Anie P. Masilamani
- Department of Neurosurgery, Medical Center – University of Freiburg, Freiburg, Germany
| | - Rana Schulzki
- Department of Neurosurgery, Medical Center – University of Freiburg, Freiburg, Germany
| | - Shuai Yuan
- Department of Neurosurgery, Medical Center – University of Freiburg, Freiburg, Germany
| | - Ira V. Haase
- Department of Neurosurgery, Medical Center – University of Freiburg, Freiburg, Germany
| | - Eva Kling
- Department of Neurosurgery, Medical Center – University of Freiburg, Freiburg, Germany
| | - Franziska Dewes
- Department of Neurosurgery, Medical Center – University of Freiburg, Freiburg, Germany
| | - Geoffroy Andrieux
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site Freiburg, Freiburg, Germany
| | - Melanie Börries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site Freiburg, Freiburg, Germany
| | - Oliver Schnell
- Department of Neurosurgery, Medical Center – University of Freiburg, Freiburg, Germany
| | - Dieter H. Heiland
- Department of Neurosurgery, Medical Center – University of Freiburg, Freiburg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site Freiburg, Freiburg, Germany
| | - Oliver Schilling
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site Freiburg, Freiburg, Germany
- Institute of Clinical Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Roberto Ferrarese
- Department of Neurosurgery, Medical Center – University of Freiburg, Freiburg, Germany
| | - Maria S. Carro
- Department of Neurosurgery, Medical Center – University of Freiburg, Freiburg, Germany
| |
Collapse
|
14
|
Van Breedam E, Nijak A, Buyle-Huybrecht T, Di Stefano J, Boeren M, Govaerts J, Quarta A, Swartenbroekx T, Jacobs EZ, Menten B, Gijsbers R, Delputte P, Alaerts M, Hassannia B, Loeys B, Berneman Z, Timmermans JP, Jorens PG, Vanden Berghe T, Fransen E, Wouters A, De Vos WH, Ponsaerts P. Luminescent Human iPSC-Derived Neurospheroids Enable Modeling of Neurotoxicity After Oxygen-glucose Deprivation. Neurotherapeutics 2022; 19:550-569. [PMID: 35289376 PMCID: PMC9226265 DOI: 10.1007/s13311-022-01212-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2022] [Indexed: 12/26/2022] Open
Abstract
Despite the considerable impact of stroke on both the individual and on society, a neuroprotective therapy for stroke patients is missing. This is partially due to the current lack of a physiologically relevant human in vitro stroke model. To address this problem, we have developed a luminescent human iPSC-derived neurospheroid model that enables real-time read-out of neural viability after ischemia-like conditions. We subjected 1- and 4-week-old neurospheroids, generated from iPSC-derived neural stem cells, to 6 h of oxygen-glucose deprivation (OGD) and measured neurospheroid luminescence. For both, we detected a decrease in luminescent signal due to ensuing neurotoxicity, as confirmed by conventional LDH assay and flow cytometric viability analysis. Remarkably, 1-week-old, but not 4-week-old neurospheroids recovered from OGD-induced injury, as evidenced by their reduced but overall increasing luminescence over time. This underscores the need for more mature neurospheroids, more faithfully recapitulating the in vivo situation. Furthermore, treatment of oxygen- and glucose-deprived neurospheroids with the pan-caspase inhibitor Z-VAD-FMK did not increase overall neural survival, despite its successful attenuation of apoptosis, in a human-based 3D environment. Nevertheless, owing to its three-dimensional organization and real-time viability reporting potential, the luminescent neurospheroids may become readily adopted in high-throughput screens aimed at identification of new therapeutic agents to treat acute ischemic stroke patients.
Collapse
Affiliation(s)
- Elise Van Breedam
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, 2610, Wilrijk, Belgium
| | - Aleksandra Nijak
- Cardiogenomics Group, Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, 2650, Edegem, Belgium
| | - Tamariche Buyle-Huybrecht
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, 2610, Wilrijk, Belgium
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp, 2610, Wilrijk, Belgium
| | - Julia Di Stefano
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, 2610, Wilrijk, Belgium
| | - Marlies Boeren
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, 2610, Wilrijk, Belgium
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp, 2610, Wilrijk, Belgium
| | - Jonas Govaerts
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, 2610, Wilrijk, Belgium
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp, 2610, Wilrijk, Belgium
| | - Alessandra Quarta
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, 2610, Wilrijk, Belgium
| | - Tine Swartenbroekx
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, 2610, Wilrijk, Belgium
| | - Eva Z Jacobs
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University Hospital, 9000, Ghent, Belgium
| | - Björn Menten
- Center for Medical Genetics Ghent, Department of Biomolecular Medicine, Ghent University Hospital, 9000, Ghent, Belgium
| | - Rik Gijsbers
- Laboratory for Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, Faculty of Medicine, KU Leuven, 3000, Leuven, Belgium
- Leuven Viral Vector Core (LVVC), KU Leuven, 3000, Leuven, Belgium
| | - Peter Delputte
- Laboratory for Microbiology, Parasitology and Hygiene, University of Antwerp, 2610, Wilrijk, Belgium
| | - Maaike Alaerts
- Cardiogenomics Group, Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, 2650, Edegem, Belgium
| | - Behrouz Hassannia
- Center for Inflammation Research (IRC), VIB-UGent, 9052, Zwijnaarde, Belgium
- Laboratory of Pathophysiology, University of Antwerp, 2610, Wilrijk, Belgium
| | - Bart Loeys
- Cardiogenomics Group, Center of Medical Genetics, University of Antwerp and Antwerp University Hospital, 2650, Edegem, Belgium
| | - Zwi Berneman
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, 2610, Wilrijk, Belgium
| | | | - Philippe G Jorens
- Laboratory of Experimental Medicine and Pediatrics (LEMP), University of Antwerp, 2610, Wilrijk, Belgium
- Department of Intensive Care Medicine, Antwerp University Hospital, 2650, Edegem, Belgium
| | - Tom Vanden Berghe
- Center for Inflammation Research (IRC), VIB-UGent, 9052, Zwijnaarde, Belgium
- Laboratory of Pathophysiology, University of Antwerp, 2610, Wilrijk, Belgium
| | - Erik Fransen
- StatUa Center for Statistics, University of Antwerp, 2000, Antwerp, Belgium
- Human Molecular Genetics group, Center of Medical Genetics, University of Antwerp, 2610, Wilrijk, Belgium
| | - An Wouters
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610, Wilrijk, Belgium
| | - Winnok H De Vos
- Laboratory of Cell Biology and Histology, University of Antwerp, 2610, Wilrijk, Belgium
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, 2610, Wilrijk, Belgium.
| |
Collapse
|
15
|
Abstract
Retrons are bacterial genetic elements involved in anti-phage defense. They have the unique ability to reverse transcribe RNA into multicopy single-stranded DNA (msDNA) that remains covalently linked to their template RNA. Retrons coupled with CRISPR-Cas9 in yeast have been shown to improve the efficiency of precise genome editing via homology-directed repair (HDR). In human cells, HDR editing efficiency has been limited by challenges associated with delivering extracellular donor DNA encoding the desired mutation. In this study, we tested the ability of retrons to produce msDNA as donor DNA and facilitate HDR by tethering msDNA to guide RNA in HEK293T and K562 cells. Through heterologous reconstitution of retrons from multiple bacterial species with the CRISPR-Cas9 system, we demonstrated HDR rates of up to 11.4%. Overall, our findings represent the first step in extending retron-based precise gene editing to human cells.
Collapse
Affiliation(s)
- Bin Zhao
- Department of Biology, Stanford University, Stanford, California, USA
| | - Shi-An A. Chen
- Department of Biology, Stanford University, Stanford, California, USA
| | - Jiwoo Lee
- Department of Biology, Stanford University, Stanford, California, USA
| | - Hunter B. Fraser
- Department of Biology, Stanford University, Stanford, California, USA
| |
Collapse
|
16
|
Targeting Neurons with Functional Oxytocin Receptors: A Novel Set of Simple Knock-In Mouse Lines for Oxytocin Receptor Visualization and Manipulation. eNeuro 2022; 9:ENEURO.0423-21.2022. [PMID: 35082173 PMCID: PMC8856715 DOI: 10.1523/eneuro.0423-21.2022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 01/13/2022] [Accepted: 01/14/2022] [Indexed: 12/19/2022] Open
Abstract
The neuropeptide oxytocin (Oxt) plays important roles in modulating social behaviors. Oxt receptor (Oxtr) is abundantly expressed in the brain and its relationship to socio-behavioral controls has been extensively studied using mouse brains. Several genetic tools to visualize and/or manipulate Oxtr-expressing cells, such as fluorescent reporters and Cre recombinase drivers, have been generated by ES-cell based gene targeting or bacterial artificial chromosome (BAC) transgenesis. However, these mouse lines displayed some differences in their Oxtr expression profiles probably because of the complex context and integrity of their genomic configurations in each line. Here, we apply our sophisticated genome-editing techniques to the Oxtr locus, systematically generating a series of knock-in mouse lines, in which its endogenous transcriptional regulations are intactly preserved and evaluate their expression profiles to ensure the reliability of our new tools. We employ the epitope tagging strategy, with which C-terminally fused tags can be detected by highly specific antibodies, to successfully visualize the Oxtr protein distribution on the neural membrane with super-resolution imaging for the first time. By using T2A self-cleaving peptide sequences, we also induce proper expressions of tdTomato reporter, codon-improved Cre recombinase (iCre), and spatiotemporally inducible Cre-ERT2 in Oxtr-expressing neurons. Electrophysiological recordings from tdTomato-positive cells in the reporter mice support the validity of our tool design. Retro-orbital injections of AAV-PHP.eB vector into the Cre line further enabled visualization of recombinase activities in the appropriate brain regions. Moreover, the first-time Cre-ERT2 line drives Cre-mediated recombination in a spatiotemporally controlled manner on tamoxifen (TMX) administration. These tools thus provide an excellent resource for future functional studies in Oxt-responsive neurons and should prove of broad interest in the field.
Collapse
|
17
|
Van Looveren D, Giacomazzi G, Thiry I, Sampaolesi M, Gijsbers R. Improved functionality and potency of next generation BinMLV viral vectors toward safer gene therapy. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 23:51-67. [PMID: 34553002 PMCID: PMC8433069 DOI: 10.1016/j.omtm.2021.07.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 07/16/2021] [Indexed: 10/27/2022]
Abstract
To develop safer retroviral murine leukemia virus (MLV)-based vectors, we previously mutated and re-engineered the MLV integrase: the W390A mutation abolished the interaction with its cellular tethering factors, BET proteins, and a retargeting peptide (the chromodomain of the CBX1 protein) was fused C-terminally. The resulting BET-independent MLVW390A-CBX was shown to integrate efficiently and more randomly, away from typical retroviral markers. In this study, we assessed the functionality and stability of expression of the redistributed MLVW390A-CBX vector in more depth, and evaluated safety using a clinically more relevant vector design encompassing a self-inactivated (SIN) LTR and a weak internal elongation factor 1α short (EFS) promoter. MLVW390A-CBX-EFS produced like MLVWT and efficiently transduced laboratory cells and primary human CD34+ hematopoetic stem cells (HSC) without transgene silencing over time, while displaying a more preferred, redistributed, and safer integration pattern. In a human mesoangioblast (MAB) stem cell model, the myogenic fusion capacity was hindered following MLVWT transduction, while this remained unaffected when applying MLVW390A-CBX. Likewise, smooth muscle cell differentiation of MABs was unaltered by MLVW390A-CBX-EFS. Taken together, our results underscore the potential of MLVW390A-CBX-EFS as a clinically relevant viral vector for ex-vivo gene therapy, combining efficient production with a preferable integration site distribution profile and stable expression over time.
Collapse
Affiliation(s)
- Dominique Van Looveren
- Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmacological and Pharmaceutical Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Giorgia Giacomazzi
- Laboratory of Translational Cardiomyology, Department of Development and Regeneration, Stem Cell Research Institute, KU Leuven, 3000 Leuven, Belgium
| | - Irina Thiry
- Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmacological and Pharmaceutical Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Maurilio Sampaolesi
- Laboratory of Translational Cardiomyology, Department of Development and Regeneration, Stem Cell Research Institute, KU Leuven, 3000 Leuven, Belgium
| | - Rik Gijsbers
- Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmacological and Pharmaceutical Sciences, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
18
|
Vara‐Pérez M, Rossi M, Van den Haute C, Maes H, Sassano ML, Venkataramani V, Michalke B, Romano E, Rillaerts K, Garg AD, Schepkens C, Bosisio FM, Wouters J, Oliveira AI, Vangheluwe P, Annaert W, Swinnen JV, Colet JM, van den Oord JJ, Fendt S, Mazzone M, Agostinis P. BNIP3 promotes HIF-1α-driven melanoma growth by curbing intracellular iron homeostasis. EMBO J 2021; 40:e106214. [PMID: 33932034 PMCID: PMC8126921 DOI: 10.15252/embj.2020106214] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 02/22/2021] [Accepted: 02/24/2021] [Indexed: 12/11/2022] Open
Abstract
BNIP3 is a mitophagy receptor with context-dependent roles in cancer, but whether and how it modulates melanoma growth in vivo remains unknown. Here, we found that elevated BNIP3 levels correlated with poorer melanoma patient's survival and depletion of BNIP3 in B16-F10 melanoma cells compromised tumor growth in vivo. BNIP3 depletion halted mitophagy and enforced a PHD2-mediated downregulation of HIF-1α and its glycolytic program both in vitro and in vivo. Mechanistically, we found that BNIP3-deprived melanoma cells displayed increased intracellular iron levels caused by heightened NCOA4-mediated ferritinophagy, which fostered PHD2-mediated HIF-1α destabilization. These effects were not phenocopied by ATG5 or NIX silencing. Restoring HIF-1α levels in BNIP3-depleted melanoma cells rescued their metabolic phenotype and tumor growth in vivo, but did not affect NCOA4 turnover, underscoring that these BNIP3 effects are not secondary to HIF-1α. These results unravel an unexpected role of BNIP3 as upstream regulator of the pro-tumorigenic HIF-1α glycolytic program in melanoma cells.
Collapse
Affiliation(s)
- Mónica Vara‐Pérez
- Cell Death Research and Therapy GroupDepartment of Cellular and Molecular MedicineKU LeuvenLeuvenBelgium
- VIB Center for Cancer Biology ResearchLeuvenBelgium
| | - Matteo Rossi
- VIB Center for Cancer Biology ResearchLeuvenBelgium
- Laboratory of Cellular Metabolism and Metabolic RegulationDepartment of OncologyKU Leuven and Leuven Cancer Institute (LKI)LeuvenBelgium
| | - Chris Van den Haute
- Research Group for Neurobiology and Gene TherapyDepartment of NeurosciencesKU LeuvenLeuvenBelgium
- Leuven Viral Vector CoreDepartment of NeurosciencesKU LeuvenLeuvenBelgium
| | - Hannelore Maes
- Cell Death Research and Therapy GroupDepartment of Cellular and Molecular MedicineKU LeuvenLeuvenBelgium
| | - Maria Livia Sassano
- Cell Death Research and Therapy GroupDepartment of Cellular and Molecular MedicineKU LeuvenLeuvenBelgium
- VIB Center for Cancer Biology ResearchLeuvenBelgium
| | - Vivek Venkataramani
- Institute of PathologyUniversity Medical Center Göttingen (UMG)GöttingenGermany
| | - Bernhard Michalke
- Helmholtz Zentrum München GmbH – German Research Center for Environmental HealthResearch Unit Analytical BioGeoChemistryNeuherbergGermany
| | - Erminia Romano
- Cell Death Research and Therapy GroupDepartment of Cellular and Molecular MedicineKU LeuvenLeuvenBelgium
| | - Kristine Rillaerts
- Cell Death Research and Therapy GroupDepartment of Cellular and Molecular MedicineKU LeuvenLeuvenBelgium
- VIB Center for Cancer Biology ResearchLeuvenBelgium
| | - Abhishek D Garg
- Cell Death Research and Therapy GroupDepartment of Cellular and Molecular MedicineKU LeuvenLeuvenBelgium
| | - Corentin Schepkens
- Laboratory of Lipid Metabolism and CancerDepartment of OncologyKU LeuvenLeuvenBelgium
- Human Biology and Toxicology UnitUniversity of MonsMonsBelgium
| | - Francesca M Bosisio
- Laboratory of Translational Cell and Tissue ResearchDepartment of Imaging and PathologyKU LeuvenLeuvenBelgium
| | - Jasper Wouters
- Laboratory of Translational Cell and Tissue ResearchDepartment of Imaging and PathologyKU LeuvenLeuvenBelgium
| | - Ana Isabel Oliveira
- VIB Center for Cancer Biology ResearchLeuvenBelgium
- Laboratory of Tumor Inflammation and AngiogenesisDepartment of OncologyKU LeuvenLeuvenBelgium
| | - Peter Vangheluwe
- Laboratory of Cellular Transport SystemsDepartment of Cellular and Molecular MedicineKU LeuvenLeuvenBelgium
| | - Wim Annaert
- Laboratory for Membrane TraffickingDepartment of NeurosciencesKU LeuvenLeuvenBelgium
- VIB Center for Brain and Disease ResearchLeuvenBelgium
| | - Johannes V Swinnen
- Laboratory of Lipid Metabolism and CancerDepartment of OncologyKU LeuvenLeuvenBelgium
| | | | - Joost J van den Oord
- Laboratory of Translational Cell and Tissue ResearchDepartment of Imaging and PathologyKU LeuvenLeuvenBelgium
| | - Sarah‐Maria Fendt
- VIB Center for Cancer Biology ResearchLeuvenBelgium
- Laboratory of Cellular Metabolism and Metabolic RegulationDepartment of OncologyKU Leuven and Leuven Cancer Institute (LKI)LeuvenBelgium
| | - Massimiliano Mazzone
- VIB Center for Cancer Biology ResearchLeuvenBelgium
- Laboratory of Tumor Inflammation and AngiogenesisDepartment of OncologyKU LeuvenLeuvenBelgium
| | - Patrizia Agostinis
- Cell Death Research and Therapy GroupDepartment of Cellular and Molecular MedicineKU LeuvenLeuvenBelgium
- VIB Center for Cancer Biology ResearchLeuvenBelgium
| |
Collapse
|
19
|
van den Akker GGH, Zacchini F, Housmans BAC, van der Vloet L, Caron MMJ, Montanaro L, Welting TJM. Current Practice in Bicistronic IRES Reporter Use: A Systematic Review. Int J Mol Sci 2021; 22:5193. [PMID: 34068921 PMCID: PMC8156625 DOI: 10.3390/ijms22105193] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/05/2021] [Accepted: 05/12/2021] [Indexed: 12/26/2022] Open
Abstract
Bicistronic reporter assays have been instrumental for transgene expression, understanding of internal ribosomal entry site (IRES) translation, and identification of novel cap-independent translational elements (CITE). We observed a large methodological variability in the use of bicistronic reporter assays and data presentation or normalization procedures. Therefore, we systematically searched the literature for bicistronic IRES reporter studies and analyzed methodological details, data visualization, and normalization procedures. Two hundred fifty-seven publications were identified using our search strategy (published 1994-2020). Experimental studies on eukaryotic adherent cell systems and the cell-free translation assay were included for further analysis. We evaluated the following methodological details for 176 full text articles: the bicistronic reporter design, the cell line or type, transfection methods, and time point of analyses post-transfection. For the cell-free translation assay, we focused on methods of in vitro transcription, type of translation lysate, and incubation times and assay temperature. Data can be presented in multiple ways: raw data from individual cistrons, a ratio of the two, or fold changes thereof. In addition, many different control experiments have been suggested when studying IRES-mediated translation. In addition, many different normalization and control experiments have been suggested when studying IRES-mediated translation. Therefore, we also categorized and summarized their use. Our unbiased analyses provide a representative overview of bicistronic IRES reporter use. We identified parameters that were reported inconsistently or incompletely, which could hamper data reproduction and interpretation. On the basis of our analyses, we encourage adhering to a number of practices that should improve transparency of bicistronic reporter data presentation and improve methodological descriptions to facilitate data replication.
Collapse
Affiliation(s)
- Guus Gijsbertus Hubert van den Akker
- Department of Orthopedic Surgery, Maastricht University, Medical Center+, 6229 ER Maastricht, The Netherlands; (G.G.H.v.d.A.); (B.A.C.H.); (L.v.d.V.); (M.M.J.C.)
| | - Federico Zacchini
- Department of Experimental, Diagnostic and Specialty Medicine, Bologna University, I-40138 Bologna, Italy; (F.Z.); (L.M.)
- Centro di Ricerca Biomedica Applicata—CRBA, Bologna University, Policlinico di Sant’Orsola, I-40138 Bologna, Italy
| | - Bas Adrianus Catharina Housmans
- Department of Orthopedic Surgery, Maastricht University, Medical Center+, 6229 ER Maastricht, The Netherlands; (G.G.H.v.d.A.); (B.A.C.H.); (L.v.d.V.); (M.M.J.C.)
| | - Laura van der Vloet
- Department of Orthopedic Surgery, Maastricht University, Medical Center+, 6229 ER Maastricht, The Netherlands; (G.G.H.v.d.A.); (B.A.C.H.); (L.v.d.V.); (M.M.J.C.)
| | - Marjolein Maria Johanna Caron
- Department of Orthopedic Surgery, Maastricht University, Medical Center+, 6229 ER Maastricht, The Netherlands; (G.G.H.v.d.A.); (B.A.C.H.); (L.v.d.V.); (M.M.J.C.)
| | - Lorenzo Montanaro
- Department of Experimental, Diagnostic and Specialty Medicine, Bologna University, I-40138 Bologna, Italy; (F.Z.); (L.M.)
- Centro di Ricerca Biomedica Applicata—CRBA, Bologna University, Policlinico di Sant’Orsola, I-40138 Bologna, Italy
- Programma Dipartimentale in Medicina di Laboratorio, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Via Albertoni 15, I-40138 Bologna, Italy
| | - Tim Johannes Maria Welting
- Department of Orthopedic Surgery, Maastricht University, Medical Center+, 6229 ER Maastricht, The Netherlands; (G.G.H.v.d.A.); (B.A.C.H.); (L.v.d.V.); (M.M.J.C.)
| |
Collapse
|
20
|
Pathogenic LRRK2 requires secondary factors to induce cellular toxicity. Biosci Rep 2021; 40:226517. [PMID: 32975566 PMCID: PMC7560525 DOI: 10.1042/bsr20202225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 09/08/2020] [Accepted: 09/24/2020] [Indexed: 12/02/2022] Open
Abstract
Pathogenic mutations in the leucine-rich repeat kinase 2 (LRRK2) gene belong to the most common genetic causes of inherited Parkinson’s disease (PD) and variations in its locus increase the risk to develop sporadic PD. Extensive research efforts aimed at understanding how changes in the LRRK2 function result in molecular alterations that ultimately lead to PD. Cellular LRRK2-based models revealed several potential pathophysiological mechanisms including apoptotic cell death, LRRK2 protein accumulation and deficits in neurite outgrowth. However, highly variable outcomes between different cellular models have been reported. Here, we have investigated the effect of different experimental conditions, such as the use of different tags and gene transfer methods, in various cellular LRRK2 models. Readouts included cell death, sensitivity to oxidative stress, LRRK2 relocalization, α-synuclein aggregation and neurite outgrowth in cell culture, as well as neurite maintenance in vivo. We show that overexpression levels and/or the tag fused to LRRK2 affect the relocalization of LRRK2 to filamentous and skein-like structures. We found that overexpression of LRRK2 per se is not sufficient to induce cellular toxicity or to affect α-synuclein-induced toxicity and aggregate formation. Finally, neurite outgrowth/retraction experiments in cell lines and in vivo revealed that secondary, yet unknown, factors are required for the pathogenic LRRK2 effects on neurite length. Our findings stress the importance of technical and biological factors in LRRK2-induced cellular phenotypes and hence imply that conclusions based on these types of LRRK2-based assays should be interpreted with caution.
Collapse
|
21
|
Lamkin DM, Bradshaw KP, Chang J, Epstein M, Gomberg J, Prajapati KP, Soliman VH, Sylviana T, Wong Y, Morizono K, Sloan EK, Cole SW. Physical activity modulates mononuclear phagocytes in mammary tissue and inhibits tumor growth in mice. PeerJ 2021; 9:e10725. [PMID: 33552733 PMCID: PMC7821756 DOI: 10.7717/peerj.10725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 12/16/2020] [Indexed: 12/29/2022] Open
Abstract
The risk for breast cancer is significantly reduced in persons who engage in greater amounts of physical activity, and greater physical activity before or after diagnosis associates with reduced disease-specific mortality. Previous mechanistic studies indicate that components of innate immunity can mediate an inhibitory effect of physical activity on several types of tumor. However, in breast cancer specifically, the myeloid compartment of innate immunity is thought to exhibit high propensity for an immunosuppressive role that obstructs anti-tumor immunity. Thus, we tested the notion that greater physical activity alters mononuclear phagocytes in mammary tissue when inhibiting nascent tumor in a murine model of breast cancer. To model greater physical activity, we placed an angled running wheel in each mouse's home cage for two weeks before tumor engraftment with EO771 mammary cancer cells that express luciferase for bioluminescent detection. Fully immunocompetent mice and mice with compromised adaptive immunity showed significantly less mammary tumor signal when given access to running wheels, although the effect size was smaller in this latter group. To investigate the role of the myeloid compartment, mononuclear phagocytes were ablated by systemic injection of clodronate liposomes at 24 h before tumor engraftment and again at the time of tumor engraftment, and this treatment reversed the inhibition in wheel running mice. However, clodronate also inhibited mammary tumor signal in sedentary mice, in conjunction with an expected decrease in gene and protein expression of the myeloid antigen, F4/80 (Adgre1), in mammary tissue. Whole transcriptome digital cytometry with CIBERSORTx was used to analyze myeloid cell populations in mammary tissue following voluntary wheel running and clodronate treatment, and this approach found significant changes in macrophage and monocyte populations. In exploratory analyses, whole transcriptome composite scores for monocytic myeloid-derived suppressor cell (M-MDSC), macrophage lactate timer, and inflammation resolution gene expression programs were significantly altered. Altogether, the results support the hypothesis that physical activity inhibits nascent mammary tumor growth by enhancing the anti-tumor potential of mononuclear phagocytes in mammary tissue.
Collapse
Affiliation(s)
- Donald M. Lamkin
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles, CA, United States of America
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, CA, United States of America
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, United States of America
| | - Karen P. Bradshaw
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles, CA, United States of America
- Department of Neuroscience, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Janice Chang
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles, CA, United States of America
| | - Ma’ayan Epstein
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles, CA, United States of America
| | - Jack Gomberg
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles, CA, United States of America
| | - Krupa P. Prajapati
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles, CA, United States of America
| | - Veronica H. Soliman
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles, CA, United States of America
| | - Thezia Sylviana
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles, CA, United States of America
| | - Yinnie Wong
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles, CA, United States of America
| | - Kouki Morizono
- Divison of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, United States of America
- UCLA AIDS Institute, David Geffen School of Medicine, University of California, Los Angeles, CA, United States of America
| | - Erica K. Sloan
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles, CA, United States of America
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, United States of America
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Division of Cancer Surgery, Peter MacCallum Cancer Centre-Victorian Comprehensive Cancer Centre, Melbourne, Victoria, Austalia
| | - Steve W. Cole
- Norman Cousins Center for PNI, Semel Institute for Neuroscience, University of California, Los Angeles, CA, United States of America
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, CA, United States of America
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, United States of America
- Divison of Hematology-Oncology, Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA, United States of America
| |
Collapse
|
22
|
RAJASEKARAN RANJANI, KIRUBAHARAN JJOHN, SHILPA P, VIDHYA M, RAJALAKSHMI S. Viral 2A-peptides mediate continuous transcription and self-cleavage of multiple heterologous genes in fowlpox virus vector. THE INDIAN JOURNAL OF ANIMAL SCIENCES 2021. [DOI: 10.56093/ijans.v90i9.109445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Applicability of viral 2A-peptides in generation of multi-cistronic transcripts to deliver separate self-cleaved proteins is well established. However, the use of viral 2A-peptides in fowlpox virus vector construction to co-express multiple heterologous genes has not been explored. To evaluate the same, a recombinant transfer plasmid pJFWPVt was constructed through two intermediate plasmid constructs, pJF7F9 and pJFHNGFP. The construction of pJF7F9 involved cloning of F7 and F9 genes of FWPV into pCI-neo with modifications in the F7-F9 intergenic region. For the construction of pJFHNGFP, a synthetic DNA adapter consisting of one synthetic early late promoter (PE/L), two viral 2A-peptides (P2A and T2A) and three multiple cloning sites (MCS1, MCS2 and MCS3) was synthesized chemically and was cloned into pUC19 to obtain pJFHNGFPi. Heterologous genes fusion (F) and haemagglutininneuraminidase (HN) of Avian Avulavirus-1 (AAv1) and marker gene AcGFP were cloned sequentially into MCS1, MCS2 and MCS3 of pJFHNGFPi to obtain pJFHNGFP. The insert (PE/L-F-P2A-HN-T2A-AcGFP) in pJFHNGFP was cloned into pJF7F9 to obtain pJFWPVt, which upon transfection in FWPV infected chicken embryo fibroblast (CEF) cells resulted in fluorescence. This confirmed the expression of AcGFP and the continuous transcription ability of viral 2A-peptides. Further, western blotting of CEF pellet showed separate protein bands of F and HN protein at 66 kDa and 74 kDa respectively, which confirmed the self-cleaving ability of viral 2A-peptides. Herein, in FWPV vector construction, continuous transcription and self-cleaving ability of viral 2A-peptides in FWPV vector construction was confirmed. This warrants scope for future viral 2A-peptide based FWPV vector construction.
Collapse
|
23
|
CRISPR-mediated knock-in in the mouse embryo using long single stranded DNA donors synthesised by biotinylated PCR. Methods 2020; 191:3-14. [PMID: 33172594 DOI: 10.1016/j.ymeth.2020.10.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 09/21/2020] [Accepted: 10/18/2020] [Indexed: 12/26/2022] Open
Abstract
Successful gene knock-in by CRISPR-Cas9 in the mouse zygote requires three components; guideRNA, Cas9 protein and a suitable donor template, which usually comprises homology flanked insert sequence. Recently, long single stranded DNA (lssDNA) donors have emerged as a popular choice of DNA donor, outperforming dsDNA templates in terms of knock-in efficiency for gene tagging and generating conditional alleles. The generation of these donors can be achieved through several methods that may introduce errors in the sequence, result in poor yields, and contain dsDNA contamination. We have developed our own cost-effective lssDNA synthesis methodology that results in high purity, sequence verified, low contamination lssDNA donors. We provide a detailed methodology on the design and generation of such donors for gene tagging experiments and generating conditional alleles.
Collapse
|
24
|
Savino AM, Fernandes SI, Olivares O, Zemlyansky A, Cousins A, Markert EK, Barel S, Geron I, Frishman L, Birger Y, Eckert C, Tumanov S, MacKay G, Kamphorst JJ, Herzyk P, Fernández-García J, Abramovich I, Mor I, Bardini M, Barin E, Janaki-Raman S, Cross JR, Kharas MG, Gottlieb E, Izraeli S, Halsey C. Metabolic adaptation of acute lymphoblastic leukemia to the central nervous system microenvironment is dependent on Stearoyl CoA desaturase. NATURE CANCER 2020; 1:998-1009. [PMID: 33479702 PMCID: PMC7116605 DOI: 10.1038/s43018-020-00115-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 08/14/2020] [Indexed: 02/07/2023]
Abstract
Metabolic reprogramming is a key hallmark of cancer, but less is known about metabolic plasticity of the same tumor at different sites. Here, we investigated the metabolic adaptation of leukemia in two different microenvironments, the bone marrow and the central nervous system (CNS). We identified a metabolic signature of fatty-acid synthesis in CNS leukemia, highlighting Stearoyl-CoA desaturase (SCD1) as a key player. In vivo SCD1 overexpression increases CNS disease, whilst genetic or pharmacological inhibition of SCD1 decreases CNS load. Overall, we demonstrated that leukemic cells dynamically rewire metabolic pathways to suit local conditions and that targeting these adaptations can be exploited therapeutically.
Collapse
Affiliation(s)
- Angela Maria Savino
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sheba Medical Center, Ramat Gan, Israel
- Molecular Pharmacology Program, Center for Cell Engineering, Center for Stem Cell Biology, Center for Experimental Therapeutics, Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sara Isabel Fernandes
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Orianne Olivares
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Anna Zemlyansky
- Schneider Children's Medical Center of Israel, Petach Tiqva, Israel
| | - Antony Cousins
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Elke K Markert
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Cancer Research UK Beatson Institute, Glasgow, UK
| | - Shani Barel
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sheba Medical Center, Ramat Gan, Israel
| | - Ifat Geron
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sheba Medical Center, Ramat Gan, Israel
| | - Liron Frishman
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sheba Medical Center, Ramat Gan, Israel
| | - Yehudit Birger
- Sheba Medical Center, Ramat Gan, Israel
- Schneider Children's Medical Center of Israel, Petach Tiqva, Israel
| | | | | | | | - Jurre J Kamphorst
- Cancer Research UK Beatson Institute, Glasgow, UK
- Rheos Medicines, Cambridge, MA, USA
| | - Pawel Herzyk
- Glasgow Polyomics, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Institute of Molecular, Cell and Systems Biology, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Jonatan Fernández-García
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Ifat Abramovich
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Inbal Mor
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Michela Bardini
- Centro Ricerca Tettamanti, Fondazione MBBM, Universita degli Studi di Milano-Bicocca, Monza, Italy
| | - Ersilia Barin
- Molecular Pharmacology Program, Center for Cell Engineering, Center for Stem Cell Biology, Center for Experimental Therapeutics, Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sudha Janaki-Raman
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Justin R Cross
- Donald B. and Catherine C. Marron Cancer Metabolism Center, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael G Kharas
- Molecular Pharmacology Program, Center for Cell Engineering, Center for Stem Cell Biology, Center for Experimental Therapeutics, Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Eyal Gottlieb
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.
| | - Shai Izraeli
- Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
- Sheba Medical Center, Ramat Gan, Israel.
- Schneider Children's Medical Center of Israel, Petach Tiqva, Israel.
- Beckman Research Institute, City of Hope, Duarte, CA, USA.
| | - Christina Halsey
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
25
|
MICAL2 is essential for myogenic lineage commitment. Cell Death Dis 2020; 11:654. [PMID: 32811811 PMCID: PMC7434877 DOI: 10.1038/s41419-020-02886-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/29/2020] [Accepted: 07/29/2020] [Indexed: 01/25/2023]
Abstract
Contractile myofiber units are mainly composed of thick myosin and thin actin (F-actin) filaments. F-Actin interacts with Microtubule Associated Monooxygenase, Calponin And LIM Domain Containing 2 (MICAL2). Indeed, MICAL2 modifies actin subunits and promotes actin filament turnover by severing them and preventing repolymerization. In this study, we found that MICAL2 increases during myogenic differentiation of adult and pluripotent stem cells (PSCs) towards skeletal, smooth and cardiac muscle cells and localizes in the nucleus of acute and chronic regenerating muscle fibers. In vivo delivery of Cas9–Mical2 guide RNA complexes results in muscle actin defects and demonstrates that MICAL2 is essential for skeletal muscle homeostasis and functionality. Conversely, MICAL2 upregulation shows a positive impact on skeletal and cardiac muscle commitments. Taken together these data demonstrate that modulations of MICAL2 have an impact on muscle filament dynamics and its fine-tuned balance is essential for the regeneration of muscle tissues.
Collapse
|
26
|
Gholobova D, Terrie L, Mackova K, Desender L, Carpentier G, Gerard M, Hympanova L, Deprest J, Thorrez L. Functional evaluation of prevascularization in one-stage versus two-stage tissue engineering approach of human bio-artificial muscle. Biofabrication 2020; 12:035021. [PMID: 32357347 DOI: 10.1088/1758-5090/ab8f36] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
A common shortcoming of current tissue engineered constructs is the lack of a functional vasculature, limiting their size and functionality. Prevascularization is a possible strategy to introduce vascular networks in these constructs. It includes among others co-culturing target cells with endothelial (precursor) cells that are able to form endothelial networks through vasculogenesis. In this paper, we compared two different prevascularization approaches of bio-artificial skeletal muscle tissue (BAM) in vitro and in vivo. In a one-stage approach, human muscle cells were directly co-cultured with endothelial cells in 3D. In a two-stage approach, a one week old BAM containing differentiated myotubes was coated with a fibrin hydrogel containing endothelial cells. The obtained endothelial networks were longer and better interconnected with the two-stage approach. We evaluated whether prevascularization had a beneficial effect on in vivo perfusion of the BAM and improved myotube survival by implantation on the fascia of the latissimus dorsi muscle of NOD/SCID mice for 5 or 14 d. Also in vivo, the two-stage approach displayed the highest vascular density. At day 14, anastomosis of implanted endothelial networks with the host vasculature was apparent. BAMs without endothelial networks contained longer and thicker myotubes in vitro, but their morphology degraded in vivo. In contrast, maintenance of myotube morphology was well supported in the two-stage prevascularized BAMs. To conclude, a two-stage prevascularization approach for muscle engineering improved the vascular density in the construct and supported myotube maintenance in vivo.
Collapse
Affiliation(s)
- D Gholobova
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven, E. Sabbelaan 53, 8500, Kortrijk, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Sorrentino A, Eroglu E, Michel T. In vivo applications of chemogenetics in redox (patho)biology. OXIDATIVE STRESS 2020:97-112. [DOI: 10.1016/b978-0-12-818606-0.00007-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
28
|
|
29
|
Debie P, Lafont C, Defrise M, Hansen I, van Willigen DM, van Leeuwen FWB, Gijsbers R, D'Huyvetter M, Devoogdt N, Lahoutte T, Mollard P, Hernot S. Size and affinity kinetics of nanobodies influence targeting and penetration of solid tumours. J Control Release 2019; 317:34-42. [PMID: 31734445 DOI: 10.1016/j.jconrel.2019.11.014] [Citation(s) in RCA: 117] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 11/10/2019] [Accepted: 11/12/2019] [Indexed: 11/30/2022]
Abstract
A compound's intratumoural distribution is an important determinant for the effectiveness of molecular therapy or imaging. Antibodies (Abs), though often used in the design of targeted compounds, struggle to achieve a homogenous distribution due to their large size and bivalent binding mechanism. In contrast, smaller compounds like nanobodies (Nbs) are expected to distribute more homogenously, though this has yet to be demonstrated in vivo at the microscopic level. We propose an intravital approach to evaluate the intratumoural distribution of different fluorescently labeled monomeric and dimeric Nb tracers and compare this with a monoclonal antibody (mAb). Monomeric and dimeric formats of the anti-HER2 (2Rb17c and 2Rb17c-2Rb17c) and control (R3B23 and R3B23-R3B23) Nb, as well as the dimeric monovalent Nb 2Rb17c-R3B23 were generated and fluorescently labeled with a Cy5 fluorophore. The mAb trastuzumab-Cy5 was also prepared. Whole-body biodistribution of all constructs was investigated in mice bearing subcutaneous xenografts (HER2+ SKOV3) using in vivo epi-fluorescence imaging. Next, for intravital experiments, GFP-expressing SKOV3 cells were grown under dorsal window chambers on athymic nude mice (n = 3/group), and imaged under a fluorescence stereo microscope immediately after intravenous injection of the tracers. Consecutive fluorescence images within the tumour were acquired over the initial 20 min after injection and later, single images were taken at 1, 3 and 24 h post-injection. Additionally, two-photon microscopy was used to investigate the colocalization of GFP (tumour cells) and Cy5 fluorescence (tracers) at higher resolution. Whole-body images showed rapid renal clearance of all Nbs, and fast tumour targeting for the specific Nbs. Specific tumour uptake of the mAb could only be clearly distinguished from background after several hours. Intravital imaging revealed that monomeric Nb tracers accumulated rapidly and distributed homogenously in the tumour mere minutes after intravenous injection. The dimeric compounds initially achieved lower fluorescence intensities than the monomeric. Furthermore, whereas the HER2-specific dimeric bivalent compound remained closely associated to the blood vessels over 24 h, the HER2-specific dimeric monovalent tracer achieved a more homogenous tumour distribution from 1 h post-injection onwards. Non-specific tracers were not retained in the tumour. Trastuzumab had the most heterogenous intratumoural distribution of all evaluated compounds, while -due to the long blood retention- achieving the highest overall tumour uptake at 24 h post-injection. In conclusion, monomeric Nbs very quickly and homogenously distribute through tumour tissue, at a rate significantly greater than dimeric Nbs and mAbs. This underlines the potential of monomeric Nb tracers and therapeutics in molecular imaging and targeted therapies.
Collapse
Affiliation(s)
- Pieterjan Debie
- Laboratory for In vivo Cellular and Molecular Imaging, ICMI-BEFY, Vrije Universiteit Brussel, Brussels, Belgium.
| | - Chrystel Lafont
- Institut de Génomique Fonctionnelle, CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - Michel Defrise
- Laboratory for In vivo Cellular and Molecular Imaging, ICMI-BEFY, Vrije Universiteit Brussel, Brussels, Belgium
| | - Inge Hansen
- Laboratory for In vivo Cellular and Molecular Imaging, ICMI-BEFY, Vrije Universiteit Brussel, Brussels, Belgium
| | - Danny M van Willigen
- Interventional Molecular Imaging laboratory, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Fijs W B van Leeuwen
- Interventional Molecular Imaging laboratory, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Rik Gijsbers
- Laboratory for Molecular Virology and Gene therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium
| | - Matthias D'Huyvetter
- Laboratory for In vivo Cellular and Molecular Imaging, ICMI-BEFY, Vrije Universiteit Brussel, Brussels, Belgium
| | - Nick Devoogdt
- Laboratory for In vivo Cellular and Molecular Imaging, ICMI-BEFY, Vrije Universiteit Brussel, Brussels, Belgium
| | - Tony Lahoutte
- Laboratory for In vivo Cellular and Molecular Imaging, ICMI-BEFY, Vrije Universiteit Brussel, Brussels, Belgium; Department of Nuclear Medicine, UZBrussel, Brussels, Belgium
| | - Patrice Mollard
- Institut de Génomique Fonctionnelle, CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - Sophie Hernot
- Laboratory for In vivo Cellular and Molecular Imaging, ICMI-BEFY, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
30
|
Tielemans B, Stoian L, Gijsbers R, Michiels A, Wagenaar A, Farre Marti R, Belge C, Delcroix M, Quarck R. Cytokines trigger disruption of endothelium barrier function and p38 MAP kinase activation in BMPR2-silenced human lung microvascular endothelial cells. Pulm Circ 2019; 9:2045894019883607. [PMID: 31692724 PMCID: PMC6811766 DOI: 10.1177/2045894019883607] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 09/24/2019] [Indexed: 12/21/2022] Open
Abstract
The bone morphogenetic protein receptor II (BMPRII) signaling pathway is impaired
in pulmonary arterial hypertension and mutations in the BMPR2
gene have been observed in both heritable and idiopathic pulmonary arterial
hypertension. However, all BMPR2 mutation carriers do not
develop pulmonary arterial hypertension, and inflammation could trigger the
development of the disease in BMPR2 mutation carriers.
Circulating levels and/or lung tissue expression of cytokines such as tumor
necrosis factor-α or interleukin-18 are elevated in patients with pulmonary
arterial hypertension and could be involved in the pathogenesis of pulmonary
arterial hypertension. We consequently hypothesized that cytokines could trigger
endothelial dysfunction in addition to impaired BMPRII signaling. Our aim was to
determine whether impairment of BMPRII signaling might affect endothelium
barrier function and adhesiveness to monocytes, in response to cytokines.
BMPR2 was silenced in human lung microvascular endothelial
cells (HLMVECs) using lentiviral vectors encoding microRNA-based hairpins.
Effects of tumor necrosis factor-α and interleukin-18 on HLMVEC adhesiveness to
the human monocyte cell line THP-1, adhesion molecule expression, endothelial
barrier function and activation of P38MAPK were investigated in vitro. Stable
BMPR2 silencing in HLMVECs resulted in impaired endothelial
barrier function and constitutive activation of P38MAPK. Adhesiveness of
BMPR2-silenced HLMVECs to THP-1 cells was enhanced by tumor
necrosis factor-α and interleukin-18 through ICAM-1 adhesion molecule.
Interestingly, tumor necrosis factor-α induced activation of P38MAPK and
disrupted endothelial barrier function in BMPR2-silenced
HLMVECs. Altogether, our findings showed that stable BMPR2
silencing resulted in impaired endothelial barrier function and activation of
P38MAPK in HLMVECs. In BMPR2-silenced HLMVECs, cytokines
enhanced adhesiveness capacities, activation of P38MAPK and impaired endothelial
barrier function suggesting that cytokines could trigger the development of
pulmonary arterial hypertension in a context of impaired BMPRII signaling
pathway.
Collapse
Affiliation(s)
- Birger Tielemans
- Division of Respiratory Diseases, Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium
| | - Leanda Stoian
- Division of Respiratory Diseases, Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium
| | - Rik Gijsbers
- Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven - University of Leuven, Leuven, Belgium.,Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven - University of Leuven, Leuven, Belgium
| | - Annelies Michiels
- Neurobiology and Gene Therapy, Department of Neurosciences, KU Leuven - University of Leuven, Leuven, Belgium.,Leuven Viral Vector Core, KU Leuven - University of Leuven, Leuven, Belgium
| | - Allard Wagenaar
- Division of Respiratory Diseases, Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium
| | - Ricard Farre Marti
- Translational Research in Gastrointestinal Disorders, Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium
| | - Catharina Belge
- Division of Respiratory Diseases, University Hospitals and Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium
| | - Marion Delcroix
- Division of Respiratory Diseases, University Hospitals and Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium
| | - Rozenn Quarck
- Division of Respiratory Diseases, University Hospitals and Department of Chronic Diseases, Metabolism & Ageing (CHROMETA), KU Leuven - University of Leuven, Leuven, Belgium
| |
Collapse
|
31
|
Abdulhaqq SA, Wu H, Schell JB, Hammond KB, Reed JS, Legasse AW, Axthelm MK, Park BS, Asokan A, Früh K, Hansen SG, Picker LJ, Sacha JB. Vaccine-Mediated Inhibition of the Transporter Associated with Antigen Processing Is Insufficient To Induce Major Histocompatibility Complex E-Restricted CD8 + T Cells in Nonhuman Primates. J Virol 2019; 93:e00592-19. [PMID: 31315990 PMCID: PMC6744250 DOI: 10.1128/jvi.00592-19] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 07/08/2019] [Indexed: 01/28/2023] Open
Abstract
Major histocompatibility complex E (MHC-E) is a highly conserved nonclassical MHC-Ib molecule that tightly binds peptides derived from leader sequences of classical MHC-Ia molecules for presentation to natural killer cells. However, MHC-E also binds diverse foreign and neoplastic self-peptide antigens for presentation to CD8+ T cells. Although the determinants of MHC-E-restricted T cell priming remain unknown, these cells are induced in humans infected with pathogens containing genes that inhibit the transporter associated with antigen processing (TAP). Indeed, mice vaccinated with TAP-inhibited autologous dendritic cells develop T cells restricted by the murine MHC-E homologue, Qa-1b. Here, we tested whether rhesus macaques (RM) vaccinated with viral constructs expressing a TAP inhibitor would develop insert-specific MHC-E-restricted CD8+ T cells. We generated viral constructs coexpressing SIVmac239 Gag in addition to one of three TAP inhibitors: herpes simplex virus 2 ICP47, bovine herpes virus 1 UL49.5, or rhesus cytomegalovirus Rh185. Each TAP inhibitor reduced surface expression of MHC-Ia molecules but did not reduce surface MHC-E expression. In agreement with modulation of surface MHC-Ia levels, TAP inhibition diminished presentation of MHC-Ia-restricted CD8+ T cell epitopes without impacting presentation of peptide antigen bound by MHC-E. Vaccination of macaques with vectors dually expressing SIVmac239 Gag with ICP47, UL49.5, or Rh185 generated Gag-specific CD8+ T cells classically restricted by MHC-Ia but not MHC-E. These data demonstrate that, in contrast to results in mice, TAP inhibition alone is insufficient for priming of MHC-E-restricted T cell responses in primates and suggest that additional unknown mechanisms govern the induction of CD8+ T cells recognizing MHC-E-bound antigen.IMPORTANCE Due to the near monomorphic nature of MHC-E in the human population and inability of many pathogens to inhibit MHC-E-mediated peptide presentation, MHC-E-restricted T cells have become an attractive vaccine target. However, little is known concerning how these cells are induced. Understanding the underlying mechanisms that induce these T cells would provide a powerful new vaccine strategy to an array of neoplasms and viral and bacterial pathogens. Recent studies have indicated a link between TAP inhibition and induction of MHC-E-restricted T cells. The significance of our research is in demonstrating that TAP inhibition alone does not prime MHC-E-restricted T cell generation and suggests that other, currently unknown mechanisms regulate their induction.
Collapse
Affiliation(s)
- Shaheed A Abdulhaqq
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Helen Wu
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - John B Schell
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Katherine B Hammond
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Jason S Reed
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Alfred W Legasse
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Michael K Axthelm
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Byung S Park
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Aravind Asokan
- Department of Surgery, Duke University, Durham, North Carolina, USA
| | - Klaus Früh
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Scott G Hansen
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Louis J Picker
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
| | - Jonah B Sacha
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Beaverton, Oregon, USA
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Oregon, USA
| |
Collapse
|
32
|
Hernandez N, Bucciol G, Moens L, Le Pen J, Shahrooei M, Goudouris E, Shirkani A, Changi-Ashtiani M, Rokni-Zadeh H, Sayar EH, Reisli I, Lefevre-Utile A, Zijlmans D, Jurado A, Pholien R, Drutman S, Belkaya S, Cobat A, Boudewijns R, Jochmans D, Neyts J, Seeleuthner Y, Lorenzo-Diaz L, Enemchukwu C, Tietjen I, Hoffmann HH, Momenilandi M, Pöyhönen L, Siqueira MM, de Lima SMB, de Souza Matos DC, Homma A, Maia MDLS, da Costa Barros TA, de Oliveira PMN, Mesquita EC, Gijsbers R, Zhang SY, Seligman SJ, Abel L, Hertzog P, Marr N, Martins RDM, Meyts I, Zhang Q, MacDonald MR, Rice CM, Casanova JL, Jouanguy E, Bossuyt X. Inherited IFNAR1 deficiency in otherwise healthy patients with adverse reaction to measles and yellow fever live vaccines. J Exp Med 2019; 216:2057-2070. [PMID: 31270247 PMCID: PMC6719432 DOI: 10.1084/jem.20182295] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 03/18/2019] [Accepted: 06/11/2019] [Indexed: 01/31/2023] Open
Abstract
We describe two unrelated patients with inherited IFNAR1 deficiency who suffered from life-threatening infections following measles or yellow fever virus vaccination and were otherwise healthy. Vaccination against measles, mumps, and rubella (MMR) and yellow fever (YF) with live attenuated viruses can rarely cause life-threatening disease. Severe illness by MMR vaccines can be caused by inborn errors of type I and/or III interferon (IFN) immunity (mutations in IFNAR2, STAT1, or STAT2). Adverse reactions to the YF vaccine have remained unexplained. We report two otherwise healthy patients, a 9-yr-old boy in Iran with severe measles vaccine disease at 1 yr and a 14-yr-old girl in Brazil with viscerotropic disease caused by the YF vaccine at 12 yr. The Iranian patient is homozygous and the Brazilian patient compound heterozygous for loss-of-function IFNAR1 variations. Patient-derived fibroblasts are susceptible to viruses, including the YF and measles virus vaccine strains, in the absence or presence of exogenous type I IFN. The patients’ fibroblast phenotypes are rescued with WT IFNAR1. Autosomal recessive, complete IFNAR1 deficiency can result in life-threatening complications of vaccination with live attenuated measles and YF viruses in previously healthy individuals.
Collapse
Affiliation(s)
- Nicholas Hernandez
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY
| | - Giorgia Bucciol
- Laboratory of Inborn Errors of Immunity, Department of Immunology, Microbiology and Transplantation, KU Leuven, Leuven, Belgium
| | - Leen Moens
- Laboratory of Inborn Errors of Immunity, Department of Immunology, Microbiology and Transplantation, KU Leuven, Leuven, Belgium
| | - Jérémie Le Pen
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY
| | - Mohammad Shahrooei
- Specialized Immunology Laboratory of Dr. Shahrooei, Sina Medical Complex, Ahvaz, Iran.,Department of Microbiology and Immunology, Clinical and Diagnostic Immunology, KU Leuven, Leuven, Belgium
| | | | - Afshin Shirkani
- Allergy and Clinical Immunology Department, Bushehr University of Medical Science, School of Medicine, Bushehr, Iran
| | | | - Hassan Rokni-Zadeh
- Department of Medical Biotechnology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Esra Hazar Sayar
- Department of Pediatrics, Division of Pediatric Immunology and Allergy, Necmettin Erbakan University, Meram Medical Faculty, Konya, Turkey
| | - Ismail Reisli
- Department of Pediatrics, Division of Pediatric Immunology and Allergy, Necmettin Erbakan University, Meram Medical Faculty, Konya, Turkey
| | - Alain Lefevre-Utile
- Pediatrics Department, Jean Verdier Hospital, Assistance Publique des Hôpitaux de Paris, Paris 13 University, Bondy, France
| | - Dick Zijlmans
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY
| | - Andrea Jurado
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY
| | - Ruben Pholien
- Laboratory of Virology and Chemotherapy, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Scott Drutman
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY
| | - Serkan Belkaya
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY
| | - Aurelie Cobat
- Pediatric Immunology-Hematology Unit, Assistance Publique-Hôpitaux de Paris, Necker Hospital for Sick Children, Paris, France
| | - Robbert Boudewijns
- Laboratory of Virology and Chemotherapy, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Dirk Jochmans
- Laboratory of Virology and Chemotherapy, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Johan Neyts
- Laboratory of Virology and Chemotherapy, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Yoann Seeleuthner
- Paris Descartes University, Imagine Institute, Paris, France.,Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Paris, France
| | - Lazaro Lorenzo-Diaz
- Paris Descartes University, Imagine Institute, Paris, France.,Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Paris, France
| | - Chibuzo Enemchukwu
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY
| | - Ian Tietjen
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY
| | | | - Mana Momenilandi
- Specialized Immunology Laboratory of Dr. Shahrooei, Sina Medical Complex, Ahvaz, Iran
| | - Laura Pöyhönen
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY
| | - Marilda M Siqueira
- National Reference Laboratory for Respiratory Viruses, Institute Oswaldo Cruz, Fiocruz, Ministry of Health, Rio de Janeiro, Brazil
| | - Sheila M Barbosa de Lima
- Laboratory of Virological Techniques, Bio-Manguinhos, Fiocruz, Ministry of Health, Rio de Janeiro, Brazil
| | - Denise C de Souza Matos
- Laboratory of Immunological Techniques, Bio-Manguinhos, Fiocruz, Ministry of Health, Rio de Janeiro, Brazil
| | - Akira Homma
- Bio-Manguinhos, Fiocruz, Ministry of Health, Rio de Janeiro, Brazil
| | | | | | | | | | - Rik Gijsbers
- Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.,Leuven Viral Vector Core, Leuven, Belgium
| | - Shen-Ying Zhang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY.,Paris Descartes University, Imagine Institute, Paris, France.,Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Paris, France
| | - Stephen J Seligman
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY.,Department of Microbiology and Immunology, New York Medical College, Valhalla, NY
| | - Laurent Abel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY.,Paris Descartes University, Imagine Institute, Paris, France.,Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Paris, France
| | - Paul Hertzog
- Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Nico Marr
- Division of Translational Medicine, Sidra Medicine, Doha, Qatar.,College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | | | - Isabelle Meyts
- Laboratory of Inborn Errors of Immunity, Department of Immunology, Microbiology and Transplantation, KU Leuven, Leuven, Belgium.,Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium.,Precision Immunology Institute and Mindich Child Health and Development Institute at the Icahn School of Medicine at Mount Sinai, New York, NY
| | - Qian Zhang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY
| | - Margaret R MacDonald
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY
| | - Charles M Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY .,Pediatric Immunology-Hematology Unit, Assistance Publique-Hôpitaux de Paris, Necker Hospital for Sick Children, Paris, France.,Paris Descartes University, Imagine Institute, Paris, France.,Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Paris, France.,Howard Hughes Medical Institute, New York, NY
| | - Emmanuelle Jouanguy
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY.,Paris Descartes University, Imagine Institute, Paris, France.,Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Paris, France
| | - Xavier Bossuyt
- Department of Microbiology, Immunology and Transplantation, Clinical and Diagnostic Immunology, KU Leuven, Leuven, Belgium.,Department of Laboratory Medicine, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
33
|
The Human Cytomegalovirus Chemokine vCXCL-1 Modulates Normal Dissemination Kinetics of Murine Cytomegalovirus In Vivo. mBio 2019; 10:mBio.01289-19. [PMID: 31239384 PMCID: PMC6593410 DOI: 10.1128/mbio.01289-19] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
An adequate in vivo analysis of HCMV’s viral chemokine vCXCL-1 has been lacking. Here we generate recombinant MCMVs expressing vCXCL-1 to study vCXCL-1 function in vivo using MCMV as a surrogate. We demonstrate that vCXCL-1 increases MCMV dissemination kinetics for both primary and secondary dissemination. Additionally, we provide evidence, that the murine neutrophil is largely a bystander in the mouse’s response to vCXCL-1. We confirm the hypothesis that vCXCL-1 is a HCMV virulence factor. Infection of severely immunocompromised mice with MCMVs expressing vCXCL-1 was lethal in more than 50% of infected animals, while all animals infected with parental virus survived during a 12-day period. This work provides needed insights into vCXCL-1 function in vivo. Human cytomegalovirus (HCMV) is a betaherpesvirus that is a significant pathogen within newborn and immunocompromised populations. Morbidity associated with HCMV infection is the consequence of viral dissemination. HCMV has evolved to manipulate the host immune system to enhance viral dissemination and ensure long-term survival within the host. The immunomodulatory protein vCXCL-1, a viral chemokine functioning primarily through the CXCR2 chemokine receptor, is hypothesized to attract CXCR2+ neutrophils to infection sites, aiding viral dissemination. Neutrophils harbor HCMV in vivo; however, the interaction between vCXCL-1 and the neutrophil has not been evaluated in vivo. Using the mouse model and mouse cytomegalovirus (MCMV) infection, we show that murine neutrophils harbor and transfer infectious MCMV and that virus replication initiates within this cell type. Utilizing recombinant MCMVs expressing vCXCL-1 from the HCMV strain (Toledo), we demonstrated that vCXCL-1 significantly enhances MCMV dissemination kinetics. Through cellular depletion experiments, we observe that neutrophils impact dissemination but that overall dissemination is largely neutrophil independent. This work adds neutrophils to the list of innate cells (i.e., dendritic and macrophages/monocytes) that contribute to MCMV dissemination but refutes the hypothesis that neutrophils are the primary cell responding to vCXCL-1.
Collapse
|
34
|
A Newly Designed EGFP-2A Peptide Monocistronic Baculoviral Vector for Concatenating the Expression of Recombinant Proteins in Insect Cells. Processes (Basel) 2019. [DOI: 10.3390/pr7050291] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Recombinant proteins produced by the baculovirus expression vector system (BVES) have been widely applied in the agricultural and medical fields. However, the procedure for protein expression is inefficient and needs to be improved. Herein, we propose a simple construct that incorporates a selectable marker (enhanced green fluorescent protein, EGFP) and a picorna viral-derived “self-cleaving” 2A-like peptide to separate the EGFP and target proteins in a monocistronic baculovirus vector to facilitate isolation of the recombinant baculovirus in the BVES. In this study, porcine adiponectin (ADN), a secreted, multimeric protein with insulin-sensitizing properties, was used to demonstrate its utility in our EGFP-2A-based expression system. EGFP and ADN were simultaneously expressed by a recombinant alphabaculovirus. Co-expression of EGFP facilitates the manipulation of the following processes, such as determining expression kinetics and harvesting ADN. The results showed that the 2A “self-cleaving” process does not interfere with EGFP activity or with signal peptide removal and the secretion of recombinant ADN. Posttranslational modifications, including glycosylation, of the recombinant ADN occurred in insect cells, and the formation of various multimers was further verified. Most importantly, the insect-produced ADN showed a similar bioactivity to that of mammalian cells. This concept provides a practical and economic approach that utilizes a new combination of alphabaculovirus/insect cell expression systems for future applications.
Collapse
|
35
|
Liu F, Liu J, Yuan Z, Qing Y, Li H, Xu K, Zhu W, Zhao H, Jia B, Pan W, Guo J, Zhang X, Cheng W, Wang W, Zhao HY, Wei HJ. Generation of GTKO Diannan Miniature Pig Expressing Human Complementary Regulator Proteins hCD55 and hCD59 via T2A Peptide-Based Bicistronic Vectors and SCNT. Mol Biotechnol 2019; 60:550-562. [PMID: 29916131 DOI: 10.1007/s12033-018-0091-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Pig-to-human organ transplantation has drawn attention in recent years due to the potential use of pigs as an alternative source of human donor organs. While GGTA1 knockout (GTKO) can protect xenografts from hyperacute rejection, complement-dependent cytotoxicity might still contribute to this type of rejection. To prolong the xenograft survival, we utilized a T2A-mediated pCMV-hCD55-T2A-hCD59-Neo vector and transfected the plasmid into GTKO Diannan miniature pig fetal fibroblasts. After G418 selection combined with single-cell cloning culture, four colonies were obtained, and three of these were successfully transfected with the hCD55 and hCD59. One of the three colonies was selected as donor cells for somatic cell nuclear transfer (SCNT). Then, the reconstructed embryos were transferred into eight recipient gilts, resulting in four pregnancies. Three of the pregnant gilts delivered, yielding six piglets. Only one piglet carried hCD55 and hCD59 genetic modification. The expression levels of the GGTA1, hCD55, and hCD59 in the tissues and fibroblasts of the piglet were determined by q-PCR, fluorescence microscopy, immunohistochemical staining, and western blotting analyses. The results showed the absence of GGTA1 and the coexpression of the hCD55 and hCD59. However, the mRNA expression levels of hCD55 and hCD59 in the GTKO/hCD55/hCD59 pig fibroblasts were lower than that in human 293T cells, which may be caused by low copy number and/or CMV promoter methylation. Furthermore, we performed human complement-mediated cytolysis assays using human serum solutions from 0 to 60%. The result showed that the fibroblasts of this triple-gene modified piglet had greater survival rates than that of wild-type and GTKO controls. Taken together, these results indicate that T2A-mediated polycistronic vector system combined with SCNT can effectively generate multiplex genetically modified pigs, additional hCD55 and hCD59 expression on top of a GTKO genetic background markedly enhance the protective effect towards human serum-mediated cytolysis than those of GTKO alone. Thus, we suggest that GTKO/hCD55/hCD59 triple-gene-modified Diannan miniature pig will be a more eligible donor for xenotransplantation.
Collapse
Affiliation(s)
- Fengjuan Liu
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Jinji Liu
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Zaimei Yuan
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Yubo Qing
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Honghui Li
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Kaixiang Xu
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China
| | - Wanyun Zhu
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China
| | - Heng Zhao
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Baoyu Jia
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Weirong Pan
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Jianxiong Guo
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China
| | - Xuezeng Zhang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201, China
| | - Wenmin Cheng
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, 650201, China
| | - Wei Wang
- Hunan Xeno Life Science Co., Ltd, Changsha, 410600, China.
- Institute for Cell Transplantation and Gene Therapy, The Third Xiangya Hospital Central-South University, Changsha, 410013, China.
| | - Hong-Ye Zhao
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China.
| | - Hong-Jiang Wei
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan Agricultural University, Kunming, 650201, China.
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, 650201, China.
| |
Collapse
|
36
|
Structure-Guided Exploration of SDS22 Interactions with Protein Phosphatase PP1 and the Splicing Factor BCLAF1. Structure 2019; 27:507-518.e5. [PMID: 30661852 DOI: 10.1016/j.str.2018.12.002] [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: 01/23/2017] [Revised: 10/09/2018] [Accepted: 12/03/2018] [Indexed: 01/29/2023]
Abstract
SDS22 is an ancient regulator of protein phosphatase-1 (PP1). Our crystal structure of SDS22 shows that its twelve leucine-rich repeats adopt a banana-shaped fold that is shielded from solvent by capping domains at its extremities. Subsequent modeling and biochemical studies revealed that the concave side of SDS22 likely interacts with PP1 helices α5 and α6, which are distal from the binding sites of many previously described PP1 interactors. Accordingly, we found that SDS22 acts as a "third" subunit of multiple PP1 holoenzymes. The crystal structure of SDS22 also revealed a large basic surface patch that enables binding of a phosphorylated form of splicing factor BCLAF1. Taken together, our data provide insights into the formation of PP1:SDS22 and the recruitment of additional interaction proteins, such as BCLAF1.
Collapse
|
37
|
Haran KP, Hajduczki A, Pampusch MS, Mwakalundwa G, Vargas-Inchaustegui DA, Rakasz EG, Connick E, Berger EA, Skinner PJ. Simian Immunodeficiency Virus (SIV)-Specific Chimeric Antigen Receptor-T Cells Engineered to Target B Cell Follicles and Suppress SIV Replication. Front Immunol 2018; 9:492. [PMID: 29616024 PMCID: PMC5869724 DOI: 10.3389/fimmu.2018.00492] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 02/26/2018] [Indexed: 11/13/2022] Open
Abstract
There is a need to develop improved methods to treat and potentially cure HIV infection. During chronic HIV infection, replication is concentrated within T follicular helper cells (Tfh) located within B cell follicles, where low levels of virus-specific CTL permit ongoing viral replication. We previously showed that elevated levels of simian immunodeficiency virus (SIV)-specific CTL in B cell follicles are linked to both decreased levels of viral replication in follicles and decreased plasma viral loads. These findings provide the rationale to develop a strategy for targeting follicular viral-producing (Tfh) cells using antiviral chimeric antigen receptor (CAR) T cells co-expressing the follicular homing chemokine receptor CXCR5. We hypothesize that antiviral CAR/CXCR5-expressing T cells, when infused into an SIV-infected animal or an HIV-infected individual, will home to B cell follicles, suppress viral replication, and lead to long-term durable remission of SIV and HIV. To begin to test this hypothesis, we engineered gammaretroviral transduction vectors for co-expression of a bispecific anti-SIV CAR and rhesus macaque CXCR5. Viral suppression by CAR/CXCR5-transduced T cells was measured in vitro, and CXCR5-mediated migration was evaluated using both an in vitro transwell migration assay, as well as a novel ex vivo tissue migration assay. The functionality of the CAR/CXCR5 T cells was demonstrated through their potent suppression of SIVmac239 and SIVE660 replication in in vitro and migration to the ligand CXCL13 in vitro, and concentration in B cell follicles in tissues ex vivo. These novel antiviral immunotherapy products have the potential to provide long-term durable remission (functional cure) of HIV and SIV infections.
Collapse
Affiliation(s)
- Kumudhini Preethi Haran
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Agnes Hajduczki
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Mary S Pampusch
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Gwantwa Mwakalundwa
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Diego A Vargas-Inchaustegui
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Eva G Rakasz
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Elizabeth Connick
- Division of Infectious Diseases, University of Arizona, Tucson, AZ, United States
| | - Edward A Berger
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Pamela J Skinner
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| |
Collapse
|
38
|
Askou AL, Benckendorff JNE, Holmgaard A, Storm T, Aagaard L, Bek T, Mikkelsen JG, Corydon TJ. Suppression of Choroidal Neovascularization in Mice by Subretinal Delivery of Multigenic Lentiviral Vectors Encoding Anti-Angiogenic MicroRNAs. Hum Gene Ther Methods 2018; 28:222-233. [PMID: 28817343 DOI: 10.1089/hgtb.2017.079] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Lentivirus-based vectors have been used for the development of potent gene therapies. Here, application of a multigenic lentiviral vector (LV) producing multiple anti-angiogenic microRNAs following subretinal delivery in a laser-induced choroidal neovascularization (CNV) mouse model is presented. This versatile LV, carrying back-to-back RNApolII-driven expression cassettes, enables combined expression of microRNAs targeting vascular endothelial growth factor A (Vegfa) mRNA and fluorescent reporters. In addition, by including a vitelliform macular dystrophy 2 (VMD2) promoter, expression of microRNAs is restricted to the retinal pigment epithelial (RPE) cells. Six days post injection (PI), robust and widespread fluorescent signals of eGFP are already observed in the retina by funduscopy. The eGFP expression peaks at day 21 PI and persists with stable expression for at least 9 months. In parallel, prominent AsRED co-expression, encoded from the VMD2-driven microRNA expression cassette, is evident in retinal sections and flat-mounts, revealing RPE-specific expression of microRNAs. Furthermore, LV-delivered microRNAs targeting the Vegfa gene in RPE cells reduced the size of laser-induced CNV in mice 28 days PI, as a consequence of diminished VEGF levels, suggesting that LVs delivered locally are powerful tools in the development of gene therapy-based strategies for treatment of age-related macular degeneration.
Collapse
Affiliation(s)
| | | | | | - Tina Storm
- 1 Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Lars Aagaard
- 1 Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Toke Bek
- 2 Department of Ophthalmology, Aarhus University Hospital , Aarhus, Denmark
| | | | - Thomas Juhl Corydon
- 1 Department of Biomedicine, Aarhus University, Aarhus, Denmark .,2 Department of Ophthalmology, Aarhus University Hospital , Aarhus, Denmark
| |
Collapse
|
39
|
Syntaxins on granules promote docking of granules via interactions with munc18. Sci Rep 2018; 8:193. [PMID: 29317735 PMCID: PMC5760731 DOI: 10.1038/s41598-017-18597-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 12/13/2017] [Indexed: 01/02/2023] Open
Abstract
SNAREs and SNARE-binding accessory proteins are believed to be central molecular components of neurotransmitter release, although the precise sequence of molecular events corresponding to distinct physiological states is unclear. The mechanism of docking of vesicles to the plasma membrane remains elusive, as the anchoring protein residing on vesicles is unknown. Here I show that targeting small amounts of syntaxin to granules by transmembrane domain alteration leads to a substantial enhancement of syntaxin clustering beneath granules, as well as of morphological granule docking. The effect was abolished without munc18 and strongly reduced by removal of the N-terminal peptide in the syntaxin mutant. Thus, in contrast to the current paradigm, I demonstrate that syntaxin acts from the vesicular membrane, strongly facilitating docking of vesicles, likely via interaction of its N-peptide with munc18. Docking was assayed by quantifying the syntaxin clusters beneath granules, using two-color Total Internal Reflectance Fluorescence microscopy in live PC-12 cells and confirmed by electron microscopy. Hereby, I propose a new model of vesicle docking, wherein munc18 bridges the few syntaxin molecules residing on granules to the syntaxin cluster on the plasma membrane, suggesting that the number of syntaxins on vesicles determines docking and conceivably fusion probability.
Collapse
|
40
|
Lee M, Zhou Y, Huang Y. An Engineered Split-TET2 Enzyme for Chemical-inducible DNA Hydroxymethylation and Epigenetic Remodeling. J Vis Exp 2017. [PMID: 29286410 DOI: 10.3791/56858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
DNA methylation is a stable and heritable epigenetic modification in the mammalian genome and is involved in regulating gene expression to control cellular functions. The reversal of DNA methylation, or DNA demethylation, is mediated by the ten-eleven translocation (TET) protein family of dioxygenases. Although it has been widely reported that aberrant DNA methylation and demethylation are associated with developmental defects and cancer, how these epigenetic changes directly contribute to the subsequent alteration in gene expression or disease progression remains unclear, largely owing to the lack of reliable tools to accurately add or remove DNA modifications in the genome at defined temporal and spatial resolution. To overcome this hurdle, we designed a split-TET2 enzyme to enable temporal control of 5-methylcytosine (5mC) oxidation and subsequent remodeling of epigenetic states in mammalian cells by simply adding chemicals. Here, we describe methods for introducing a chemical-inducible epigenome remodeling tool (CiDER), based on an engineered split-TET2 enzyme, into mammalian cells and quantifying the chemical inducible production of 5-hydroxymethylcytosine (5hmC) with immunostaining, flow cytometry or a dot-blot assay. This chemical-inducible epigenome remodeling tool will find broad use in interrogating cellular systems without altering the genetic code, as well as in probing the epigenotype-phenotype relations in various biological systems.
Collapse
Affiliation(s)
- Minjung Lee
- Centre for Epigenetics and Disease Prevention, Department of Molecular and Cellular Medicine, Institute of Biosciences and Technology, College of Medicine, Texas A&M University
| | - Yubin Zhou
- Centre for Translational Cancer Research, Department of Medical Physiology, Institute of Biosciences and Technology, College of Medicine, Texas A&M University;
| | - Yun Huang
- Centre for Epigenetics and Disease Prevention, Department of Molecular and Cellular Medicine, Institute of Biosciences and Technology, College of Medicine, Texas A&M University;
| |
Collapse
|
41
|
Figueras A, Alsina-Sanchís E, Lahiguera Á, Abreu M, Muinelo-Romay L, Moreno-Bueno G, Casanovas O, Graupera M, Matias-Guiu X, Vidal A, Villanueva A, Viñals F. A Role for CXCR4 in Peritoneal and Hematogenous Ovarian Cancer Dissemination. Mol Cancer Ther 2017; 17:532-543. [PMID: 29146630 DOI: 10.1158/1535-7163.mct-17-0643] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 09/08/2017] [Accepted: 11/03/2017] [Indexed: 11/16/2022]
Abstract
Epithelial ovarian cancer is characterized by a low recovery rate because the disease is typically diagnosed at an advanced stage, by which time most patients (80%) already exhibit disseminated neoplasia. The cytokine receptor CXCR4 has been implicated in the development of metastasis in various tumor types. Using a patient-derived tissue macroarray and mRNA expression analysis, we observed high CXCR4 levels in high-grade serous epithelial ovarian carcinomas, the most metastatic tumor, compared with those in endometrioid carcinomas. CXCR4 inhibition by treatment with the CXCR4 antagonist AMD3100 or by expression of shRNA anti-CXCR4 similarly inhibited angiogenesis in several models of ovarian carcinomas orthotopically grown in nude mice, but the effect on tumor growth was correlated with the levels of CXCR4 expression. Moreover, CXCR4 inhibition completely blocked dissemination and metastasis. This effect was associated with reduced levels of active Src, active ERKs, the inhibition of EMT transition, and block of hematogenous ovarian cancer dissemination decreasing circulating human tumoral cells (CTC). In tumors, CXCR4-expressing cells also had more mesenchymal characteristics. In conclusion, our results indicate that CXCR4 expression confers a proinvasive phenotype to ovarian carcinoma cells. Thus, anti-CXCR4 therapy is a possible agent for a complementary treatment of advanced disseminated epithelial high-grade serous ovarian cancer patients. Mol Cancer Ther; 17(2); 532-43. ©2017 AACR.
Collapse
Affiliation(s)
- Agnès Figueras
- Program Against Cancer Therapeutic Resistance (ProCURE), Institut Català d'Oncologia, Hospital Duran i Reynals, Barcelona, Spain.,Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Elisenda Alsina-Sanchís
- Program Against Cancer Therapeutic Resistance (ProCURE), Institut Català d'Oncologia, Hospital Duran i Reynals, Barcelona, Spain.,Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Álvaro Lahiguera
- Program Against Cancer Therapeutic Resistance (ProCURE), Institut Català d'Oncologia, Hospital Duran i Reynals, Barcelona, Spain.,Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Manuel Abreu
- Liquid Biopsy Analysis Unit, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Laura Muinelo-Romay
- Liquid Biopsy Analysis Unit, Oncomet, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Gema Moreno-Bueno
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Departamento de Bioquímica, Universidad Autónoma de Madrid (UAM), Instituto de Investigaciones Biomédicas "Alberto Sols" (CSIC-UAM), IdiPaz, Madrid, Spain.,MD Anderson International Foundation, Madrid, Spain
| | - Oriol Casanovas
- Program Against Cancer Therapeutic Resistance (ProCURE), Institut Català d'Oncologia, Hospital Duran i Reynals, Barcelona, Spain.,Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Mariona Graupera
- Program Against Cancer Therapeutic Resistance (ProCURE), Institut Català d'Oncologia, Hospital Duran i Reynals, Barcelona, Spain.,Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain.,Laboratori d'Oncologia Molecular, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain
| | - Xavier Matias-Guiu
- Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.,Servei d'Anatomia Patològica, Hospital Universitari de Bellvitge, Barcelona, Spain
| | - August Vidal
- Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain.,Servei d'Anatomia Patològica, Hospital Universitari de Bellvitge, Barcelona, Spain.,Xenopat, Carrer de la Feixa Llarga S/N, L'Hospitalet de Llobregat, Barcelona, Spain.,Departament de Patologia i Terapèutica Experimental, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Alberto Villanueva
- Program Against Cancer Therapeutic Resistance (ProCURE), Institut Català d'Oncologia, Hospital Duran i Reynals, Barcelona, Spain.,Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain.,Xenopat, Carrer de la Feixa Llarga S/N, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Francesc Viñals
- Program Against Cancer Therapeutic Resistance (ProCURE), Institut Català d'Oncologia, Hospital Duran i Reynals, Barcelona, Spain. .,Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain.,Departament de Ciències Fisiològiques, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
42
|
Functional Evaluation of an IKBKG Variant Suspected to Cause Immunodeficiency Without Ectodermal Dysplasia. J Clin Immunol 2017; 37:801-810. [PMID: 28993958 DOI: 10.1007/s10875-017-0448-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 09/20/2017] [Indexed: 10/18/2022]
Abstract
Hypomorphic IKBKG mutations in males are typically associated with anhidrotic ectodermal dysplasia with immunodeficiency (EDA-ID). Some mutations cause immunodeficiency without EDA (NEMO-ID). The immunological profile associated with these NEMO-ID variants is not fully documented. We present a 2-year-old patient with suspected immunodeficiency in which a hemizygous p.Glu57Lys IKBKG variant was identified. At the age of 1 year, he had an episode of otitis media that evolved into a bilateral mastoiditis (Pseudomonas spp). Hypogammaglobulinemia, specific (polysaccharide) antibody deficiency, and low switched memory B cell subsets were noticed. The mother was heterozygous for the variant but had no signs of incontinentia pigmenti. Patient peripheral blood mononuclear cells produced low amounts of IL-6 after stimulation with IL-1β, Pam3CSK4, and FSL-1. In patient fibroblasts, IκB-α was degraded normally upon stimulation with IL-1β or TNF-α. Transduction of wild-type and variant NEMO in NEMO-/- deficient SV40 fibroblasts revealed a slight but significant reduction of IL-6 production upon stimulation with IL-1β and TNF-α. In conclusion, we demonstrated that p.Glu57Lys leads to specific immunological defects in vitro. No other pathogenic PID variants were identified through whole exome sequencing. As rare polymorphisms have been described in IKBKG and polygenic inheritance remains an option in the presented case, this study emphasizes the need for thorough functional and genetic evaluation when encountering and interpreting suspected disease-causing NEMO-ID variants.
Collapse
|
43
|
Genome editing in Shiraia bambusicola using CRISPR-Cas9 system. J Biotechnol 2017; 259:228-234. [DOI: 10.1016/j.jbiotec.2017.06.1204] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 05/29/2017] [Accepted: 06/29/2017] [Indexed: 12/20/2022]
|
44
|
Deng H, Gao R, Liao X, Cai Y. Characterization of a major facilitator superfamily transporter in Shiraia bambusicola. Res Microbiol 2017; 168:664-672. [DOI: 10.1016/j.resmic.2017.05.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 05/03/2017] [Accepted: 05/15/2017] [Indexed: 10/19/2022]
|
45
|
Askou AL, Benckendorff JNE, Holmgaard A, Storm T, Aagaard L, Bek T, Mikkelsen JG, Corydon TJ. Suppression of choroidal neovascularization in mice by subretinal delivery of multigenic lentiviral vectors encoding anti-angiogenic microRNAs. Hum Gene Ther Methods 2017. [DOI: 10.1089/hum.2017.079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Affiliation(s)
| | | | | | - Tina Storm
- Aarhus University, Department of Biomedicine, Aarhus C, Denmark
| | - Lars Aagaard
- Aarhus University, Department of Biomedicine, Aarhus C, Denmark
| | - Toke Bek
- Aarhus University Hospital, Department of Ophthalmology, Aarhus C, Denmark
| | | | - Thomas Juhl Corydon
- Aarhus University, Department of Biomedicine, Wilhelm Meyers Alle, Aarhus C, Denmark, 8000
| |
Collapse
|
46
|
Ni X, Ou C, Guo J, Liu B, Zhang J, Wu Z, Li H, Chen M. Lentiviral vector-mediated co-overexpression of VEGF and Bcl-2 improves mesenchymal stem cell survival and enhances paracrine effects in vitro. Int J Mol Med 2017. [PMID: 28627637 PMCID: PMC5505017 DOI: 10.3892/ijmm.2017.3019] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cell (MSC) transplantation has emerged as a promising therapy for ischemic heart disease; however, the low survival rate of transplanted cells limits their therapeutic efficacy. The aim of this study was to investigate whether the dual genetic modification of vascular endothelial growth factor (VEGF) and B-cell lymphoma-2 (Bcl-2) confers a higher expression level of the target genes, better survival and a stronger paracrine effect in MSCs in an adverse environment than the modification of the individual genes. For this purpse, a lentiviral vector was constructed by using a self-cleaving T2A peptide sequence to link and achieve the co-overexpression of VEGF and Bcl-2. Rat MSCs were transfected to obtain cell lines that exhibited a stable overexpression. An in vitro model of oxygen glucose deprivation (OGD) was applied to mimic the ischemic microenvironment, and cell apoptosis, autophagy and the paracrine effects were then determined. Compared with the MSCs in which individual genes were modified and the control MSCs, the MSCs which were subjected to dual genetic modification had a higher expression level of the target genes, a more rapid proliferation, reduced apoptosis, decreased autophagy and an enhanced paracrine effect. Furthermore, the suppression of autophagy was found to contribute to the inhibition of apoptosis in this in vitro OGD model. On the whole, these data indicate that the co-overexpression of VEGF and Bcl-2 protects MSCs in an ischemic environment by inhibiting apoptosis, suppressing autophagy and enhancing the paracrine effects.
Collapse
Affiliation(s)
- Xiaobin Ni
- Department of Cardiology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Caiwen Ou
- Department of Cardiology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Jingbin Guo
- Department of Cardiology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Bei Liu
- Department of Cardiology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Jianwu Zhang
- Department of Cardiology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Zhiye Wu
- Department of Cardiology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Hekai Li
- Department of Cardiology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Minsheng Chen
- Department of Cardiology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| |
Collapse
|
47
|
El Ashkar S, Van Looveren D, Schenk F, Vranckx LS, Demeulemeester J, De Rijck J, Debyser Z, Modlich U, Gijsbers R. Engineering Next-Generation BET-Independent MLV Vectors for Safer Gene Therapy. MOLECULAR THERAPY-NUCLEIC ACIDS 2017. [PMID: 28624199 PMCID: PMC5415309 DOI: 10.1016/j.omtn.2017.04.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Retroviral vectors have shown their curative potential in clinical trials correcting monogenetic disorders. However, therapeutic benefits were compromised due to vector-induced dysregulation of cellular genes and leukemia development in a subset of patients. Bromodomain and extraterminal domain (BET) proteins act as cellular cofactors that tether the murine leukemia virus (MLV) pre-integration complex to host chromatin via interaction with the MLV integrase (IN) and thereby define the typical gammaretroviral integration distribution. We engineered next-generation BET-independent (Bin) MLV vectors to retarget their integration to regions where they are less likely to dysregulate nearby genes. We mutated MLV IN to uncouple BET protein interaction and fused it with chromatin-binding peptides. The addition of the CBX1 chromodomain to MLV INW390A efficiently targeted integration away from gene regulatory elements. The retargeted vector produced at high titers and efficiently transduced CD34+ hematopoietic stem cells, while fewer colonies were detected in a serial colony-forming assay, a surrogate test for genotoxicity. Our findings underscore the potential of the engineered vectors to reduce the risk of insertional mutagenesis without compromising transduction efficiency. Ultimately, combined with other safety features in vector design, next-generation BinMLV vectors can improve the safety of gammaretroviral vectors for gene therapy.
Collapse
Affiliation(s)
- Sara El Ashkar
- Laboratory for Molecular Virology and Drug Discovery, Department of Pharmaceutical and Pharmacological Sciences, 3000 Leuven, KU Leuven, Belgium
| | - Dominique Van Looveren
- Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Franziska Schenk
- RG Gene Modification in Stem Cells, LOEWE Center for Cell and Gene Therapy Frankfurt, Paul-Ehrlich-Institute, 63225 Langen, Germany
| | - Lenard S Vranckx
- Laboratory for Molecular Virology and Drug Discovery, Department of Pharmaceutical and Pharmacological Sciences, 3000 Leuven, KU Leuven, Belgium
| | - Jonas Demeulemeester
- Laboratory for Molecular Virology and Drug Discovery, Department of Pharmaceutical and Pharmacological Sciences, 3000 Leuven, KU Leuven, Belgium
| | - Jan De Rijck
- Laboratory for Molecular Virology and Drug Discovery, Department of Pharmaceutical and Pharmacological Sciences, 3000 Leuven, KU Leuven, Belgium
| | - Zeger Debyser
- Laboratory for Molecular Virology and Drug Discovery, Department of Pharmaceutical and Pharmacological Sciences, 3000 Leuven, KU Leuven, Belgium
| | - Ute Modlich
- RG Gene Modification in Stem Cells, LOEWE Center for Cell and Gene Therapy Frankfurt, Paul-Ehrlich-Institute, 63225 Langen, Germany
| | - Rik Gijsbers
- Laboratory for Viral Vector Technology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, 3000 Leuven, Belgium; Leuven Viral Vector Core, KU Leuven, 3000 Leuven, Belgium.
| |
Collapse
|
48
|
Lee M, Li J, Liang Y, Ma G, Zhang J, He L, Liu Y, Li Q, Li M, Sun D, Zhou Y, Huang Y. Engineered Split-TET2 Enzyme for Inducible Epigenetic Remodeling. J Am Chem Soc 2017; 139:4659-4662. [PMID: 28294608 PMCID: PMC5385525 DOI: 10.1021/jacs.7b01459] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The Ten-eleven translocation (TET) family of 5-methylcytosine (5mC) dioxygenases catalyze the conversion of 5mC into 5-hydroxymethylcytosine (5hmC) and further oxidized species to promote active DNA demethylation. Here we engineered a split-TET2 enzyme to enable temporal control of 5mC oxidation and subsequent remodeling of epigenetic states in mammalian cells. We further demonstrate the use of this chemically inducible system to dissect the correlation between DNA hydroxymethylation and chromatin accessibility in the mammalian genome. This chemical-inducible epigenome remodeling tool will find broad use in interrogating cellular systems without altering the genetic code, as well as in probing the epigenotype-phenotype relations in various biological systems.
Collapse
Affiliation(s)
- Minjung Lee
- Centre for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University , 2121 W Holcombe Boulevard, Houston, Texas 77030, United States
| | - Jia Li
- Centre for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University , 2121 W Holcombe Boulevard, Houston, Texas 77030, United States
| | - Yi Liang
- Centre for Translational Cancer Research, Institute of Biosciences and Technology, Department of Medical Physiology, College of Medicine, Texas A&M University , 2121 W Holcombe Boulevard, Houston, Texas 77030, United States
| | - Guolin Ma
- Centre for Translational Cancer Research, Institute of Biosciences and Technology, Department of Medical Physiology, College of Medicine, Texas A&M University , 2121 W Holcombe Boulevard, Houston, Texas 77030, United States
| | - Jixiang Zhang
- Centre for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University , 2121 W Holcombe Boulevard, Houston, Texas 77030, United States.,Department of Gastroenterology, Renmin Hospital of Wuhan University , Wuhan, Hubei 430060, China
| | - Lian He
- Centre for Translational Cancer Research, Institute of Biosciences and Technology, Department of Medical Physiology, College of Medicine, Texas A&M University , 2121 W Holcombe Boulevard, Houston, Texas 77030, United States
| | - Yuliang Liu
- Centre for Translational Cancer Research, Institute of Biosciences and Technology, Department of Medical Physiology, College of Medicine, Texas A&M University , 2121 W Holcombe Boulevard, Houston, Texas 77030, United States
| | - Qian Li
- Centre for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University , 2121 W Holcombe Boulevard, Houston, Texas 77030, United States
| | - Minyong Li
- School of Pharmacy, Shandong University , 44 Wenhua Road W., Jinan, Shandong 250012, China
| | - Deqiang Sun
- Centre for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University , 2121 W Holcombe Boulevard, Houston, Texas 77030, United States
| | - Yubin Zhou
- Centre for Translational Cancer Research, Institute of Biosciences and Technology, Department of Medical Physiology, College of Medicine, Texas A&M University , 2121 W Holcombe Boulevard, Houston, Texas 77030, United States
| | - Yun Huang
- Centre for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University , 2121 W Holcombe Boulevard, Houston, Texas 77030, United States
| |
Collapse
|
49
|
Affiliation(s)
- Alan S.L. Wong
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong
| | - Gigi C.G. Choi
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, University of Hong Kong, Pokfulam, Hong Kong
| | - Timothy K. Lu
- Synthetic Biology Group, Research Laboratory of Electronics, Department of Biological Engineering and Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139;
| |
Collapse
|
50
|
Towards a Safer, More Randomized Lentiviral Vector Integration Profile Exploring Artificial LEDGF Chimeras. PLoS One 2016; 11:e0164167. [PMID: 27788138 PMCID: PMC5082951 DOI: 10.1371/journal.pone.0164167] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 09/20/2016] [Indexed: 11/19/2022] Open
Abstract
The capacity to integrate transgenes into the host cell genome makes retroviral vectors an interesting tool for gene therapy. Although stable insertion resulted in successful correction of several monogenic disorders, it also accounts for insertional mutagenesis, a major setback in otherwise successful clinical gene therapy trials due to leukemia development in a subset of treated patients. Despite improvements in vector design, their use is still not risk-free. Lentiviral vector (LV) integration is directed into active transcription units by LEDGF/p75, a host-cell protein co-opted by the viral integrase. We engineered LEDGF/p75-based hybrid tethers in an effort to elicit a more random integration pattern to increase biosafety, and potentially reduce proto-oncogene activation. We therefore truncated LEDGF/p75 by deleting the N-terminal chromatin-reading PWWP-domain, and replaced this domain with alternative pan-chromatin binding peptides. Expression of these LEDGF-hybrids in LEDGF-depleted cells efficiently rescued LV transduction and resulted in LV integrations that distributed more randomly throughout the host-cell genome. In addition, when considering safe harbor criteria, LV integration sites for these LEDGF-hybrids distributed more safely compared to LEDGF/p75-mediated integration in wild-type cells. This approach should be broadly applicable to introduce therapeutic or suicide genes for cell therapy, such as patient-specific iPS cells.
Collapse
|