1
|
Swanson J, Tonne J, Sangsuwannukul T, Thompson J, Kendall B, Liseth O, Metko M, Vile R. APOBEC3B expression in 293T viral producer cells drives mutations in chimeric antigen receptors and reduces CAR T cell efficacy. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200873. [PMID: 39403625 PMCID: PMC11472098 DOI: 10.1016/j.omton.2024.200873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 08/15/2024] [Accepted: 09/05/2024] [Indexed: 11/07/2024]
Abstract
Chimeric antigen receptor (CAR) T cells are a clinically approved therapy for blood cancers. To produce clinical-grade CAR T cells, a retroviral or lentiviral vector is used to deliver the CAR and associated genes to patient T cells. Apolipoprotein B editing enzyme, catalytic polypeptide 3 (APOBEC3) enzymes are known to be upregulated after transfection and retroviral infection and to deaminate cytidine to uracil in nucleic acids, resulting in cytidine-to-thymine mutations in DNA. Here, we hypothesized that APOBEC3 enzymes, induced during the production of CAR T cells, impact the efficacy of the resulting CAR T cells. We demonstrated that APOBEC3 family member APOBEC3B was upregulated at the RNA and protein levels after transfection of HEK293T cells with plasmids to make lentivirus, and that APOBEC3 signature mutations were present in the CAR construct. APOBEC3B overexpression in HEK293T cells led to further mutations in the resulting CAR T cells, and significantly decreased CAR T cell killing. APOBEC3B knockout in HEK293T cells led to reduced mutations in the CAR construct and significantly increased in CAR T cell killing. These results suggest that generation of CAR-expressing viruses from producer cell lines deficient in genome-modifying proteins such as APOBEC3B could enhance the quality of CAR T cell production.
Collapse
Affiliation(s)
- Jack Swanson
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Jason Tonne
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Jill Thompson
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Benjamin Kendall
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Olivia Liseth
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Muriel Metko
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Richard Vile
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
2
|
Sun Y, Kronenberg NM, Sethi SK, Dash SN, Kovalik ME, Sempowski B, Strickland S, Raina R, Sperati CJ, Tian X, Ishibe S, Hall G, Gather MC. CRB2 Depletion Induces YAP Signaling and Disrupts Mechanosensing in Podocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.22.619513. [PMID: 39484460 PMCID: PMC11527017 DOI: 10.1101/2024.10.22.619513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Focal Segmental Glomerulosclerosis (FSGS) is a histologic lesion caused by a variety of injurious stimuli that lead to dysfunction/loss of glomerular visceral epithelial cells (i.e. podocytes). Pathogenic mutations in CRB2, encoding the type 1 transmembrane protein Crumb 2 Homolog Protein, have been shown to cause early-onset corticosteroid-resistant nephrotic syndrome (SRNS)/FSGS. Here, we identified a 2-generation East Asian kindred (DUK40595) with biopsy-proven SRNS/FSGS caused by a compound heterozygous mutation in CRB2 comprised of the previously described truncating mutation p.Gly1036_Alafs*43 and a rare 9-bp deletion mutation p.Leu1074_Asp1076del. Because compound heterozygous mutations involving the truncating p.Gly1036_Alafs*43 variant have been associated with reduced CRB2 expression in podocytes and autosomal recessive SRNS/FSGS, we sought to define the pathogenic effects of CRB2 deficiency in podocytes. We show that CRB2 knockdown induces YAP activity and target gene expression in podocytes. It upregulates YAP-mediated mechanosignaling and increases the density of focal adhesion and F-actin. Using Elastic Resonator Interference Stress Microscopy (ERISM), we demonstrate that CRB2 knockdown also enhances podocyte contractility in a substrate stiffness-dependent manner. The knockdown effect decreases with increasing substrate stiffness, indicating impaired mechanosensing in CRB2 knockdown cells at low substrate stiffness. While the mechanical activation of CRB2 knockdown cells is associated with increased YAP activity, the enhanced cell contractility is not significantly reduced by the selective YAP inhibitors K-975 and verteporfin, suggesting that multiple pathways may be involved in mechanosignaling downstream of CRB2. Taken together, these studies provide the first evidence that CRB2 deficiency may impair podocyte mechanotransduction via disruption of YAP signaling in podocytes.
Collapse
Affiliation(s)
- Yingyu Sun
- Humboldt Centre for Nano- and Biophotonics, Department of Chemistry, University of Cologne, Cologne, Germany
| | - Nils M. Kronenberg
- Humboldt Centre for Nano- and Biophotonics, Department of Chemistry, University of Cologne, Cologne, Germany
| | - Sidharth K. Sethi
- Pediatric Nephrology and Pediatric Kidney Transplantation, Medanta Kidney and Urology Institute, The Medicity Hospital, Gurgaon, Haryana, India
| | - Surjya N. Dash
- Division of Nephrology, Department of Medicine, Duke University, Durham, North Carolina, U.S.A
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, U.S.A
| | - Maria E. Kovalik
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, U.S.A
| | - Benjamin Sempowski
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, U.S.A
| | - Shelby Strickland
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, U.S.A
| | - Rupresh Raina
- Division of Nephrology, Department of Medicine, Yale University, New Haven, Connecticut, U.S.A
- Cleveland Clinic Akron General Medical Center, Akron Nephrology Associates, Akron, Ohio, USA
| | - C. John Sperati
- Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, U.S.A
| | - Xuefei Tian
- Cleveland Clinic Akron General Medical Center, Akron Nephrology Associates, Akron, Ohio, USA
| | - Shuta Ishibe
- Cleveland Clinic Akron General Medical Center, Akron Nephrology Associates, Akron, Ohio, USA
| | - Gentzon Hall
- Division of Nephrology, Department of Medicine, Duke University, Durham, North Carolina, U.S.A
- Duke Molecular Physiology Institute, Duke University, Durham, North Carolina, U.S.A
| | - Malte C. Gather
- Humboldt Centre for Nano- and Biophotonics, Department of Chemistry, University of Cologne, Cologne, Germany
- Centre of Biophotonics, SUPA, School of Physics and Astronomy, University of St Andrews, St Andrews, U.K
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Disease (CECAD), University of Cologne, Cologne, Germany
| |
Collapse
|
3
|
Eid LE, Deane-Alder K, Rujan RM, Mariam Z, Oqua AI, Manchanda Y, Belousoff MJ, Bernardino de la Serna J, Sloop KW, Rutter GA, Montoya A, Withers DJ, Millership SJ, Bouzakri K, Jones B, Reynolds CA, Sexton PM, Wootten D, Deganutti G, Tomas A. In vivo functional profiling and structural characterisation of the human Glp1r A316T variant. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.19.619191. [PMID: 39484598 PMCID: PMC11527029 DOI: 10.1101/2024.10.19.619191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Glucagon-like peptide-1 receptor (GLP-1R) agonists are a highly effective therapy class for type 2 diabetes (T2D) and obesity, yet there are variable patient responses. Variation in the human Glp1r gene leading to altered receptor structure, signal transduction, and function might be directly linked to therapeutic responses in patients. A naturally occurring, low-frequency, gain-of-function missense variant, rs10305492 G>A (A316T), protects against T2D and cardiovascular disease. Here we employ CRISPR/Cas9 technology to generate a humanised knock-in mouse model bearing the homozygous Glp1r A316T substitution. Human Glp1r A316T/A316T mice displayed lower fasting blood glucose levels and improved glucose tolerance, as well as increased plasma insulin levels and insulin secretion responses, even under metabolic stress. They also exhibited alterations in islet cytoarchitecture and β-cell identity indicative of compensatory mechanisms under a high-fat, high-sucrose (HFHS) diet challenge. Across all models investigated, the human Glp1r A316T variant exhibited characteristics of constitutive activation but blunted incretin-induced responses. Our results are further supported by cryo-EM analysis and molecular dynamics (MD) simulations of the GLP-1R A316T structure, demonstrating that the A316T Glp1r variant governs basal receptor activity and pharmacological responses to GLP-1R-targeting anti-diabetic therapies, highlighting the importance of the precise molecular characterisation of human Glp1r variants to predict individual therapy responses.
Collapse
|
4
|
Keyer V, Syzdykova L, Ingirbay B, Sedova E, Zauatbayeva G, Kulatay T, Shevtsov A, Shustov AV. Non-industrial production of therapeutic lentiviral vectors: How to provide vectors to academic CAR-T. Biotechnol Bioeng 2024; 121:3252-3268. [PMID: 38963234 DOI: 10.1002/bit.28794] [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: 03/26/2024] [Revised: 05/31/2024] [Accepted: 06/20/2024] [Indexed: 07/05/2024]
Abstract
Bringing effective cancer therapy in the form of chimeric antigen receptor technology to untapped markets faces numerous challenges, including a global shortage of therapeutic lentiviral or retroviral vectors on which all current clinical therapies using genetically modified T cells are based. Production of these lentiviral vectors in academic settings in principle opens the way to local production of therapeutic cells, which is the only economically viable approach to make this therapy available to patients in developing countries. The conditions for obtaining and concentrating lentiviral vectors have been optimized and described. The calcium phosphate precipitation method was found to be suitable for transfecting high cell-density cultures, a prerequisite for high titers. We describe protocols for gradually increasing production from 6-well plates to P100 plates, T-175 flasks, and 5-layer stacks while maintaining high titers, >108 transducing units. Concentration experiments using ultracentrifugation revealed the advantage of lower centrifugation speeds compared to competing protocols. The resulting batches of lentiviral vectors had a titer of 1010 infectious particles and were used to transduce primary human T lymphocytes generating chimeric antigen receptor T cells, the quality of which was checked and found potential applicability for treatment.
Collapse
Affiliation(s)
- Viktoriya Keyer
- Laboratory for Genetic Engineering, National Center for Biotechnology, Astana, Kazakhstan
| | - Laura Syzdykova
- Laboratory for Genetic Engineering, National Center for Biotechnology, Astana, Kazakhstan
| | - Bakytkali Ingirbay
- Laboratory for Genetic Engineering, National Center for Biotechnology, Astana, Kazakhstan
| | - Elena Sedova
- Laboratory for Genetic Engineering, National Center for Biotechnology, Astana, Kazakhstan
| | - Gulzat Zauatbayeva
- Laboratory for Genetic Engineering, National Center for Biotechnology, Astana, Kazakhstan
| | - Tolganay Kulatay
- Laboratory for Genetic Engineering, National Center for Biotechnology, Astana, Kazakhstan
| | - Alexandr Shevtsov
- Laboratory for Genetic Engineering, National Center for Biotechnology, Astana, Kazakhstan
| | - Alexandr V Shustov
- Laboratory for Genetic Engineering, National Center for Biotechnology, Astana, Kazakhstan
| |
Collapse
|
5
|
Mohamud Y, Lin JC, Hwang SW, Bahreyni A, Wang ZC, Luo H. Coxsackievirus B3 Activates Macrophages Independently of CAR-Mediated Viral Entry. Viruses 2024; 16:1456. [PMID: 39339932 PMCID: PMC11437450 DOI: 10.3390/v16091456] [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: 08/23/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Enteroviruses are a genus of small RNA viruses that are responsible for approximately one billion global infections annually. These infections range in severity from the common cold and flu-like symptoms to more severe diseases, such as viral myocarditis, pancreatitis, and neurological disorders, that continue to pose a global health challenge with limited therapeutic strategies currently available. In the current study, we sought to understand the interaction between coxsackievirus B3 (CVB3), which is a model enterovirus, and macrophage cells, as there is limited understanding of how this virus interacts with macrophage innate immune cells. Our study demonstrated that CVB3 can robustly activate macrophages without apparent viral replication in these cells. We also showed that myeloid cells lacked the viral entry receptor coxsackievirus and adenovirus receptor (CAR). However, the expression of exogenous CAR in RAW264.7 macrophages was unable to overcome the viral replication deficit. Interestingly, the CAR expression was associated with altered inflammatory responses during prolonged infection. Additionally, we identified the autophagy protein LC3 as a novel stimulus for macrophage activation. These findings provide new insights into the mechanisms of CVB3-induced macrophage activation and its implications for viral pathogenesis.
Collapse
Affiliation(s)
- Yasir Mohamud
- Centre for Heart Lung Innovation, University of British Columbia, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Jingfei Carly Lin
- Centre for Heart Lung Innovation, University of British Columbia, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Sinwoo Wendy Hwang
- Centre for Heart Lung Innovation, University of British Columbia, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Amirhossein Bahreyni
- Centre for Heart Lung Innovation, University of British Columbia, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Zhihan Claire Wang
- Centre for Heart Lung Innovation, University of British Columbia, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| | - Honglin Luo
- Centre for Heart Lung Innovation, University of British Columbia, St. Paul's Hospital, Vancouver, BC V6Z 1Y6, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6Z 1Y6, Canada
| |
Collapse
|
6
|
Shi H, Li J, Yan T, Zhou L, Zhu Y, Guo F, Yang S, Kong X, Zhou H. Krüppel-like factor 12 decreases progestin sensitivity in endometrial cancer by inhibiting the progesterone receptor signaling pathway. Transl Oncol 2024; 47:102041. [PMID: 38959708 PMCID: PMC11269804 DOI: 10.1016/j.tranon.2024.102041] [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: 11/02/2023] [Revised: 05/11/2024] [Accepted: 06/20/2024] [Indexed: 07/05/2024] Open
Abstract
OBJECTIVE This study aimed to clarify the mechanism by which Krüppel-like factor 12 (KLF12) affects progesterone sensitivity in endometrial cancer (EC) through the progesterone receptor PGR signaling pathway. METHODS The relationship of KLF12 with PGR in EC patients was examined by immunohistochemistry, and the expression of KLF12 and PGR in EC cell lines was detected by real-time PCR and western blotting. Cell proliferation assay, plate clone formation, cell apoptosis assay, and cell cycle analysis were conducted to determine the impact of KLF12 intervention on progesterone therapy. CUT&Tag analysis and the dual-luciferase reporter experiment were used to determine the underlying regulatory effect of KLF12 on the PGR DNA sequence. A subcutaneous xenograft nude mouse model was established to validate the in vivo effect of KLF12 on progesterone sensitivity via PGR expression modulation. RESULTS KLF12 demonstrated decreased progesterone sensitivity and a negative correlation with PGR expression in EC tissues. Progesterone sensitivity was increased by KLF12 deficiency through PGR overexpression, a result that could be significantly reversed by PGR downregulation. PGR was identified as a target gene of KLF12, which could directly bind to the PGR promotor region and inhibit its expression. CONCLUSION This study is the first to investigate the effect of KLF12 expression on EC cell resistance to progesterone. Our results offer important mechanistic insight into the direct regulation of the PGR promoter region, demonstrating that KLF12 expression strongly suppressed the PGR signaling pathway and, as a result, reduced progesterone sensitivity in EC patients.
Collapse
Affiliation(s)
- Haimeng Shi
- Department of Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University Medical School, Nanjing, PR China
| | - Jian Li
- Department of Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University Medical School, Nanjing, PR China
| | - Tong Yan
- Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, PR China
| | - Ling Zhou
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing 100044, PR China
| | - Yu Zhu
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing 210008, PR China
| | - Feifei Guo
- Department of Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University Medical School, Nanjing, PR China
| | - Sihui Yang
- Department of Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University Medical School, Nanjing, PR China
| | - Xiangyi Kong
- Department of Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University Medical School, Nanjing, PR China
| | - Huaijun Zhou
- Department of Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing University Medical School, Nanjing, PR China; Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, PR China.
| |
Collapse
|
7
|
He R, Dong T, Chu J, Wang Y, Wang X, Song Y, Sun Y. Phaser-Trim: A Phase Separation Based Genetically Encoded Reporter for H3K9 Trimethylation in Living Cells. Anal Chem 2024. [PMID: 39101551 DOI: 10.1021/acs.analchem.4c00992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/06/2024]
Abstract
Histone methylation is a key epigenetic modification that regulates the chromatin structure and gene expression for proper cellular and physiological processes. Aberrant histone methylation patterns are implicated in many diseases. Therefore, monitoring histone methylation dynamics in living cells and species is essential for elucidating its regulatory mechanisms and identifying potential therapeutic targets. However, current methods for detecting histone methylation are limited by their low sensitivity and specificity. To overcome this challenge, we have developed a genetically encoded biosensor named Phaser-Trim (Phase separation based genetically encoded reporter for H3K9 Trimethylation) to detect the dynamic changes of H3K9me3 in living cells and species through the generation and disappearance of phase-separated droplets. Phaser-Trim demonstrates advantages of clear phenotypic characteristics, convenient operation, quantitative accuracy, biocompatibility, high specificity, and superior imaging performance with high signal-to-background ratio (SBR) for in vivo animal imaging. Using Phaser-Trim, we have successfully detected the dynamics of the H3K9me3 level during the differentiation of neural stem cells in Drosophila. Furthermore, Phaser-Trim also holds promise for application in high-throughput screening systems to facilitate the discovery of novel anticancer drugs.
Collapse
Affiliation(s)
- Renxi He
- National Biomedical Imaging Center, College of Future Technology, State Key Laboratory of Membrane Biology, School of Life Sciences, and Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, 100871, China
| | - Tian Dong
- National Biomedical Imaging Center, College of Future Technology, State Key Laboratory of Membrane Biology, School of Life Sciences, and Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, 100871, China
| | - Jingyi Chu
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Yao Wang
- National Biomedical Imaging Center, College of Future Technology, State Key Laboratory of Membrane Biology, School of Life Sciences, and Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, 100871, China
| | - Xiaotian Wang
- National Biomedical Imaging Center, College of Future Technology, State Key Laboratory of Membrane Biology, School of Life Sciences, and Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, 100871, China
| | - Yan Song
- Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, School of Life Sciences, Peking University, Beijing, 100871, China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Yujie Sun
- National Biomedical Imaging Center, College of Future Technology, State Key Laboratory of Membrane Biology, School of Life Sciences, and Biomedical Pioneering Innovation Center (BIOPIC), Peking University, Beijing, 100871, China
| |
Collapse
|
8
|
Zhang R, He Z, Shi Y, Sun X, Chen X, Wang G, Zhang Y, Gao P, Wu Y, Lu S, Duan J, Sun S, Yang N, Fan W, Zhao K, Yang B, Xia Y, Zhang Y, Zhang Y, Yin H. Amplification editing enables efficient and precise duplication of DNA from short sequence to megabase and chromosomal scale. Cell 2024; 187:3936-3952.e19. [PMID: 38936359 DOI: 10.1016/j.cell.2024.05.056] [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: 12/25/2023] [Revised: 03/24/2024] [Accepted: 05/31/2024] [Indexed: 06/29/2024]
Abstract
Duplication is a foundation of molecular evolution and a driver of genomic and complex diseases. Here, we develop a genome editing tool named Amplification Editing (AE) that enables programmable DNA duplication with precision at chromosomal scale. AE can duplicate human genomes ranging from 20 bp to 100 Mb, a size comparable to human chromosomes. AE exhibits activity across various cell types, encompassing diploid, haploid, and primary cells. AE exhibited up to 73.0% efficiency for 1 Mb and 3.4% for 100 Mb duplications, respectively. Whole-genome sequencing and deep sequencing of the junctions of edited sequences confirm the precision of duplication. AE can create chromosomal microduplications within disease-relevant regions in embryonic stem cells, indicating its potential for generating cellular and animal models. AE is a precise and efficient tool for chromosomal engineering and DNA duplication, broadening the landscape of precision genome editing from an individual genetic locus to the chromosomal scale.
Collapse
Affiliation(s)
- Ruiwen Zhang
- Departments of Urology and Laboratory Medicine, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, State Key Laboratory of Virology, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China; Department of Rheumatology and Immunology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Zhou He
- Departments of Urology and Laboratory Medicine, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, State Key Laboratory of Virology, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Yajing Shi
- Departments of Urology and Laboratory Medicine, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, State Key Laboratory of Virology, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Xiangkun Sun
- Departments of Urology and Laboratory Medicine, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, State Key Laboratory of Virology, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Xinyu Chen
- Departments of Urology and Laboratory Medicine, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, State Key Laboratory of Virology, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Guoquan Wang
- Departments of Urology and Laboratory Medicine, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, State Key Laboratory of Virology, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Yizhou Zhang
- Departments of Urology and Laboratory Medicine, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, State Key Laboratory of Virology, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Pan Gao
- Departments of Urology and Laboratory Medicine, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, State Key Laboratory of Virology, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Ying Wu
- Departments of Urology and Laboratory Medicine, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, State Key Laboratory of Virology, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Shuhan Lu
- Departments of Urology and Laboratory Medicine, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, State Key Laboratory of Virology, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Junyi Duan
- Departments of Urology and Laboratory Medicine, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, State Key Laboratory of Virology, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Shangwu Sun
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Na Yang
- Center for Gene Diagnosis and Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Wei Fan
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Kaitao Zhao
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan 430071, China
| | - Bei Yang
- Shanghai Institute for Advanced Immunochemical Studies and School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yuchen Xia
- State Key Laboratory of Virology and Hubei Province Key Laboratory of Allergy and Immunology, Institute of Medical Virology, TaiKang Center for Life and Medical Sciences, TaiKang Medical School, Wuhan University, Wuhan 430071, China
| | - Yan Zhang
- Department of Clinical Laboratory, Institute of Translational Medicine, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ying Zhang
- Departments of Urology and Laboratory Medicine, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, State Key Laboratory of Virology, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China; Department of Rheumatology and Immunology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Hao Yin
- Departments of Urology and Laboratory Medicine, Frontier Science Center for Immunology and Metabolism, Medical Research Institute, Zhongnan Hospital of Wuhan University, State Key Laboratory of Virology, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
9
|
Yin E, Esbin MN. Optimized CRISPR-based knockout in BeWo cells. Placenta 2024:S0143-4004(24)00300-X. [PMID: 38997889 DOI: 10.1016/j.placenta.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/04/2024] [Accepted: 07/05/2024] [Indexed: 07/14/2024]
Abstract
CRISPR genome editing is a widely used tool to perturb genes of interest within cells and tissues and can be used as a research tool to study the connection between genotypes and cellular phenotypes. Highly efficient genome editing is limited in certain cell types due to low transfection efficiency or single-cell survivability. This is true for BeWo cells, an in vitro model of placental syncytiotrophoblast cell-cell fusion and hormone secretion. Here we describe an optimized and easy-to-use protocol for knockout in BeWo cells using CRISPR Cas9 ribonucleoprotein (RNP) complexes delivered via electroporation. Further, we describe parameters for successful guide RNA design and how to assess genetic knockouts in BeWo cells so that users can apply this technique to their own genes of interest. We provide a positive control for inducing highly efficient knockout of the cell-cell fusion protein Syncytin-2 (ERVFRD-1) and assessing editing efficiency at this locus. We anticipate that efficient RNP-mediated genetic knockouts in BeWo cells will facilitate the study of new genes involved in cell-cell fusion and hormone secretion in this important cellular model system. Furthermore, this strategy of optimized nucleofection and RNP delivery may be of use in other difficult-to-edit trophoblast cells or could be applied to efficiently deliver transgenes to BeWo cells.
Collapse
Affiliation(s)
- Eric Yin
- University of California, Berkeley, United States
| | | |
Collapse
|
10
|
Maghe C, Gavard J. Protocol for qualitative analysis of lysosome immunoprecipitation from patient-derived glioblastoma stem-like cells. STAR Protoc 2024; 5:103121. [PMID: 38850538 PMCID: PMC11215108 DOI: 10.1016/j.xpro.2024.103121] [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: 03/28/2024] [Revised: 05/03/2024] [Accepted: 05/17/2024] [Indexed: 06/10/2024] Open
Abstract
Lysosomes are critical for the sustenance of glioblastoma stem-like cells (GSCs) properties. We present a protocol to enrich and purify lysosomes from patient-derived GSCs in culture. We describe the steps required to stably express a tagged lysosomal protein in GSCs, mechanically lyse cells, magnetically immunopurify lysosomes, and qualitatively assess these organelles. We then detail the procedure for retrieving intact and purified lysosomes from GSCs. We also specify cell culture conditions, storage procedures, and sample preparation for immunoblotting. For complete details on the use and execution of this protocol, please refer to Maghe et al.1.
Collapse
Affiliation(s)
- Clément Maghe
- Team SOAP, CRCI(2)NA, Nantes Université, INSERM, CNRS, Université d'Angers, 44000 Nantes, France; Equipe Labellisée Ligue Nationale Contre le Cancer, 75013 Paris, France
| | - Julie Gavard
- Team SOAP, CRCI(2)NA, Nantes Université, INSERM, CNRS, Université d'Angers, 44000 Nantes, France; Equipe Labellisée Ligue Nationale Contre le Cancer, 75013 Paris, France; Institut de Cancérologie de l'Ouest, Saint Herblain, France.
| |
Collapse
|
11
|
Deleuze V, Soler E, Andrieu-Soler C. Protocol for efficient CRISPR-Cas9-mediated fluorescent tag knockin in hard-to-transfect erythroid cell lines. STAR Protoc 2024; 5:103016. [PMID: 38640065 PMCID: PMC11044133 DOI: 10.1016/j.xpro.2024.103016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/01/2024] [Accepted: 03/27/2024] [Indexed: 04/21/2024] Open
Abstract
Precise insertion of fluorescent tags by CRISPR-Cas9-mediated homologous recombination (HR) in mammalian genes is a powerful tool allowing to study gene function and protein gene products. Here, we present a protocol for efficient HR-mediated targeted insertion of fluorescent markers in the genome of hard-to-transfect erythroid cell lines MEL (mouse erythroleukemic) and MEDEP (mouse ES cell-derived erythroid progenitor line). We describe steps for plasmid construction, electroporation, amplification, and verification of genome editing. We then detail procedures for isolating positive clones and validating knockin clones. For complete details on the use and execution of this protocol, please refer to Deleuze et al.1.
Collapse
Affiliation(s)
- Virginie Deleuze
- IGMM University Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Université' de Paris, Paris, France
| | - Eric Soler
- IGMM University Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Université' de Paris, Paris, France.
| | - Charlotte Andrieu-Soler
- IGMM University Montpellier, CNRS, Montpellier, France; Laboratory of Excellence GR-Ex, Université' de Paris, Paris, France.
| |
Collapse
|
12
|
Sripada SA, Barbieri E, Shastry S, Wuestenhagen E, Aldinger A, Rammo O, Schulte MM, Daniele M, Menegatti S. Multiangle Light Scattering as a Lentivirus Purification Process Analytical Technology. Anal Chem 2024; 96:9593-9600. [PMID: 38804040 DOI: 10.1021/acs.analchem.4c01209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
The limited biomolecular and functional stability of lentiviral vectors (LVVs) for cell therapy poses the need for analytical tools that can monitor their titers and activity throughout the various steps of expression and purification. In this study, we describe a rapid (25 min) and reproducible (coefficient of variance ∼0.5-2%) method that leverages size exclusion chromatography coupled with multiangle light scattering detection (SEC-MALS) to determine size, purity, and particle count of LVVs purified from bioreactor harvests. The SEC-MALS data were corroborated by orthogonal methods, namely, dynamic light scattering (DLS) and transmission electron microscopy. The method was also evaluated for robustness in the range of 2.78 × 105-2.67 × 107 particles per sample. Notably, MALS-based particle counts correlated with the titer of infectious LVVs measured via transduction assays (R2 = 0.77). Using a combination of SEC-MALS and DLS, we discerned the effects of purification parameters on LVV quality, such as the separation between heterogeneous LV, which can facilitate critical decision-making in the biomanufacturing of gene and cell therapies.
Collapse
Affiliation(s)
- Sobhana A Sripada
- Department of Chemical and Biomolecular Engineering, NC State University, 911 Partners Way, Raleigh, North Carolina 27606, United States
- NC-VVIRAL, NC State University, 1840 Entrepreneur Dr, Raleigh, North Carolina 27606, United States
| | - Eduardo Barbieri
- Department of Chemical and Biomolecular Engineering, NC State University, 911 Partners Way, Raleigh, North Carolina 27606, United States
- LigaTrap Technologies LLC, 1791 Varsity Drive, Suite #150, Raleigh, North Carolina 27606, United States
| | - Shriarjun Shastry
- Department of Chemical and Biomolecular Engineering, NC State University, 911 Partners Way, Raleigh, North Carolina 27606, United States
- Biomanufacturing Training and Education Center, NC State University, 850 Oval Dr, Raleigh, North Carolina 27606, United States
- NC-VVIRAL, NC State University, 1840 Entrepreneur Dr, Raleigh, North Carolina 27606, United States
| | | | | | | | | | - Michael Daniele
- Department of Electrical and Computer Engineering, NC State University, 890 Oval Dr, Raleigh, North Carolina 27606, United States
- Joint Department of Biomedical Engineering, NC State University and UNC Chapel Hill, 1840 Entrepreneur Dr, Raleigh, North Carolina 27606, United States
- NC-VVIRAL, NC State University, 1840 Entrepreneur Dr, Raleigh, North Carolina 27606, United States
| | - Stefano Menegatti
- Department of Chemical and Biomolecular Engineering, NC State University, 911 Partners Way, Raleigh, North Carolina 27606, United States
- LigaTrap Technologies LLC, 1791 Varsity Drive, Suite #150, Raleigh, North Carolina 27606, United States
- Biomanufacturing Training and Education Center, NC State University, 850 Oval Dr, Raleigh, North Carolina 27606, United States
- NC-VVIRAL, NC State University, 1840 Entrepreneur Dr, Raleigh, North Carolina 27606, United States
| |
Collapse
|
13
|
Ajenu EO, Seideneck AM, Pandellapalli E, Shinsky EM, Humphries CL, Aparicio NL, Sharma M, Marden JH, Krasilnikova MM. ABCG2 transporter reduces protein aggregation in cigarette smoke condensate-exposed A549 lung cancer cells. PLoS One 2024; 19:e0297661. [PMID: 38442133 PMCID: PMC10914296 DOI: 10.1371/journal.pone.0297661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 01/09/2024] [Indexed: 03/07/2024] Open
Abstract
Cigarette smoke-induced protein aggregation damages the lung cells in emphysema and COPD; however, lung cancer cells continue to thrive, evolving to persist in the toxic environment. Here, we showed that upon the cigarette smoke condensate exposure, A549 lung cancer cells exhibit better survival and reduced level of protein aggregation when compared to non-cancerous Beas-2B and H-6053 cells. Our data suggests that upregulation of efflux pumps in cancer cells assists in reducing smoke toxicity. Specifically, we demonstrated that inhibition of the ABCG2 transporter in A549 by febuxostat or its downregulation by shRNA-mediated RNA interference resulted in a significant increase in protein aggregation due to smoke exposure.
Collapse
Affiliation(s)
- Emmanuella O. Ajenu
- Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, United states of America
| | - Ashley M. Seideneck
- Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, United states of America
| | - Esh Pandellapalli
- Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, United states of America
| | - Emily M. Shinsky
- Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, United states of America
| | - Casey L. Humphries
- Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, United states of America
| | - Nicholas L. Aparicio
- Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, United states of America
| | - Mahak Sharma
- Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, United states of America
| | - James H. Marden
- Department of Biology, Penn State University, University Park, Pennsylvania, United states of America
| | - Maria M. Krasilnikova
- Department of Biochemistry and Molecular Biology, Penn State University, University Park, Pennsylvania, United states of America
| |
Collapse
|
14
|
Jin M, Mi Y, Li F, Ren F, Deng Y, Zheng P. ZNF131 facilitates the growth of hepatocellular carcinoma by acting as a transcriptional activator of SMC4 expression. Biochem Biophys Res Commun 2024; 696:149515. [PMID: 38241815 DOI: 10.1016/j.bbrc.2024.149515] [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: 12/28/2023] [Accepted: 01/10/2024] [Indexed: 01/21/2024]
Abstract
ZNF131 is a Zinc finger protein that acts as a transcription factor with oncogenic effects in multiple cancers. In this study, we aimed to explore the alternative splicing profile of ZNF131 in hepatocellular carcinoma (HCC), its regulatory effects on cell-cycle progression, and the downstream effectors. ZNF131 transcriptional profile and HCC survival analysis were conducted using data from the Cancer Genome Atlas (TCGA)-Liver Hepatocellular Cancer (LIHC) dataset. Chromatin immunoprecipitation (ChIP)-qPCR and dual-luciferase reporter assays were utilized to explore transcriptional regulation. CCK-8, colony formation and xenograft tumor models were used to study HCC tumor growth. Results showed that ZNF131 isoform 2 is upregulated in HCC tissues and its upregulation was associated with unfavorable overall survival (OS) and progression-free interval (PFI). Knockdown of endogenous ZNF131 inhibits HCC cell growth and induces G2/M cell-cycle arrest. ZNF131 binds to the SMC4 promoter by interacting with ZBTB33 and the ZBTB33 recognizing motif. ZNF131 transcriptionally activates SMC4 expression in HCC cells. The tumor-suppressive effects of ZNF131 shRNA could be partially reversed by enforced SMC4 overexpression. In summary, this study highlights the ZNF131/ZBTB33/SMC4 axis as a driver of pathological cell cycling and proliferation in HCC.
Collapse
Affiliation(s)
- Meng Jin
- Henan Key Laboratory of Helicobacter Pylori & Microbiota and Gastrointestinal Cancer, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China
| | - Yang Mi
- Henan Key Laboratory of Helicobacter Pylori & Microbiota and Gastrointestinal Cancer, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China; Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Fazhan Li
- Henan Key Laboratory of Helicobacter Pylori & Microbiota and Gastrointestinal Cancer, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China
| | - Feifei Ren
- Henan Key Laboratory of Helicobacter Pylori & Microbiota and Gastrointestinal Cancer, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China
| | - Ying Deng
- Cancer Center, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, Chengdu, Sichuan, China
| | - Pengyuan Zheng
- Henan Key Laboratory of Helicobacter Pylori & Microbiota and Gastrointestinal Cancer, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China; Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
15
|
Alfar HR, Pariser DN, Chanzu H, Joshi S, Coenen DM, Lykins J, Prakhya KS, Potash MJ, Chao W, Kelschenbach J, Volsky DJ, Metcalf-Pate K, Whiteheart SW. Protocol for optimizing production and quality control of infective EcoHIV virions. STAR Protoc 2023; 4:102368. [PMID: 37342907 PMCID: PMC10511864 DOI: 10.1016/j.xpro.2023.102368] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/03/2023] [Accepted: 05/18/2023] [Indexed: 06/23/2023] Open
Abstract
EcoHIV is a model of HIV infection that recapitulates aspects of HIV-1 pathology in mice. However, there are limited published protocols to guide EcoHIV virion production. Here, we present a protocol for producing infective EcoHIV virions and essential quality controls. We describe steps for viral purification, titering, and multiple techniques to analyze infection efficacy. This protocol produces high infectivity in C57BL/6 mice which will aid investigators in generating preclinical data.
Collapse
Affiliation(s)
- Hammodah R Alfar
- University of Kentucky, College of Medicine, Lexington, KY 40506, USA
| | - Daphne N Pariser
- Massachusetts Institute of Technology, Division of Comparative Medicine, 77 Massachusetts Avenue, Cambridge, MA 02139, USA; Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Harry Chanzu
- University of Kentucky, College of Medicine, Lexington, KY 40506, USA
| | - Smita Joshi
- University of Kentucky, College of Medicine, Lexington, KY 40506, USA
| | - Daniëlle M Coenen
- University of Kentucky, College of Medicine, Lexington, KY 40506, USA
| | - Joshua Lykins
- University of Kentucky, College of Medicine, Lexington, KY 40506, USA
| | | | - Mary Jane Potash
- Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Wei Chao
- Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Jennifer Kelschenbach
- Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - David J Volsky
- Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY 10029, USA
| | - Kelly Metcalf-Pate
- Massachusetts Institute of Technology, Division of Comparative Medicine, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | | |
Collapse
|
16
|
Ahmadi SE, Shabestari RM, kojabad AA, Safa M. A straightforward microfluidic-based approach toward optimizing transduction efficiency of HIV-1-derived lentiviral vectors in BCP-ALL cells. BIOTECHNOLOGY REPORTS (AMSTERDAM, NETHERLANDS) 2023; 38:e00792. [PMID: 36950261 PMCID: PMC10025989 DOI: 10.1016/j.btre.2023.e00792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/27/2023] [Accepted: 03/10/2023] [Indexed: 03/13/2023]
Abstract
Background HIV-1-derived lentiviral vectors (LVs) are capable of transducing human cells by integrating the transgene into the host genome. In order to do that, LVs should have enough time and space to interact with the surface of the target cells. Herein, we used a microfluidic system to facilitate the transduction of BCP-ALL cells. Methods and Results We used a SU-8 mold to fabricate a PDMS microfluidic chip containing three channels with a 50 μm height and a surface matching 96-well plates. In order to produce LVs, we used HEK293T cells to package the second generation of LVs. First, we evaluated the cell recovery from the microfluidic chip. Cell recovery assessment showcased that 3 h and 6 h of incubation in microfluidic channels containing 100,000 NALM-6 (BCP-ALL) cells with 2μL of culture media yielded 87±7.2% and 80.6 ± 10% of cell recovery, respectively. Afterward, the effects of LV-induced toxicity were evaluated using 10-30% LV concentrations in time frames ranging from 3 h to 24 h. In 96-well plates, it took 12-24 h for the viruses with 20% and 30% concentrations to affect the cell survival significantly. These effects were intensified in the microfluidic system implying that microfluidic is capable of enhancing LV transduction. Based on the evidence of cell recovery and cell survival we chose 6 h of incubation with 20% LV. Conclusion The results from EGFP expression showcased that a microfluidic system could increase the LV transduction in BCP-ALL cells by almost 9-folds. All in all, the microfluidic system seems to be a great armamentarium in optimizing LV-based transduction.
Collapse
|
17
|
Brundu S, Napolitano V, Franzolin G, Lo Cascio E, Mastrantonio R, Sardo G, Cascardi E, Verginelli F, Sarnataro S, Gambardella G, Pisacane A, Arcovito A, Boccaccio C, Comoglio PM, Giraudo E, Tamagnone L. Mutated axon guidance gene PLXNB2 sustains growth and invasiveness of stem cells isolated from cancers of unknown primary. EMBO Mol Med 2023; 15:e16104. [PMID: 36722641 PMCID: PMC9994481 DOI: 10.15252/emmm.202216104] [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: 03/31/2022] [Revised: 12/28/2022] [Accepted: 01/11/2023] [Indexed: 02/02/2023] Open
Abstract
The genetic changes sustaining the development of cancers of unknown primary (CUP) remain elusive. The whole-exome genomic profiling of 14 rigorously selected CUP samples did not reveal specific recurring mutation in known driver genes. However, by comparing the mutational landscape of CUPs with that of most other human tumor types, it emerged a consistent enrichment of changes in genes belonging to the axon guidance KEGG pathway. In particular, G842C mutation of PlexinB2 (PlxnB2) was predicted to be activating. Indeed, knocking down the mutated, but not the wild-type, PlxnB2 in CUP stem cells resulted in the impairment of self-renewal and proliferation in culture, as well as tumorigenic capacity in mice. Conversely, the genetic transfer of G842C-PlxnB2 was sufficient to promote CUP stem cell proliferation and tumorigenesis in mice. Notably, G842C-PlxnB2 expression in CUP cells was associated with basal EGFR phosphorylation, and EGFR blockade impaired the viability of CUP cells reliant on the mutated receptor. Moreover, the mutated PlxnB2 elicited CUP cell invasiveness, blocked by EGFR inhibitor treatment. In sum, we found that a novel activating mutation of the axon guidance gene PLXNB2 sustains proliferative autonomy and confers invasive properties to stem cells isolated from cancers of unknown primary, in EGFR-dependent manner.
Collapse
Affiliation(s)
| | - Virginia Napolitano
- Department of Life Sciences and Public HealthUniversità Cattolica del Sacro CuoreRomeItaly
| | | | - Ettore Lo Cascio
- Department of Biotechnological Sciences and Intensive CareUniversità Cattolica del Sacro CuoreRomeItaly
| | - Roberta Mastrantonio
- Department of Life Sciences and Public HealthUniversità Cattolica del Sacro CuoreRomeItaly
| | | | - Eliano Cascardi
- Candiolo Cancer InstituteFPO‐IRCCSTurinItaly
- Department of Medical SciencesUniversity of TurinTurinItaly
| | | | | | - Gennaro Gambardella
- Telethon Institute of Genetic and MedicinePozzuoliItaly
- Department of Electrical Engineering and Information TechnologyUniversity of Naples Federico IINaplesItaly
| | | | - Alessandro Arcovito
- Department of Biotechnological Sciences and Intensive CareUniversità Cattolica del Sacro CuoreRomeItaly
- Fondazione Policlinico Gemelli (FPG) – IRCCSRomeItaly
| | - Carla Boccaccio
- Candiolo Cancer InstituteFPO‐IRCCSTurinItaly
- Department of OncologyUniversity of TurinTurinItaly
| | | | - Enrico Giraudo
- Candiolo Cancer InstituteFPO‐IRCCSTurinItaly
- Department of Science and Drug TechnologyUniversity of TurinTurinItaly
| | - Luca Tamagnone
- Department of Life Sciences and Public HealthUniversità Cattolica del Sacro CuoreRomeItaly
- Fondazione Policlinico Gemelli (FPG) – IRCCSRomeItaly
| |
Collapse
|
18
|
Xiong Q, Zhang H, Ji X, Zhang Y, Shi G, Dai L, Cheng F, Wang H, Luo J, Xu J, Ji Y, Su X, Yang W, Zhang L, Deng H. A novel membrane-bound interleukin-2 promotes NK-92 cell persistence and anti-tumor activity. Oncoimmunology 2022; 11:2127282. [PMID: 36185809 PMCID: PMC9519007 DOI: 10.1080/2162402x.2022.2127282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
A major challenge in natural killer (NK) cell immunotherapy is the limited persistence of NK cells in vivo. However, the proliferation of NK cells is dependent on cytokines such as interleukin-2 (IL-2). Although IL-2 is a critical cytokine for NK cell activation and survival, IL-2 administration in adoptive NK cell therapy can induce adverse toxicities. To improve the persistence of NK cells and attenuate the systemic toxicity of IL-2, we constructed a cell-restricted artificial IL-2, named membrane-bound IL-2 (mbIL-2), comprising human IL-2 and human IL-2Rα joined by a classic linker. We found that mbIL-2-activated NK-92 cells can survive and proliferate in vitro and in vivo, independent of exogenous IL-2, while mbIL-2-expressing NK-92 cells do not support bystander cell survival or proliferation. Additionally, mbIL-2 enhanced NK-92 cell-mediated antitumor activity by tuning the IL-2 receptor downstream signals and NK cell receptor repertoire expression. To conclude, our novel mbIL-2 improves NK-92 cell persistence and enhances NK-92 cell-mediated antitumor activity. NK-92 cells genetically modified to express the novel mbIL-2 with potential significance for clinical development.
Collapse
Affiliation(s)
- Qi Xiong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, P.R. China
| | - Hantao Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, P.R. China
| | - Xuanle Ji
- The College of Life Sciences, Sichuan University, Chengdu, P.R. China
| | - Yong Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, P.R. China
| | - Gang Shi
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, P.R. China
| | - Lei Dai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, P.R. China
| | - Fuyi Cheng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, P.R. China
| | - Huiling Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, P.R. China
| | - Jieyan Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, P.R. China
| | - Jia Xu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, P.R. China
| | - Yanhong Ji
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, P.R. China
| | - Xiaolan Su
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, P.R. China
| | - Weixiao Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, P.R. China
| | - Lin Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, P.R. China
| | - Hongxin Deng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, P.R. China
- Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Beijing, P.R. China
| |
Collapse
|
19
|
Protocol for viral vector-mediated measurement of transcription factor activity of mouse brain. STAR Protoc 2022; 3:101633. [PMID: 36035800 PMCID: PMC9405533 DOI: 10.1016/j.xpro.2022.101633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Here, we provide a step-by-step protocol to measure the activities of multiple transcription factors (TFs) in the same mouse brain. This protocol includes a procedure to construct a virus-based TF activity reporter, in utero transfection, and PCR-based measurement of TF activity to obtain the transcription factor activity profile (TFAP). Our protocol facilitates a systematic analysis of TF activity of the brain in vivo and will aid trans-omics understanding of the molecular mechanism underlying the brain functions. For complete details on the use and execution of this protocol, please refer to Abe and Abe, (2022). Protocol allows quantitative measurement of TF activity in mouse brain A protocol for preparation and in utero injection of LV-based transcription reporter Workflow of behavioral experiment and brain sampling for TF activity measurement
Publisher’s note: Undertaking any experimental protocol requires adherence to local institutional guidelines for laboratory safety and ethics.
Collapse
|
20
|
Quach ABV, Little SR, Shih SCC. Viral Generation, Packaging, and Transduction on a Digital Microfluidic Platform. Anal Chem 2022; 94:4039-4047. [PMID: 35192339 DOI: 10.1021/acs.analchem.1c05227] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Viral-based systems are a popular delivery method for introducing exogenous genetic material into mammalian cells. Unfortunately, the preparation of lentiviruses containing the machinery to edit the cells is labor-intensive, with steps requiring optimization and sensitive handling. To mitigate these challenges, we introduce the first microfluidic method that integrates lentiviral generation, packaging, and transduction. The new method allows the production of viral titers between 106 and 107 (similar to macroscale production) and high transduction efficiency for hard-to-transfect cell lines. We extend the technique for gene editing applications and show how this technique can be used to knock out and knock down estrogen receptor gene─a gene prominently responsible for 70% of breast cancer cases. This new technique is automated with multiplexing capabilities, which have the potential to standardize the methods for viral-based genome engineering.
Collapse
Affiliation(s)
- Angela B V Quach
- Department of Biology, Concordia University, 7141 Sherbrooke Street West, Montréal, Québec H4B 1R6, Canada.,Centre for Applied Synthetic Biology, Concordia University, 7141 Sherbrooke Street West, Montréal, Québec H4B 1R6, Canada
| | - Samuel R Little
- Centre for Applied Synthetic Biology, Concordia University, 7141 Sherbrooke Street West, Montréal, Québec H4B 1R6, Canada.,Department of Electrical and Computer Engineering, Concordia University, 1455 de Maisonneuve Blvd. West, Montréal, Québec H3G 1M8, Canada
| | - Steve C C Shih
- Department of Biology, Concordia University, 7141 Sherbrooke Street West, Montréal, Québec H4B 1R6, Canada.,Centre for Applied Synthetic Biology, Concordia University, 7141 Sherbrooke Street West, Montréal, Québec H4B 1R6, Canada.,Department of Electrical and Computer Engineering, Concordia University, 1455 de Maisonneuve Blvd. West, Montréal, Québec H3G 1M8, Canada
| |
Collapse
|
21
|
Di Franco S, Zhang L, Gaggianesi M, Lo Iacono M, Medema JP, Stassi G. FACS-based protocol to assess cytotoxicity and clonogenic potential of colorectal cancer stem cells using a Wnt/β-catenin signaling pathway reporter. STAR Protoc 2021; 2:100880. [PMID: 34712995 PMCID: PMC8529548 DOI: 10.1016/j.xpro.2021.100880] [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] [Indexed: 12/29/2022] Open
Abstract
Cancer stem cells (CSCs) play a key role in tumor initiation and progression. A real-time tool to evaluate the activation of CSC-specific signaling pathways is crucial for the study of this cancer cell subset. Here, we present a protocol to monitor, in vitro, the activation of Wnt/β-catenin signaling pathway, which is considered a functional biomarker for colorectal CSCs (CR-CSCs). This flow-cytometry-based protocol allows it to isolate CR-CSCs and to evaluate their cytotoxicity upon anti-tumor treatments. For complete details on the use and execution of this protocol, please refer to Di Franco et al. (2021).
Collapse
Affiliation(s)
- Simone Di Franco
- Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Palermo 90127, Italy
| | - Le Zhang
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands.,Oncode Institute, Amsterdam UMC, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Miriam Gaggianesi
- Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Palermo 90127, Italy
| | - Melania Lo Iacono
- Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Palermo 90127, Italy
| | - Jan Paul Medema
- Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Cancer Center Amsterdam, Amsterdam UMC, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands.,Oncode Institute, Amsterdam UMC, University of Amsterdam, Amsterdam 1105 AZ, the Netherlands
| | - Giorgio Stassi
- Department of Surgical Oncological and Stomatological Sciences, University of Palermo, Palermo 90127, Italy
| |
Collapse
|
22
|
Portillo AL, Hogg R, Ashkar AA. Production of human CAR-NK cells with lentiviral vectors and functional assessment in vitro. STAR Protoc 2021; 2:100956. [PMID: 34825217 PMCID: PMC8605094 DOI: 10.1016/j.xpro.2021.100956] [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] [Indexed: 11/23/2022] Open
Abstract
Although natural killer (NK) cells have become a promising immune effector cell for chimeric antigen receptor (CAR)-based therapy, generating human CAR-NK cells with high transgene efficiency has been challenging. In this protocol, we describe how to generate CAR-NK cells with transduction efficiencies >15% from healthy donor ex vivo expanded NK cells using third generation lentiviral vectors (LVs). We also show how to assess CAR-NK cell anti-tumor function in vitro using a flow cytometry-based killing assay. For complete details on the use and execution of this protocol, please refer to Portillo et al. (2021). PBMC isolation and expansion of human NK cells from healthy donors Production of third generation lentiviral vectors for NK cell transduction A protocol for the generation of CAR-modified NK cells from human expanded NK cells A flow cytometry-based assay to assess CAR-NK cell cytotoxicity in vitro
Collapse
Affiliation(s)
- Ana L Portillo
- Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada.,McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Richard Hogg
- Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada.,McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Ali A Ashkar
- Department of Medicine, McMaster University, Hamilton, ON L8N 3Z5, Canada.,McMaster Immunology Research Centre, McMaster University, Hamilton, ON L8S 4K1, Canada
| |
Collapse
|
23
|
Bow AJ, Masi TJ, Dhar MS. Etched 3D-Printed Polycaprolactone Constructs Functionalized with Reduced Graphene Oxide for Enhanced Attachment of Dental Pulp-Derived Stem Cells. Pharmaceutics 2021; 13:2146. [PMID: 34959426 PMCID: PMC8704510 DOI: 10.3390/pharmaceutics13122146] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/03/2021] [Accepted: 12/10/2021] [Indexed: 01/08/2023] Open
Abstract
A core challenge in the field of tissue engineering is the ability to establish pipeline workflows for the design and characterization of scaffold technologies with clinically translatable attributes. The parallel development of biomaterials and stem cell populations represents a self-sufficient and streamlined approach for establishing such a pipeline. In the current study, rat dental pulp stem cell (rDPSC) populations were established to assess functionalized polycaprolactone (PCL) constructs. Initial optimization and characterization of rDPSC extraction and culture conditions confirmed that cell populations were readily expandable and demonstrated surface markers associated with multi-potency. Subset populations were transduced to express DsRed fluorescent protein as a mechanism of tracking both cells and cell-derived extracellular matrix content on complex scaffold architecture. Thermoplastic constructs included reduced graphene oxide (rGO) as an additive to promote cellular attachment and were further modified by surface etching a weak acetic acid solution to roughen surface topographical features, which was observed to dramatically improve cell surface coverage in vitro. Based on these data, the modified rGO-functionalized PCL constructs represent a versatile platform for bone tissue engineering, capable of being applied as a standalone matrix or in conjunction with bio-active payloads such as DPSCs or other bio-inks.
Collapse
Affiliation(s)
- Austin J. Bow
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37931, USA;
| | - Thomas J. Masi
- School of Medicine, University of Tennessee Graduate, Knoxville, TN 37920, USA;
| | - Madhu S. Dhar
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37931, USA;
| |
Collapse
|
24
|
Abstract
The CRISPR-Cas system coupled with Combinatorial Genetics En Masse (CombiGEM) enables systematic analysis of high-order genetic perturbations that are important for understanding biological processes and discovering therapeutic target combinations. Here, we present detailed steps and technical considerations for building multiplexed guide RNA libraries and carrying out a combinatorial CRISPR screen in mammalian cells. We also present an analytical pipeline, CombiPIPE, for mapping two- and three-way genetic interactions. For complete details on the use and execution of this protocol, please refer to Zhou et al. (2020).
Collapse
Affiliation(s)
- Peng Zhou
- Laboratory of Combinatorial Genetics and Synthetic Biology, School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Yuk Kei Wan
- Laboratory of Combinatorial Genetics and Synthetic Biology, School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Becky K.C. Chan
- Laboratory of Combinatorial Genetics and Synthetic Biology, School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Gigi C.G. Choi
- Laboratory of Combinatorial Genetics and Synthetic Biology, School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Alan S.L. Wong
- Laboratory of Combinatorial Genetics and Synthetic Biology, School of Biomedical Sciences, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
- Department of Electrical and Electronic Engineering, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| |
Collapse
|