1
|
de Souza AP, Bastos AP, da Fonseca FN, Pandolfi JR, Costamilan CADVLR, Marques MG. Polyethyleneimine-mediated gene transfection in porcine fetal fibroblasts. Anim Reprod 2024; 21:e20240026. [PMID: 39629009 PMCID: PMC11614137 DOI: 10.1590/1984-3143-ar2024-0026] [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: 05/06/2024] [Accepted: 09/04/2024] [Indexed: 12/06/2024] Open
Abstract
Polyethylenimine (PEI) has been explored as an efficient non-viral system for delivering genes to cells; however, there were no protocols for its use in porcine fetal fibroblasts (PFF). Therefore, we compared different concentrations of FITC-PEI (0.625, 1.25, 2.5, 5, 10, 20, 40, or 80 µg/mL) and incubation times (30 min, 1 h, or 2 h). It was observed that the incubation time did not affect the internalization of the PEI-FITC and that 30 min was sufficient to capture the complex. The concentrations higher than 10 µg/mL could reach many marked PFF (>90%). Then, two PEI concentrations were tested, 10 or 40 µg/mL, combined with an N/P of 2 with the pmhyGENIE-5 for 30 min. The percentage of PFF-GFP positive was similar between the PEI concentrations in the evaluation time points (24 h, 48 h, and 72 h). However, 40 µg/mL caused higher membrane damage rates. Thus, it can be concluded that concentrations between 10 - 80 µg/ml of PEI promote high incorporation rates, even in periods as short as 30 minutes. Furthermore, it can be stated that the transfection condition used in Polyplexes 1 (10 µg/mL of PEI and 37.5 µg/mL of pmhyGENIE-5 for 30 min) efficiently produces genetically edited porcine fetal fibroblasts with low cell damage.
Collapse
Affiliation(s)
| | - Ana Paula Bastos
- Empresa Brasileira de Pesquisa Agropecuária, Embrapa Suínos e Aves, Concórdia, SC, Brasil
| | | | - José Rodrigo Pandolfi
- Empresa Brasileira de Pesquisa Agropecuária, Embrapa Suínos e Aves, Concórdia, SC, Brasil
| | | | - Mariana Groke Marques
- Empresa Brasileira de Pesquisa Agropecuária, Embrapa Suínos e Aves, Concórdia, SC, Brasil
- Pós-Graduação em Produção e Sanidade Animal, Instituto Federal Catarinense, Concórdia, SC, Brasil
| |
Collapse
|
2
|
Bai JH, Yu QT, Wang YW, Chen CF, Ma LJ, Yuan Y, Gan YJ, Yang JQ, Zhu S, Ran YH, Zhang LL, Qian H, Zhao ZH, Liu Q. Polyethyleneimine-mediated assembly of DNA nanotubes for KRAS siRNA delivery in lung adenocarcinoma therapy. J Mater Chem B 2024; 12:6442-6451. [PMID: 38860876 DOI: 10.1039/d4tb00791c] [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: 06/12/2024]
Abstract
Self-assembled DNA nanostructures hold great promise in biosensing, drug delivery and nanomedicine. Nevertheless, challenges like instability and inefficiency in cellular uptake of DNA nanostructures under physiological conditions limit their practical use. To tackle these obstacles, this study proposes a novel approach that integrates the cationic polymer polyethyleneimine (PEI) with DNA self-assembly. The hypothesis is that the positively charged linear PEI can facilitate the self-assembly of DNA nanostructures, safeguard them against harsh conditions and impart them with the cellular penetration characteristic of PEI. As a demonstration, a DNA nanotube (PNT) was successfully synthesized through PEI mediation, and it exhibited significantly enhanced stability and cellular uptake efficiency compared to conventional Mg2+-assembled DNA nanotubes. The internalization mechanism was further found to be both clathrin-mediated and caveolin-mediated endocytosis, influenced by both PEI and DNA. To showcase the applicability of this hybrid nanostructure for biomedical settings, the KRAS siRNA-loaded PNT was efficiently delivered into lung adenocarcinoma cells, leading to excellent anticancer effects in vitro. These findings suggest that the PEI-mediated DNA assembly could become a valuable tool for future biomedical applications.
Collapse
Affiliation(s)
- Jia-Hao Bai
- Laboratory of Pharmacy and Chemistry, and Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, Chongqing 400016, China.
| | - Qing-Tao Yu
- Laboratory of Pharmacy and Chemistry, and Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, Chongqing 400016, China.
| | - Yi-Wu Wang
- Laboratory of Pharmacy and Chemistry, and Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, Chongqing 400016, China.
| | - Chun-Fa Chen
- Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Street, Chongqing 400037, China.
| | - Lian-Ju Ma
- Laboratory of Pharmacy and Chemistry, and Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, Chongqing 400016, China.
| | - Yue Yuan
- Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Street, Chongqing 400037, China.
| | - Yong-Jun Gan
- Laboratory of Pharmacy and Chemistry, and Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, Chongqing 400016, China.
| | - Jia-Qi Yang
- Laboratory of Pharmacy and Chemistry, and Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, Chongqing 400016, China.
| | - Shu Zhu
- Laboratory of Pharmacy and Chemistry, and Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, Chongqing 400016, China.
| | - Yu-Hang Ran
- Laboratory of Pharmacy and Chemistry, and Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, Chongqing 400016, China.
| | - Li-Lei Zhang
- Laboratory of Pharmacy and Chemistry, and Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, Chongqing 400016, China.
| | - Hang Qian
- Institute of Respiratory Diseases, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Street, Chongqing 400037, China.
| | - Zheng-Huan Zhao
- College of Basic Medical Sciences, Chongqing Medical University, Chongqing 400016, China.
| | - Qian Liu
- Laboratory of Pharmacy and Chemistry, and Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
3
|
Affinito A, Quintavalle C, Chianese RV, Roscigno G, Fiore D, D'Argenio V, Thomas G, Savarese A, Ingenito F, Cocca L, Nuzzo S, Berezovski MV, Stoppelli MP, Condorelli G. MCT4-driven CAF-mediated metabolic reprogramming in breast cancer microenvironment is a vulnerability targetable by miR-425-5p. Cell Death Discov 2024; 10:140. [PMID: 38485929 PMCID: PMC10940713 DOI: 10.1038/s41420-024-01910-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 02/26/2024] [Accepted: 03/06/2024] [Indexed: 03/18/2024] Open
Abstract
Multiple oncogenic alterations contribute to breast cancer development. Metabolic reprogramming, deeply contributing to tumor microenvironment (TME) education, is now widely recognized as a hallmark of cancer. The reverse Warburg effect induces cancer-associated fibroblasts (CAFs) to produce and secrete L-lactate, enhancing malignant characteristics such as neoangiogenesis, metastatic dissemination, and treatment resistance. Monocarboxylate transporter (MCT) 4 is involved in lactate efflux from CAFs into stromal and epithelial cells. Here, we first assess the expression of miR-425-5p and its target MCT4 in breast cancer CAFs and normal fibroblasts. We analyzed the metabolic changes induced by miR-425-5p in CAFs and its role in the education of breast cancer epithelial cells. We show that miR-425-5p-induced MCT4 knockdown decreased lactate extrusion from CAFs and its availability in the TME. miR-425-5p overexpression induced profound metabolic transformation in CAFs, ultimately influencing breast cancer metabolism. Furthermore, miR-425-5p impaired the capacity of CAFs to sustain vessel formation and breast cancer cell migration, viability, and proliferation. These findings emphasize the key role of miR-425-5p in breast cancer metabolism and aggressiveness, and its possible importance for breast cancer therapy and monitoring.
Collapse
Affiliation(s)
- Alessandra Affinito
- Department of Molecular Medicine and Medical Biotechnology, "Federico II" University of Naples, Naples, Italy
- AKA Biotech, Naples, Italy
| | - Cristina Quintavalle
- Institute Experimental Endocrinology and Oncology "Gaetano Salvatore" (IEOS), CNR, Naples, Italy
| | - Rosario Vincenzo Chianese
- Department of Molecular Medicine and Medical Biotechnology, "Federico II" University of Naples, Naples, Italy
| | - Giuseppina Roscigno
- Department of Molecular Medicine and Medical Biotechnology, "Federico II" University of Naples, Naples, Italy
| | - Danilo Fiore
- Department of Molecular Medicine and Medical Biotechnology, "Federico II" University of Naples, Naples, Italy
- Institute Experimental Endocrinology and Oncology "Gaetano Salvatore" (IEOS), CNR, Naples, Italy
| | - Valeria D'Argenio
- Department of Human Sciences and Quality of Life Promotion, San Raffaele Open University, Roma, Italy
- CEINGE-Biotecnologie Avanzate Franco Salvatore, Napoli, Italy
| | | | - Alessia Savarese
- Department of Molecular Medicine and Medical Biotechnology, "Federico II" University of Naples, Naples, Italy
| | - Francesco Ingenito
- Department of Molecular Medicine and Medical Biotechnology, "Federico II" University of Naples, Naples, Italy
| | - Lorenza Cocca
- Department of Molecular Medicine and Medical Biotechnology, "Federico II" University of Naples, Naples, Italy
| | | | - Maxim V Berezovski
- Department of Chemistry and Biomolecular Sciences and John L. Holmes Mass Spectrometry Facility, University of Ottawa, Ottawa, ON, Canada
| | | | - Gerolama Condorelli
- Department of Molecular Medicine and Medical Biotechnology, "Federico II" University of Naples, Naples, Italy.
- Institute Experimental Endocrinology and Oncology "Gaetano Salvatore" (IEOS), CNR, Naples, Italy.
| |
Collapse
|
4
|
Cheng B, Li Y, Ji YB, Shi W, Li M, Zheng J, Ding L, Liu K, Fang L, Xu Y, Li H, Shao X. Polyethylenimine Triggers Dll4 Degradation to Regulate Angiogenesis In Vitro. ACS OMEGA 2024; 9:7502-7510. [PMID: 38405519 PMCID: PMC10882680 DOI: 10.1021/acsomega.3c06050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 11/28/2023] [Accepted: 01/17/2024] [Indexed: 02/27/2024]
Abstract
The Dll4-Notch signaling pathway plays a crucial role in the regulation of angiogenesis and is a promising therapeutic target for diseases associated with abnormal angiogenesis, such as cancer and ophthalmic diseases. Here, we find that polyethylenimine (PEI), a cationic polymer widely used as nucleic acid transfection reagents, can target the Notch ligand Dll4. By immunostaining and immunoblotting, we demonstrate that PEI significantly induces the clearance of cell-surface Dll4 and facilitates its degradation through the lysosomal pathway. As a result, the activation of Notch signaling in endothelial cells is effectively inhibited by PEI, as evidenced by the observed decrease in the generation of the activated form of Notch and expression of Notch target genes Hes1 and Hey1. Furthermore, through blocking Dll4-mediated Notch signaling, PEI treatment enhances angiogenesis in vitro. Together, our study reveals a novel biological effect of PEI and establishes a foundation for the development of a Dll4-targeted biomaterial for the treatment of angiogenesis-related disease.
Collapse
Affiliation(s)
- Binghua Cheng
- Guangdong
Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials,
Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanyan Li
- Guangdong
Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials,
Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- University
of Chinese Academy of Sciences, Beijing 100049, China
| | - Ya-Bin Ji
- Guangdong
Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials,
Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Wenli Shi
- Guangdong
Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials,
Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Meiqing Li
- Guangdong
Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials,
Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Sino-Euro
Center of Biomedicine and Health, Shenzhen 518024, China
| | - Jiwei Zheng
- Guangdong
Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials,
Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Li Ding
- Guangdong
Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials,
Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Ke Liu
- Guangdong
Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials,
Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Sino-Euro
Center of Biomedicine and Health, Shenzhen 518024, China
| | - Lijing Fang
- Guangdong
Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials,
Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Sino-Euro
Center of Biomedicine and Health, Shenzhen 518024, China
| | - Ye Xu
- General
Hospital of Southern Theatre Command, Guangzhou 510010, China
| | - Hongchang Li
- Guangdong
Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials,
Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Sino-Euro
Center of Biomedicine and Health, Shenzhen 518024, China
| | - Ximing Shao
- Guangdong
Key Laboratory of Nanomedicine, CAS-HK Joint Lab of Biomaterials,
Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Sino-Euro
Center of Biomedicine and Health, Shenzhen 518024, China
| |
Collapse
|
5
|
He S, Fu X, Wang L, Xue Y, Zhou L, Qiao S, An J, Xia T. Self-Assemble Silk Fibroin Microcapsules for Cartilage Regeneration through Gene Delivery and Immune Regulation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2302799. [PMID: 37264755 DOI: 10.1002/smll.202302799] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/22/2023] [Indexed: 06/03/2023]
Abstract
Effective treatments for cartilage defects are currently lacking. Gene delivery using proper delivery systems has shown great potential in cartilage regeneration. However, the inflammatory microenvironment generated by the defected cartilage severely affects the system's delivery efficiency. Therefore, this study reports a silk fibroin microcapsule (SFM) structure based on layer-by-layer self-assembly, in which interleukin-4 (IL-4) is modified on silk by click chemistry and loaded with lysyl oxidase plasmid DNA (LOX pDNA). The silk microcapsules display good biocompatibility and the release rate of genes can be adjusted by controlling the number of self-assembled layers. Moreover, the functionalized SFMs mixed with methacrylated gelatin (GelMA) exhibit good injectability. The IL-4 on the outer layer of the SFM can regulate macrophages to polarize toward the M2 type, thereby promoting cartilage matrix repair and inhibiting inflammation. The LOX pDNA loaded inside can be effectively delivered into cells to promote extracellular matrix generation, significantly promoting cartilage regeneration. The results of this study provide a promising biomaterial for cartilage repair, and this novel silk-based microcapsule delivery system can also provide strategies for the treatment of other diseases.
Collapse
Affiliation(s)
- Shuangjian He
- Department of orthopedics, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, 215000, China
| | - Xuejie Fu
- Institute of Clinical Medicine Research, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, 215000, China
| | - Liang Wang
- Department of orthopedics, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, 215000, China
| | - Yangyang Xue
- Department of orthopedics, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, 215000, China
| | - Long Zhou
- Department of orthopedics, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, 215000, China
| | - Shigang Qiao
- Institute of Clinical Medicine Research, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, 215000, China
| | - Jianzhong An
- Institute of Clinical Medicine Research, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, 215000, China
| | - Tingting Xia
- Institute of Clinical Medicine Research, Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, 215000, China
| |
Collapse
|
6
|
Martínez-Morales JC, González-Ruiz KD, Romero-Ávila MT, Rincón-Heredia R, Reyes-Cruz G, García-Sáinz JA. Lysophosphatidic acid receptor LPA 1 trafficking and interaction with Rab proteins, as evidenced by Förster resonance energy transfer. Mol Cell Endocrinol 2023; 570:111930. [PMID: 37054840 DOI: 10.1016/j.mce.2023.111930] [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: 02/10/2023] [Revised: 03/31/2023] [Accepted: 04/06/2023] [Indexed: 04/15/2023]
Abstract
LPA1 internalization to endosomes was studied employing Förster Resonance Energy Transfer (FRET) in cells coexpressing the mCherry-lysophosphatidic acid LPA1 receptors and distinct eGFP-tagged Rab proteins. Lysophosphatidic acid (LPA)-induced internalization was rapid and decreased afterward: phorbol myristate acetate (PMA) action was slower and sustained. LPA stimulated LPA1-Rab5 interaction rapidly but transiently, whereas PMA action was rapid but sustained. Expression of a Rab5 dominant-negative mutant blocked LPA1-Rab5 interaction and receptor internalization. LPA-induced LPA1-Rab9 interaction was only observed at 60 min, and LPA1-Rab7 interaction after 5 min with LPA and after 60 min with PMA. LPA triggered immediate but transient rapid recycling (i.e., LPA1-Rab4 interaction), whereas PMA action was slower but sustained. Agonist-induced slow recycling (LPA1-Rab11 interaction) increased at 15 min and remained at this level, whereas PMA action showed early and late peaks. Our results indicate that LPA1 receptor internalization varies with the stimuli.
Collapse
Affiliation(s)
| | - Karla D González-Ruiz
- Departamento de Biología Celular y Desarrollo, Ap. Postal 70-600, Ciudad de México, 04510, Mexico
| | - M Teresa Romero-Ávila
- Departamento de Biología Celular y Desarrollo, Ap. Postal 70-600, Ciudad de México, 04510, Mexico
| | - Ruth Rincón-Heredia
- Unidad de Imagenología, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad Universitaria, Ap. Postal 70-600, Ciudad de México, 04510, Mexico
| | - Guadalupe Reyes-Cruz
- Departamento de Biología Celular, Centro de Investigación y Estudios Avanzados-Instituto Politécnico Nacional, Colonia San Pedro Zacatenco, Ciudad de México, 07360, Mexico
| | | |
Collapse
|
7
|
Chen W, Wang C, Liu W, Zhao B, Zeng Z, Long F, Wang C, Li S, Lin N, Zhou J. A Matrix-Metalloproteinase-Responsive Hydrogel System for Modulating the Immune Microenvironment in Myocardial Infarction. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2209041. [PMID: 36754377 DOI: 10.1002/adma.202209041] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 02/02/2023] [Indexed: 06/18/2023]
Abstract
Injectable hydrogels carrying therapeutic factors to modulate the infarct immune microenvironment show great potential in the treatment of myocardial infarction (MI). However, conventional injectable hydrogels release therapeutic factors in an uncontrolled manner, which leads to poor treatment efficacy and acute side effects on normal tissues. In this work, a matrix metalloproteinase (MMP)2/9-responsive hydrogel system (MPGC4) is developed, considering the characteristics of the post-MI microenvironment. MPGC4 consists of tetra-poly(ethylene glycol) (PEG) hydrogels and a composite gene nanocarrier (CTL4) that is composed of carbon dots (CDots) coupled with interleukin-4 plasmid DNA via electrostatic interactions. MPGC4 can be automatically triggered to release CTL4 on demand after MI to regulate the infarct immune microenvironment. In addition, due to the photoluminescence properties of CDots, a large amount of viscoelastic MPGC4 is found to be retained in situ after injection into the infarct region without leakage. The in vitro results demonstrate that CTL4 promotes proinflammatory M1 macrophage polarization to the anti-inflammatory M2 subtype and contributes to cardiomyocyte survival through macrophage transition. In a rat model of MI, MPGC4 clears MMPs and precisely targets CTL4 to the infarcted region. In particular, MPGC4 improves cardiac function by modulating macrophage transition to reduce early inflammatory responses and proangiogenic activity.
Collapse
Affiliation(s)
- Wei Chen
- Beijing Institute of Basic Medical Sciences, 27 Taiping Rd, Beijing, 100850, P. R. China
- The Higher Educational Key Laboratory for Biomedical Engineering of Fujian Province, Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, 422 Siming Nan Road, Xiamen, 361005, P. R. China
| | - Changyong Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Rd, Beijing, 100850, P. R. China
| | - Wei Liu
- Beijing Institute of Basic Medical Sciences, 27 Taiping Rd, Beijing, 100850, P. R. China
| | - Bicheng Zhao
- The Higher Educational Key Laboratory for Biomedical Engineering of Fujian Province, Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, 422 Siming Nan Road, Xiamen, 361005, P. R. China
| | - Zhicheng Zeng
- The Higher Educational Key Laboratory for Biomedical Engineering of Fujian Province, Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, 422 Siming Nan Road, Xiamen, 361005, P. R. China
| | - Fen Long
- The Higher Educational Key Laboratory for Biomedical Engineering of Fujian Province, Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, 422 Siming Nan Road, Xiamen, 361005, P. R. China
| | - Chunlan Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Rd, Beijing, 100850, P. R. China
| | - Siwei Li
- Beijing Institute of Basic Medical Sciences, 27 Taiping Rd, Beijing, 100850, P. R. China
| | - Naibo Lin
- The Higher Educational Key Laboratory for Biomedical Engineering of Fujian Province, Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, 422 Siming Nan Road, Xiamen, 361005, P. R. China
| | - Jin Zhou
- Beijing Institute of Basic Medical Sciences, 27 Taiping Rd, Beijing, 100850, P. R. China
| |
Collapse
|
8
|
Ren R, Guo J, Liu G, Kang H, Machens HG, Schilling AF, Slobodianski A, Zhang Z. Nucleic acid direct delivery to fibroblasts: a review of nucleofection and applications. J Biol Eng 2022; 16:30. [PMID: 36329479 PMCID: PMC9635183 DOI: 10.1186/s13036-022-00309-5] [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: 07/08/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022] Open
Abstract
The fibroblast is one of the ideal target cell candidates for cell-based gene therapy approaches to promote tissue repair. Gene delivery to fibroblasts by viral transfection has been confirmed to have high transfection efficiency. However, in addition to immunogenic effects of viruses, the random integration of viral genes may damage the genome, affect the cell phenotype or even cause cancerous mutations in the transfected cells. Due to these potential biohazards and unknown long-term risks, the clinical use of viral transfection has been very limited. In contrast, initial non-viral transfection methods have been simple and safe to implement, with low immunogenicity, insertional mutagenesis, and risk of carcinogenesis, but their transfection efficiency has been relatively low. Nucleofection, a more recent non-viral transfection method, now combines the advantages of high transfection efficiency and direct nucleic acid delivery to the nucleus with a high safety.Here, we reviewed recent articles on fibroblast nucleofection, summarized different research points, improved methods and application scopes, and opened up ideas for promoting the further improvement and development of fibroblast nucleofection to meet the needs of a variety of disease research and clinical applications.
Collapse
Affiliation(s)
- Ranyue Ren
- grid.412793.a0000 0004 1799 5032Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei China
| | - Jiachao Guo
- grid.412793.a0000 0004 1799 5032Department of Pediatric Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei China
| | - Guangwu Liu
- grid.412793.a0000 0004 1799 5032Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei China
| | - Hao Kang
- grid.412793.a0000 0004 1799 5032Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei China
| | - Hans-Günther Machens
- grid.15474.330000 0004 0477 2438Department of Plastic Surgery and Hand Surgery, Faculty of Medicine, Klinikum Rechts Der Isar, Technische Universität München, Munich, Germany
| | - Arndt F. Schilling
- grid.411984.10000 0001 0482 5331Department of Trauma Surgery, Orthopaedics and Plastic Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Alex Slobodianski
- grid.15474.330000 0004 0477 2438Department of Plastic Surgery and Hand Surgery, Faculty of Medicine, Klinikum Rechts Der Isar, Technische Universität München, Munich, Germany
| | - Ziyang Zhang
- grid.412793.a0000 0004 1799 5032Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei China
| |
Collapse
|
9
|
Shayestehfar M, Farahi S, Kheiri Yeganeh Azar B, Memari A, Baluchnejadmojarad T, Faghihi F. Generating Human Induced Pluripotent Stem Cell Via Low-Dose Polyethylenimine-Mediated Transfection: An Optimized Protocol. DNA Cell Biol 2022; 41:903-916. [PMID: 35984994 DOI: 10.1089/dna.2022.0331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Human dermal fibroblasts (HDFs) can be reprogrammed through different strategies to generate human induced pluripotent stem cells (hiPSCs). However, most of these strategies require high-cost materials and specific equipment not readily accessible in most laboratories. Hence, liposomal and virus-based techniques can replace with polyethylenimine (PEI)-mediated transfection to overcome these challenges. However, few researchers have addressed the PEI's ability to transfect HDFs. This study used PEI reagent to transfer oriP/EBNA1-based vector into HDFs to produce hiPSC lines. We first described conditions allowing the efficient transfection of HDFs with low cytotoxicity and without specific types of equipment and optimized several parameters relevant to the transfection procedure. We then monitored the effect of different N/P ratios on transfection efficiency and cytotoxicity using flow cytometry and fluorescent microscopy. By the results, we found that transfection efficiency was greatly affected by plasmid DNA concentration, PEI concentration, order of combining reagents, serum presence in polyplexes, and the duration of serum starvations. Moreover, using the optimized condition, we found that the N/P ratio of 3 achieved the highest percentage of HDFs positive for green fluorescent protein plasmid (∼40%) with minimal cell toxicity. We finally generated hiPSCs using the optimized protocol and oriP/EBNA1-based vectors. We confirmed hiPSC formation by characterizing tests: alkaline phosphatase staining, immunocytochemistry assay, real-time PCR analysis, in vitro differentiation into three germ layers, and karyotyping test. In conclusion, our results indicated that 25 kDa branched PEI could efficiently transfect HDFs toward generating hiPSCs via a simple, cost-effective, and optimized condition.
Collapse
Affiliation(s)
- Monir Shayestehfar
- Department of Neuroscience, Faculty of Advanced Technology in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sara Farahi
- Biotechnology Department, Shahid Beheshti University of medical science, Tehran, Iran
| | - Behjat Kheiri Yeganeh Azar
- Department of Molecular Medicine, Faculty of Advanced Technology in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amirhossein Memari
- Sports Medicine Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Faezeh Faghihi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
KIZILYER A. An optimized protocol for the electroporation of NCI H929 multiple myeloma cells. MEHMET AKIF ERSOY ÜNIVERSITESI VETERINER FAKÜLTESI DERGISI 2022. [DOI: 10.24880/maeuvfd.1126466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Multiple myeloma cell lines are difficult to transfect with non-viral nucleic acid delivery methods. While electroporation is the most efficient tool for the transfection of most hard-to-transfect cells, human multiple myeloma cells differ in their permissiveness and each cell type require different electroporation conditions for an efficient transgene delivery. In this study, various parameters for NCI H929 human multiple myeloma cells are tested to generate an optimized electroporation protocol. Findings from this paper showed that besides the voltage and capacitance settings, cell count, the cell cycle status of cells, the amount of nucleic acid and removal of death cells all impacted the electroporation efficiency and viable cell count. These results are expected to serve as a starting point and a guide for researchers.
Collapse
|
11
|
Singh S, Dahiya S, Singh YJ, Beeton K, Jain A, Sarkar R, Dubey A, Tehseen A, Sehrawat S. Robust anti-SARS-CoV2 single domain antibodies cross neutralize multiple viruses. iScience 2022; 25:104549. [PMID: 35702569 PMCID: PMC9181070 DOI: 10.1016/j.isci.2022.104549] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/08/2022] [Accepted: 06/02/2022] [Indexed: 11/24/2022] Open
Abstract
We report robust SARS-CoV2 neutralizing sdAbs targeting the viral peptides encompassing the polybasic cleavage site (CSP) and in the receptor binding domain (RBD) of the spike (S) protein. Both the sdAbs inhibited infectivity of the CoV2 S protein expressing pseudoviruses (LV-CoV2S). Both anti-CSP and RBD intrabodies (IB) inhibited the output of LV(CoV2 S). Anti-CSP IB altered the proteolytic processing and targeted the viral S protein for degradation. Because of cross-reactive CSPs in the entry mediators, the anti-CSP sdAb neutralized in vitro and in vivo the infectivity of SARS-CoV2 unrelated viruses such as herpes simplex virus 1 (HSV1) and pestes des petits ruminants virus (PPRV). Conversely, anti-HSV1 and anti-PPRV sera neutralized LV(CoV2 S) owing to the presence of CSP reactive antibodies indicating that a prior infection with such pathogens could impact on the pattern of COVID-19.
Collapse
Affiliation(s)
- Sudhakar Singh
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar Knowledge City PO Manauli, Mohali, Punjab 140306, India
| | - Surbhi Dahiya
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar Knowledge City PO Manauli, Mohali, Punjab 140306, India
| | - Yuviana J. Singh
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar Knowledge City PO Manauli, Mohali, Punjab 140306, India
| | - Komal Beeton
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar Knowledge City PO Manauli, Mohali, Punjab 140306, India
| | - Ayush Jain
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar Knowledge City PO Manauli, Mohali, Punjab 140306, India
| | - Roman Sarkar
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar Knowledge City PO Manauli, Mohali, Punjab 140306, India
| | - Abhishek Dubey
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar Knowledge City PO Manauli, Mohali, Punjab 140306, India
| | - Azeez Tehseen
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar Knowledge City PO Manauli, Mohali, Punjab 140306, India
| | - Sharvan Sehrawat
- Department of Biological Sciences, Indian Institute of Science Education and Research Mohali, Sector 81, SAS Nagar Knowledge City PO Manauli, Mohali, Punjab 140306, India
| |
Collapse
|
12
|
Dabbaghi M, Hashemi K, Oskuee RK, Afkhami-Goli A. Reverse relation between cytotoxicity and Polyethylenimine/DNA ratio, the effect of using HEPES-buffered saline (HBS) medium in gene delivery. Toxicol In Vitro 2022; 83:105414. [DOI: 10.1016/j.tiv.2022.105414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 05/17/2022] [Accepted: 06/03/2022] [Indexed: 11/25/2022]
|
13
|
Liu F, Liu J, Xiang H, Sun Z, Li Y, Li X, Liu Y, Liu J. Dihydroartemisinin protects blood-brain barrier permeability during sepsis by inhibiting the transcription factor SNAI1. Clin Exp Pharmacol Physiol 2022; 49:979-987. [PMID: 35651290 PMCID: PMC9543489 DOI: 10.1111/1440-1681.13683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 04/07/2022] [Accepted: 05/30/2022] [Indexed: 11/28/2022]
Abstract
Blood–brain barrier (BBB) injury is involved in the pathogenesis of sepsis‐associated encephalopathy. In this study, we used dihydroartemisinin (DHA), a derivative of artemisinin, to treat a cecal ligation and puncture (CLP)‐induced mouse sepsis model and a tumour necrosis factor α (TNF‐α)‐stimulated human cerebral microvessel endothelial cells (hCMEC)/D3 cell line. We found that DHA decreased BBB permeability and increased the expression of the tight junction protein occludin (OCLN) in the CLP model. In hCMEC/D3 cells, DHA decreased TNF‐α‐induced hyperpermeability and increased the expression of OCLN. DHA also repressed SNAI1 expression in the CLP mouse model and in TNF‐α‐stimulated hCMEC/D3 cells. These data suggest that DHA protects BBB permeability during sepsis by stimulating the expression of OCLN, by downregulating the expression of the SNAI1 transcription factor.
Collapse
Affiliation(s)
- Fuhong Liu
- School of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.,Medical Research Center, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, 16766 Jingshi Road, Jinan, Shandong, China
| | - Jing Liu
- Medical Research Center, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, 16766 Jingshi Road, Jinan, Shandong, China
| | - Hongjie Xiang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, 16766 Jingshi Road, Jinan, Shandong, China
| | - Zongguo Sun
- Medical Research Center, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, 16766 Jingshi Road, Jinan, Shandong, China
| | - Yan Li
- School of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiao Li
- School of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yanjun Liu
- School of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ju Liu
- Medical Research Center, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, 16766 Jingshi Road, Jinan, Shandong, China
| |
Collapse
|
14
|
de Campos NSP, de Oliveira Beserra A, Pereira PHB, Chaves AS, Fonseca FLA, da Silva Medina T, dos Santos TG, Wang Y, Marasco WA, Suarez ER. Immune Checkpoint Blockade via PD-L1 Potentiates More CD28-Based than 4-1BB-Based Anti-Carbonic Anhydrase IX Chimeric Antigen Receptor T Cells. Int J Mol Sci 2022; 23:ijms23105448. [PMID: 35628256 PMCID: PMC9141239 DOI: 10.3390/ijms23105448] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 02/01/2023] Open
Abstract
The complete regression of clear cell renal cell carcinoma (ccRCC) obtained pre-clinically with anti-carbonic anhydrase IX (CAIX) G36 chimeric antigen receptor (CAR) T cells in doses equivalent to ≅108 CAR T cells/kg renewed the potential of this target to treat ccRCC and other tumors in hypoxia. The immune checkpoint blockade (ICB) brought durable clinical responses in advanced ccRCC and other tumors. Here, we tested CD8α/4-1BB compared to CD28-based anti-CAIX CAR peripheral blood mononuclear cells (PBMCs) releasing anti-programmed cell death ligand-1 (PD-L1) IgG4 for human ccRCC treatment in vitro and in an orthotopic NSG mice model in vivo. Using a ≅107 CAR PBMCs cells/kg dose, anti-CAIX CD28 CAR T cells releasing anti-PD-L1 IgG highly decrease both tumor volume and weight in vivo, avoiding the occurrence of metastasis. This antitumoral superiority of CD28-based CAR PBMCs cells compared to 4-1BB occurred under ICB via PD-L1. Furthermore, the T cell exhaustion status in peripheral CD4 T cells, additionally to CD8, was critical for CAR T cells efficiency. The lack of hepatotoxicity and nephrotoxicity upon the administration of a 107 CAR PMBCs cells/kg dose is the basis for carrying out clinical trials using anti-CAIX CD28 CAR PBMCs cells releasing anti-PD-L1 antibodies or anti-CAIX 4-1BB CAR T cells, offering exciting new prospects for the treatment of refractory ccRCC and hypoxic tumors.
Collapse
Affiliation(s)
| | - Adriano de Oliveira Beserra
- A.C. Camargo Cancer Center, Centro Internacional de Pesquisa, Sao Paulo 01508-010, SP, Brazil; (A.d.O.B.); (P.H.B.P.); (A.S.C.); (T.d.S.M.); (T.G.d.S.)
| | - Pedro Henrique Barbosa Pereira
- A.C. Camargo Cancer Center, Centro Internacional de Pesquisa, Sao Paulo 01508-010, SP, Brazil; (A.d.O.B.); (P.H.B.P.); (A.S.C.); (T.d.S.M.); (T.G.d.S.)
| | - Alexandre Silva Chaves
- A.C. Camargo Cancer Center, Centro Internacional de Pesquisa, Sao Paulo 01508-010, SP, Brazil; (A.d.O.B.); (P.H.B.P.); (A.S.C.); (T.d.S.M.); (T.G.d.S.)
| | - Fernando Luiz Affonso Fonseca
- Laboratório de Análises Clínicas, Centro Universitário Faculdade de Medicina do ABC, Santo Andre 09060-870, SP, Brazil;
- Departamento de Ciências Farmacêuticas, Universidade Federal de Aso Paulo, Diadema 09920-000, SP, Brazil
| | - Tiago da Silva Medina
- A.C. Camargo Cancer Center, Centro Internacional de Pesquisa, Sao Paulo 01508-010, SP, Brazil; (A.d.O.B.); (P.H.B.P.); (A.S.C.); (T.d.S.M.); (T.G.d.S.)
| | - Tiago Goss dos Santos
- A.C. Camargo Cancer Center, Centro Internacional de Pesquisa, Sao Paulo 01508-010, SP, Brazil; (A.d.O.B.); (P.H.B.P.); (A.S.C.); (T.d.S.M.); (T.G.d.S.)
| | - Yufei Wang
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA;
- Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Wayne Anthony Marasco
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02215, USA;
- Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
- Correspondence: (W.A.M.); (E.R.S.)
| | - Eloah Rabello Suarez
- Center for Natural and Human Sciences, Federal University of ABC, Santo Andre 09210-580, SP, Brazil;
- Correspondence: (W.A.M.); (E.R.S.)
| |
Collapse
|
15
|
Guzmán-Silva A, Martínez-Morales JC, Medina LDC, Romero-Ávila MT, Villegas-Comonfort S, Solís KH, García-Sáinz JA. Mutation of putative phosphorylation sites in the free fatty acid receptor 1: Effects on signaling, receptor phosphorylation, and internalization. Mol Cell Endocrinol 2022; 545:111573. [PMID: 35065200 DOI: 10.1016/j.mce.2022.111573] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 01/07/2022] [Accepted: 01/17/2022] [Indexed: 11/16/2022]
Abstract
Free fatty acid receptor 1 phosphorylation sites were studied using mutants, including a) a mutant with T215V in the third intracellular loop (3IL), b) another with changes in the carboxyl terminus (C-term): T287V, T293V, S298A, and c) a mutant with all of these changes (3IL/C-term). Agonist-induced increases in intracellular calcium were similar between cells expressing wild-type or mutant receptors. In contrast, agonist-induced FFA1 receptor phosphorylation was reduced in mutants compared to wild type. Phorbol ester-induced FFA1 receptor phosphorylation was rapid and robust in cells expressing the wild-type receptor and essentially abolished in the mutants. Agonist-induced ERK 1/2 phosphorylation and receptor internalization were decreased in cells expressing the mutant receptors compared to those expressing the wild-type receptor. Our data suggest that the identified sites might participate in receptor phosphorylation, signaling, and internalization.
Collapse
Affiliation(s)
- Alejandro Guzmán-Silva
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito exterior s/n, Ciudad Universitaria, Ap. Postal 70-600, Ciudad de México, 04510, Mexico
| | - Juan Carlos Martínez-Morales
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito exterior s/n, Ciudad Universitaria, Ap. Postal 70-600, Ciudad de México, 04510, Mexico
| | - Luz Del Carmen Medina
- Departamento de Biología de la Reproducción, División de CBS, Universidad Autónoma Metropolitana-Iztapalapa, Av. San Rafael Atlixco 186, Ciudad de México, 09340, Mexico
| | - M Teresa Romero-Ávila
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito exterior s/n, Ciudad Universitaria, Ap. Postal 70-600, Ciudad de México, 04510, Mexico
| | - Sócrates Villegas-Comonfort
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito exterior s/n, Ciudad Universitaria, Ap. Postal 70-600, Ciudad de México, 04510, Mexico
| | - Karina Helivier Solís
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito exterior s/n, Ciudad Universitaria, Ap. Postal 70-600, Ciudad de México, 04510, Mexico
| | - J Adolfo García-Sáinz
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Circuito exterior s/n, Ciudad Universitaria, Ap. Postal 70-600, Ciudad de México, 04510, Mexico.
| |
Collapse
|
16
|
Feng S, Wang Z, Li A, Xie X, Liu J, Li S, Li Y, Wang B, Hu L, Yang L, Guo T. Strategies for High-Efficiency Mutation Using the CRISPR/Cas System. Front Cell Dev Biol 2022; 9:803252. [PMID: 35198566 PMCID: PMC8860194 DOI: 10.3389/fcell.2021.803252] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 12/22/2021] [Indexed: 12/15/2022] Open
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPR)-associated systems have revolutionized traditional gene-editing tools and are a significant tool for ameliorating gene defects. Characterized by high target specificity, extraordinary efficiency, and cost-effectiveness, CRISPR/Cas systems have displayed tremendous potential for genetic manipulation in almost any organism and cell type. Despite their numerous advantages, however, CRISPR/Cas systems have some inherent limitations, such as off-target effects, unsatisfactory efficiency of delivery, and unwanted adverse effects, thereby resulting in a desire to explore approaches to address these issues. Strategies for improving the efficiency of CRISPR/Cas-induced mutations, such as reducing off-target effects, improving the design and modification of sgRNA, optimizing the editing time and the temperature, choice of delivery system, and enrichment of sgRNA, are comprehensively described in this review. Additionally, several newly emerging approaches, including the use of Cas variants, anti-CRISPR proteins, and mutant enrichment, are discussed in detail. Furthermore, the authors provide a deep analysis of the current challenges in the utilization of CRISPR/Cas systems and the future applications of CRISPR/Cas systems in various scenarios. This review not only serves as a reference for improving the maturity of CRISPR/Cas systems but also supplies practical guidance for expanding the applicability of this technology.
Collapse
Affiliation(s)
- Shuying Feng
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Zilong Wang
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Aifang Li
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Xin Xie
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Junjie Liu
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Shuxuan Li
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yalan Li
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Baiyan Wang
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Lina Hu
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Lianhe Yang
- Medical College, Henan University of Chinese Medicine, Zhengzhou, China
| | - Tao Guo
- Department of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, China
| |
Collapse
|
17
|
Adaptor protein 3BP2 regulates dectin-1-mediated cellular signalling to induce cytokine expression and NF-κB activation. Biochem J 2022; 479:503-523. [PMID: 35129602 DOI: 10.1042/bcj20210707] [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: 10/01/2021] [Revised: 01/31/2022] [Accepted: 02/07/2022] [Indexed: 11/17/2022]
Abstract
The adaptor protein c-Abl Src homology 3 domain-binding protein-2 (3BP2) is phosphorylated by spleen tyrosine kinase (Syk), and the phosphorylation of Tyr183 is important in the regulation of immune responses. Recently, we reported that 3BP2 plays important roles in phagocytosis and chemokine expression mediated by the Fc receptor for IgG. Although it is well established that various phagocytic cells express Syk-coupled C-type lectin receptors (CLRs) to induce innate immune responses, the functions of 3BP2 and the physiological relevance of the phosphorylation of Tyr183 remain elusive. In this study, we generated genome-edited mice and observed that 3BP2 influenced the development of bone marrow-derived dendritic cells (BMDCs) induced by granulocyte-macrophage colony-stimulating factor. In addition, we found that 3BP2 was critical for cytokine expression induced by Syk-coupled CLRs-dectin-1 and macrophage-inducible C-type lectin. Immunoblotting analyses revealed that 3BP2 was required for the dectin-1-induced activation of NF-κB p65. The impaired expression of cytokines and activation of NF-κB in 3BP2-mutant cells were restored by wild-type 3BP2, suggesting that 3BP2 was involved in the dectin-1-mediated signalling that led to NF-κB activation. Furthermore, we found that the phosphorylation of Tyr183 is not essential for cytokine expression and that 3BP2 in combination with caspase recruitment domain family member 9 activates NF-κB in HEK-293T cells. Collectively, these results indicate that in addition to the development of BMDCs, 3BP2 plays an important role in the dectin-1-induced activation of NF-κB and cytokine expression.
Collapse
|
18
|
Jiang Q, Guan S, Zhang Y, Sun Y, Jiang X. Targeted and fluorescence traceable multifunctional host-guest supramolecular gene delivery platform based on poly(cyclodextrin) and rhodamine conjugated disulfide-containing azobenzene-terminated branched polymer. INT J POLYM MATER PO 2022. [DOI: 10.1080/00914037.2022.2029438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Qimin Jiang
- Jiangsu Key Laboratory of Environmentally Friendly Polymeric Materials, School of Materials Science and Engineering, Jiangsu Collaborative Innovation Center of Photovolatic Science and Engineering, Changzhou University, Changzhou, P. R. China
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, P. R. China
| | - Shuyi Guan
- Jiangsu Key Laboratory of Environmentally Friendly Polymeric Materials, School of Materials Science and Engineering, Jiangsu Collaborative Innovation Center of Photovolatic Science and Engineering, Changzhou University, Changzhou, P. R. China
| | - Yunti Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, P. R. China
| | - Yuhua Sun
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, P. R. China
| | - Xulin Jiang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, P. R. China
| |
Collapse
|
19
|
Tortajada L, Felip C, Vicent MJ. Polymer-based Non-viral Vectors for Gene Therapy in the Skin. Polym Chem 2022. [DOI: 10.1039/d1py01485d] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Gene therapy has emerged as a versatile technique with the potential to treat a range of human diseases; however, examples of the topical application of gene therapy as a treatment...
Collapse
|
20
|
Tang QL, Gu LX, Xu Y, Liao XH, Zhou Y, Zhang TC. Establishing functional lentiviral vector production in a stirred bioreactor for CAR-T cell therapy. Bioengineered 2021; 12:2095-2105. [PMID: 34047682 PMCID: PMC8806440 DOI: 10.1080/21655979.2021.1931644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 11/01/2022] Open
Abstract
As gene delivery tools, lentiviral vectors (LV) have broad applications in chimeric antigen receptor therapy (CAR-T). Large-scale production of functional LV is limited by the adherent, serum-dependent nature of HEK293T cells used in the manufacturing. HEK293T adherent cells were adapted to suspension cells in a serum-free medium to establish large-scale processes for functional LV production in a stirred bioreactor without micro-carriers. The results showed that 293 T suspension was successfully cultivated in F media (293 CD05 medium and SMM293-TII with 1:1 volume ratio), and the cells retained the capacity for LV production. After cultivation in a 5.5 L bioreactor for 4 days, the cells produced 1.5 ± 0.3 × 107 TU/mL raw LV, and the lentiviral transduction efficiency was 48.6 ± 2.8% in T Cells. The yield of LV equaled to the previous shake flask. The critical process steps were completed to enable a large-scale LV production process. Besides, a cryopreservation solution was developed to reduce protein involvement, avoid cell grafting and reduce process cost. The process is cost-effective and easy to scale up production, which is expected to be highly competitive.
Collapse
Affiliation(s)
- Qu-Lai Tang
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education & Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| | - Li-Xing Gu
- College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Yao Xu
- College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Xing-Hua Liao
- College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Yong Zhou
- College of Life Science and Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Tong-Cun Zhang
- Key Laboratory of Industrial Fermentation Microbiology of the Ministry of Education & Tianjin Key Laboratory of Industrial Microbiology, College of Biotechnology, Tianjin University of Science and Technology, Tianjin, China
| |
Collapse
|
21
|
Mancinelli S, Turcato A, Kisslinger A, Bongiovanni A, Zazzu V, Lanati A, Liguori GL. Design of transfections: Implementation of design of experiments for cell transfection fine tuning. Biotechnol Bioeng 2021; 118:4488-4502. [PMID: 34406655 PMCID: PMC9291525 DOI: 10.1002/bit.27918] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 08/02/2021] [Accepted: 08/12/2021] [Indexed: 11/25/2022]
Abstract
Transfection is the process by which nucleic acids are introduced into eukaryotic cells. This is fundamental in basic research for studying gene function and modulation of gene expression as well as for many bioprocesses in the manufacturing of clinical‐grade recombinant biologics from cells. Transfection efficiency is a critical parameter to increase biologics' productivity; the right protocol has to be identified to ensure high transfection efficiency and therefore high product yield. Design of experiments (DoE) is a mathematical method that has become a key tool in bioprocess development. Based on the DoE method, we developed an operational flow that we called “Design of Transfections” (DoT) for specific transfection modeling and identification of the optimal transfection conditions. As a proof of principle, we applied the DoT workflow to optimize a cell transfection chemical protocol for neural progenitors, using polyethyleneimine (PEI). We simultaneously varied key influencing factors, namely concentration and type of PEI, DNA concentration, and cell density. The transfection efficiency was measured by fluorescence imaging followed by automatic counting of the green fluorescent transfected cells. Taking advantage of the DoT workflow, we developed a new simple, efficient, and economically advantageous PEI transfection protocol through which we were able to obtain a transfection efficiency of 34%.
Collapse
Affiliation(s)
- Sara Mancinelli
- Institute of Genetics and Biophysics (IGB), National Research Council (CNR), Naples, Italy
| | | | - Annamaria Kisslinger
- Institute of Experimental Endocrinology and Oncology (IEOS), National Research Council (CNR), Naples, Italy
| | - Antonella Bongiovanni
- Institute for Research and Biomedical Innovation (IRIB), National Research Council (CNR), Palermo, Italy
| | - Valeria Zazzu
- Institute of Genetics and Biophysics (IGB), National Research Council (CNR), Naples, Italy
| | | | - Giovanna Lucia Liguori
- Institute of Genetics and Biophysics (IGB), National Research Council (CNR), Naples, Italy
| |
Collapse
|
22
|
Cousin N, Cap S, Dihr M, Tacconi C, Detmar M, Dieterich LC. Lymphatic PD-L1 Expression Restricts Tumor-Specific CD8 + T-cell Responses. Cancer Res 2021; 81:4133-4144. [PMID: 34099493 PMCID: PMC9398148 DOI: 10.1158/0008-5472.can-21-0633] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/23/2021] [Accepted: 06/01/2021] [Indexed: 01/07/2023]
Abstract
Lymph node (LN)-resident lymphatic endothelial cells (LEC) mediate peripheral tolerance by self-antigen presentation on MHC-I and constitutive expression of T-cell inhibitory molecules, including PD-L1 (CD274). Tumor-associated LECs also upregulate PD-L1, but the specific role of lymphatic PD-L1 in tumor immunity is not well understood. In this study, we generated a mouse model lacking lymphatic PD-L1 expression and challenged these mice with two orthotopic tumor models, B16F10 melanoma and MC38 colorectal carcinoma. Lymphatic PD-L1 deficiency resulted in consistent expansion of tumor-specific CD8+ T cells in tumor-draining LNs in both tumor models, reduced primary tumor growth in the MC38 model, and increased efficacy of adoptive T-cell therapy in the B16F10 model. Strikingly, lymphatic PD-L1 acted primarily by inducing apoptosis in tumor-specific CD8+ central memory T cells. Overall, these findings demonstrate that LECs restrain tumor-specific immunity via PD-L1, which may explain why some patients with cancer without PD-L1 expression in the tumor microenvironment still respond to PD-L1/PD-1-targeted immunotherapy. SIGNIFICANCE: A new lymphatic-specific PD-L1 knockout mouse model reveals that lymphatic endothelial PD-L1 expression reduces tumor immunity, inducing apoptosis in tumor-specific CD8+ central memory cells in tumor-draining lymph nodes.
Collapse
Affiliation(s)
| | | | | | | | | | - Lothar C. Dieterich
- Corresponding Author: Lothar C. Dieterich, ETH Zurich, Institute of Pharmaceutical Sciences, Vladimir-Prelog-Weg 1–5/10, 8093 Zurich, Switzerland. Phone: 41-44-63-37392; Fax: 41-44-63-31344; E-mail:
| |
Collapse
|
23
|
Patiño MI, Restrepo LM, Becerra NY, van der Mei HC, van Kooten TG, Sharma PK. Nonviral Expression of LL-37 in a Human Skin Equivalent to Prevent Infection in Skin Wounds. Hum Gene Ther 2021; 32:1147-1157. [PMID: 33980038 DOI: 10.1089/hum.2021.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Inefficient autologous tissue recovery in skin wounds increases the susceptibility of patients to infections caused by multidrug resistant microorganisms, resulting in a high mortality rate. Genetic modification of skin cells has become an important field of study because it could lead to the construction of more functional skin grafts, through the overexpression of antimicrobial peptides that would prevent early contamination and infection with bacteria. In this study, we produce and evaluate human skin equivalents (HSEs) containing transfected human primary fibroblasts and keratinocytes by polyplexes to express the antimicrobial peptide LL-37. The effect of LL-37 on the metabolic activity of normal HSEs was evaluated before the construction of the transfected HSEs, and the antimicrobial efficacy against Pseudomonas aeruginosa and Staphylococcus aureus was evaluated. Subsequently, the levels of LL-37 in the culture supernatants of transfected HSEs, as well as the local expression, were determined. It was found that LL-37 treatment significantly promoted the cellular proliferation of HSEs. Furthermore, HSEs that express elevated levels of LL-37 were shown to possess histological characteristics close to the normal skin and display enhanced antimicrobial activity against S. aureus in vitro. These findings demonstrate that HSEs expressing LL-37 through nonviral modification of skin cells are a promising approach for the prevention of bacterial colonization in wounds.
Collapse
Affiliation(s)
- Maria Isabel Patiño
- Tissue Engineering and Cell Therapy Group, Faculty of Medicine, University of Antioquia, Medellín, Colombia.,Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Luz Marina Restrepo
- Tissue Engineering and Cell Therapy Group, Faculty of Medicine, University of Antioquia, Medellín, Colombia
| | - Natalia Yiset Becerra
- Tissue Engineering and Cell Therapy Group, Faculty of Medicine, University of Antioquia, Medellín, Colombia
| | - Henny C van der Mei
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Theo G van Kooten
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Prashant K Sharma
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
24
|
Therapeutic delivery of siRNA with polymeric carriers to down-regulate STAT5A expression in high-risk B-cell acute lymphoblastic leukemia (B-ALL). PLoS One 2021; 16:e0251719. [PMID: 34157051 PMCID: PMC8219370 DOI: 10.1371/journal.pone.0251719] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 05/02/2021] [Indexed: 11/19/2022] Open
Abstract
Overexpression and persistent activation of STAT5 play an important role in the development and progression of acute lymphoblastic leukemia (ALL), the most common pediatric cancer. Small interfering RNA (siRNA)-mediated downregulation of STAT5 represents a promising therapeutic approach for ALL to overcome the limitations of current treatment modalities such as high relapse rates and poor prognosis. However, to effectively transport siRNA molecules to target cells, development of potent carriers is of utmost importance to surpass hurdles of delivery. In this study, we investigated the use of lipopolymers as non-viral delivery systems derived from low molecular weight polyethylenimines (PEI) substituted with lauric acid (Lau), linoleic acid (LA) and stearic acid (StA) to deliver siRNA molecules to ALL cell lines and primary samples. Among the lipid-substituted polymers explored, Lau- and LA-substituted PEI displayed excellent siRNA delivery to SUP-B15 and RS4;11 cells. STAT5A gene expression was downregulated (36-92%) in SUP-B15 and (32%) in RS4;11 cells using the polymeric delivery systems, which consequently reduced cell growth and inhibited the formation of colonies in ALL cells. With regard to ALL primary cells, siRNA-mediated STAT5A gene silencing was observed in four of eight patient cells using our leading polymeric delivery system, 1.2PEI-Lau8, accompanied by the significant reduction in colony formation in three of eight patients. In both BCR-ABL positive and negative groups, three of five patients demonstrated marked cell growth inhibition in both MTT and trypan blue exclusion assays using 1.2PEI-Lau8/siRNA complexes in comparison with their control siRNA groups. Three patient samples did not show any positive results with our delivery systems. Differential therapeutic responses to siRNA therapy observed in different patients could result from variable genetic profiles and patient-to-patient variability in delivery. This study supports the potential of siRNA therapy and the designed lipopolymers as a delivery system in ALL therapy.
Collapse
|
25
|
Kucharski M, Mrowiec P, Ocłoń E. Current standards and pitfalls associated with the transfection of primary fibroblast cells. Biotechnol Prog 2021; 37:e3152. [PMID: 33774920 DOI: 10.1002/btpr.3152] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/24/2021] [Accepted: 03/24/2021] [Indexed: 12/25/2022]
Abstract
Cultured fibroblast cells, especially dermal cells, are used for various types of scientific research, particularly within the medical field. Desirable features of the cells include their ease of isolation, rapid cellular growth, and high degree of robustness. Currently, fibroblasts are mainly used to obtain pluripotent cells via a reprogramming process. Dermal fibroblasts, are particularly useful for gene therapies used for promoting wound healing or minimizing skin aging. In recent years, fibroblast transfection efficiencies have significantly improved. In order to introduce molecules (most often DNA or RNA) into cells, viral-based systems (transduction) or non-viral methods (transfection) that include physical/mechanical processes or lipid reagents may be used. In this article, we describe critical points that should be considered when selecting a method for transfecting fibroblasts. The most effective methods used for the transfection of fibroblasts include both viral-based and non-viral nucleofection systems. These methods result in a high level of transgene expression and are superior in terms of transfection efficacy and viability.
Collapse
Affiliation(s)
- Mirosław Kucharski
- Department of Animal Physiology and Endocrinology, University of Agriculture in Krakow, Krakow, Poland
| | - Patrycja Mrowiec
- Department of Animal Reproduction, Anatomy and Genomics, University of Agriculture in Krakow, Krakow, Poland
| | - Ewa Ocłoń
- Centre for Experimental and Innovative Medicine, Laboratory of Recombinant Proteins Production, University of Agriculture in Krakow, Krakow, Poland
| |
Collapse
|
26
|
Flores-Espinoza E, Meizoso-Huesca A, Villegas-Comonfort S, Reyes-Cruz G, García-Sáinz JA. Effect of docosahexaenoic acid, phorbol myristate acetate, and insulin on the interaction of the FFA4 (short isoform) receptor with Rab proteins. Eur J Pharmacol 2020; 889:173595. [PMID: 32986985 DOI: 10.1016/j.ejphar.2020.173595] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/12/2020] [Accepted: 09/22/2020] [Indexed: 12/29/2022]
Abstract
Human embryonic kidney (HEK) 293 cells were co-transfected with plasmids for the expression of mCherry fluorescent protein-tagged FFA4 receptors and the enhanced green fluorescent protein-tagged Rab proteins involved in retrograde transport and recycling, to study their possible interaction through Förster Resonance Energy Transfer (FRET), under the action of agents that induce FFA4 receptor phosphorylation and internalization through different processes, i.e., the agonist, docosahexaenoic acid, the protein kinase C activator phorbol myristate acetate, and insulin. Data indicate that FFA4 receptor internalization varied depending on the agent that induced the process. Agonist activation (docosahexaenoic acid) induced an association with early endosomes (as suggested by interaction with Rab5) and rapid recycling to the plasma membrane (as indicated by receptor interaction with Rab4). More prolonged agonist stimulation also appears to allow the FFA4 receptors to interact with late endosomes (interaction with Rab9), slow recycling (interaction with Rab 11), and target to degradation (Rab7). Phorbol myristate acetate, triggered a rapid association with early endosomes (Rab5), slow recycling to the plasma membrane (Rab11), and some receptor degradation (Rab7). Insulin-induced FFA4 receptor internalization appears to be associated with interaction with early endosomes (Rab5) and late endosomes (Rab9) and fast and slow recycling to the plasma membrane (Rab4, Rab11). Additionally, we observed that agonist- and PMA-induced FFA4 internalization was markedly reduced by paroxetine, which suggests a possible role of G protein-coupled receptor kinase 2.
Collapse
Affiliation(s)
- Emmanuel Flores-Espinoza
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Aldo Meizoso-Huesca
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Sócrates Villegas-Comonfort
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Guadalupe Reyes-Cruz
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional-CINVESTAV, Av. Instituto Politécnico Nacional, 2508, Col. San Pedro Zacatenco, Mexico City, Mexico
| | - J Adolfo García-Sáinz
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico.
| |
Collapse
|
27
|
Najafov A, Luu HS, Mookhtiar AK, Mifflin L, Xia HG, Amin PP, Ordureau A, Wang H, Yuan J. RIPK1 Promotes Energy Sensing by the mTORC1 Pathway. Mol Cell 2020; 81:370-385.e7. [PMID: 33271062 DOI: 10.1016/j.molcel.2020.11.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 08/31/2020] [Accepted: 11/04/2020] [Indexed: 02/03/2023]
Abstract
The mechanisms of cellular energy sensing and AMPK-mediated mTORC1 inhibition are not fully delineated. Here, we discover that RIPK1 promotes mTORC1 inhibition during energetic stress. RIPK1 is involved in mediating the interaction between AMPK and TSC2 and facilitate TSC2 phosphorylation at Ser1387. RIPK1 loss results in a high basal mTORC1 activity that drives defective lysosomes in cells and mice, leading to accumulation of RIPK3 and CASP8 and sensitization to cell death. RIPK1-deficient cells are unable to cope with energetic stress and are vulnerable to low glucose levels and metformin. Inhibition of mTORC1 rescues the lysosomal defects and vulnerability to energetic stress and prolongs the survival of RIPK1-deficient neonatal mice. Thus, RIPK1 plays an important role in the cellular response to low energy levels and mediates AMPK-mTORC1 signaling. These findings shed light on the regulation of mTORC1 during energetic stress and unveil a point of crosstalk between pro-survival and pro-death pathways.
Collapse
Affiliation(s)
- Ayaz Najafov
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Ludwig Center, Harvard Medical School, Boston, MA 02115, USA.
| | - Hoang Son Luu
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Adnan K Mookhtiar
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Lauren Mifflin
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Hong-Guang Xia
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Palak P Amin
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Alban Ordureau
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Huibing Wang
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Junying Yuan
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Ludwig Center, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
28
|
Wang Y, Chan LLY, Grimaud M, Fayed A, Zhu Q, Marasco WA. High-Throughput Image Cytometry Detection Method for CAR-T Transduction, Cell Proliferation, and Cytotoxicity Assays. Cytometry A 2020; 99:689-697. [PMID: 33191639 DOI: 10.1002/cyto.a.24267] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/14/2020] [Accepted: 11/11/2020] [Indexed: 12/27/2022]
Abstract
Chimeric antigen receptor (CAR)-T cell therapy has drawn much attention due to its recent clinical success in B-cell malignancies. In general, the CAR-T cell discovery process consists of CAR identification, T-cell activation, transduction, and expansion, as well as assessment of CAR-T cytotoxicity. The current evaluation methods for the CAR-T discovery process can be time-consuming, low-throughput and requires the preparation of multiple sacrificial samples in order to produce kinetic data. In this study, we employed the use of a plate-based image cytometer to monitor anti-CAIX (carbonic anhydrase IX) G36 CAR-T generation and assess its cytotoxic potency of direct and selective killing against CAIX+ SKRC-59 human renal cell carcinoma cells. The transduction efficiency and cytotoxicity results were analyzed using image cytometry and compared directly to flow cytometry and Chromium 51 (51 Cr) release assays, showing that image cytometry was comparable against these conventional methods. Image cytometry method streamlines the assays required during the CAR-T cell discovery process by analyzing a plate of T cells from CAR-T generation to in vitro functional assays with minimum disruption. The proposed method can reduce assay time and uses less cell samples by imaging and analyze the same plate over time without the need to sacrifice any cells. The ability to monitor kinetic data can allow additional insights into the behavior and interaction between CAR-T and target tumor cells. © 2020 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Yufei Wang
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, 02215, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Leo Li-Ying Chan
- Department of Advanced Technology R&D, Nexcelom Bioscience LLC., Lawrence, Massachusetts, 01843, USA
| | - Marion Grimaud
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, 02215, USA
| | - Atef Fayed
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, 02215, USA
| | - Quan Zhu
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, 02215, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Wayne A Marasco
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, 02215, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
29
|
de-Los-Santos-Cocotle G, Martínez-Morales JC, Romero-Ávila MT, Reyes-Cruz G, García-Sáinz JA. Effects of agonists and phorbol esters on α 1A-adrenergic receptor-Rab protein interactions. Eur J Pharmacol 2020; 885:173423. [PMID: 32750368 DOI: 10.1016/j.ejphar.2020.173423] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 07/21/2020] [Accepted: 07/25/2020] [Indexed: 11/29/2022]
Abstract
In a cell line, stably expressing α1A-adrenoceptors fused to the mCherry red fluorescent protein, noradrenaline, methoxamine, and oxymetazoline induced concentration-dependent increases in intracellular calcium. All of these agents increase α1A-adrenoceptor phosphorylation and internalization. Transient co-expression of these receptors with Rab proteins tagged with the enhanced Green Fluorescent Protein was employed to estimate α1A-adrenoceptor-Rab interaction using Förster Resonance Energy Transfer. Noradrenaline and methoxamine increased α1A-adrenoceptor interaction with Rab5 and Rab7 but did not modify it with Rab9. Oxymetazoline induced adrenoceptor interaction with Rab5 and Rab9 and only an insignificant increase in Rab7 signal. Phorbol myristate acetate increased α1A-adrenoceptor interaction with Rab5 and Rab9 but did not modify it with Rab7. The agonists and the active phorbol ester, all of which induce receptor phosphorylation and internalization, favor receptor interaction with Rab5, i.e., association with early endosomes. Cell stimulation with phorbol myristate acetate induced the α1A-adrenoceptors to interact with the late endosomal marker, Rab9, suggesting that the receptors are directed to slow recycling endosomes once they have transited to the Trans-Golgi network to be retrieved to the plasma membrane. The agonists noradrenaline and methoxamine likely induce a faster recycling and might direct some of the adrenoceptors toward degradation and/or very slow recycling to the plasma membrane. Oxymetazoline produced a mixed pattern of interaction with the Rab proteins. These data indicate that α1A-adrenoceptor agonists can trigger different vesicular traffic and receptor fates within the cells.
Collapse
Affiliation(s)
- Gustavo de-Los-Santos-Cocotle
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Juan Carlos Martínez-Morales
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - M Teresa Romero-Ávila
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Guadalupe Reyes-Cruz
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional-CINVESTAV, Av. Instituto Politécnico Nacional 2508; Col, San Pedro Zacatenco, Mexico City, Mexico
| | - J Adolfo García-Sáinz
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico.
| |
Collapse
|
30
|
Nanoparticle-siRNA: A potential strategy for rheumatoid arthritis therapy? J Control Release 2020; 325:380-393. [PMID: 32653501 DOI: 10.1016/j.jconrel.2020.07.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/05/2020] [Accepted: 07/07/2020] [Indexed: 02/07/2023]
Abstract
Rheumatoid arthritis (RA) is a common clinical inflammatory disease of the autoimmune system manifested by persistent synovitis, cartilage damage and even deformities. Despite significant progress in the clinical treatment of RA, long-term administration of anti-rheumatic drugs can cause a series of problems, including infections, gastrointestinal reactions, and abnormal liver and kidney functions. The emergence of RNA interference (RNAi) drugs has brought new hope for the treatment of RA. Designing a reasonable vector for RNAi drugs will greatly expand the application prospects of RNAi. Nanoparticles as a promising drug carrier provide reliable support for RNAi drugs. The review summarizes the pathogenesis of RA as a possible target for small interference RNA (siRNA) design. At the same time, the review also analyzes the nanoparticles used in siRNA carriers in recent years, laying the foundation and prospect for the next step in the development of intelligent nanocarriers.
Collapse
|
31
|
Elshereef AA, Jochums A, Lavrentieva A, Stuckenberg L, Scheper T, Solle D. High cell density transient transfection of CHO cells for TGF-β1 expression. Eng Life Sci 2020; 19:730-740. [PMID: 32624966 DOI: 10.1002/elsc.201800174] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 07/30/2019] [Accepted: 08/20/2019] [Indexed: 01/08/2023] Open
Abstract
High cell densities for transient transfection with polyethyleneimine (PEI) can be used for rapid and maximal production of recombinant proteins. High cell densities can be obtained by different cultivation systems, such as batch or perfusion systems. Herein, densities up to 18 million cells/mL were obtained by centrifugation for transfection evaluation. PEI transfection efficiency was easily determined by transfected enhanced green fluorescence protein (EGFP) reporter plasmid DNA (pDNA). A linear correlation between fluorescence intensity and transfection efficiency was improved. The transfection efficiency of PEI was highly dependent on the transfection conditions and directly related to the level of recombinant protein. Several factors were required to optimize the transient transfection process; these factors included the media type (which is compatible with low or high cell density transfection), the preculture CHO-K1 suspension cell density, and the pDNA to PEI level. Based on design of experiment (DoE) analyses, the optimal transfection conditions for 10 × 106 cells/mL in the CHOMACS CD medium achieved 73% transfection efficiency and a cell viability of over 80%. These results were confirmed for the production of transforming growth factor-beta 1 (TGF-β1) in a shake flask. The purified TGF-β1 protein concentration from 60 mL supernatant was 27 µg/mL, and the protein was biologically active.
Collapse
Affiliation(s)
- Abdalla A Elshereef
- Institute of Technical Chemistry Gottfried Wilhelm Leibniz University of Hannover Hannover Germany.,Chemistry of Natural and Microbial Products Department Pharmaceutical and Drug Industries Research Division National Research Centre Giza Egypt
| | - André Jochums
- Institute of Technical Chemistry Gottfried Wilhelm Leibniz University of Hannover Hannover Germany
| | - Antonina Lavrentieva
- Institute of Technical Chemistry Gottfried Wilhelm Leibniz University of Hannover Hannover Germany
| | - Lena Stuckenberg
- Institute of Technical Chemistry Gottfried Wilhelm Leibniz University of Hannover Hannover Germany
| | - Thomas Scheper
- Institute of Technical Chemistry Gottfried Wilhelm Leibniz University of Hannover Hannover Germany
| | - Dörte Solle
- Institute of Technical Chemistry Gottfried Wilhelm Leibniz University of Hannover Hannover Germany
| |
Collapse
|
32
|
Novák J, Vopálenský V, Pospíšek M, Vedeler A. Co-localization of Interleukin-1α and Annexin A2 at the plasma membrane in response to oxidative stress. Cytokine 2020; 133:155141. [PMID: 32615410 DOI: 10.1016/j.cyto.2020.155141] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/11/2020] [Accepted: 05/15/2020] [Indexed: 12/19/2022]
Abstract
Interleukin-1α (IL-1α) and Annexin A2 (AnxA2) are pleiotropic molecules with both intracellular and extracellular roles. They share several characteristics including unconventional secretion aided by S100 proteins, anchoring of the externalized proteins at the outer surface of the plasma membrane and response to oxidative stress. Although IL-1α and AnxA2 have been implicated in a variety of biological processes, including cancer, little is known about the mechanisms of their cellular release. In the present study, employing the non-cancerous breast epithelial MCF10A cells, we demonstrate that IL-1α and AnxA2 establish a close association in response to oxidative stress. Stress conditions lead to translocation of both proteins towards lamellipodia rich in vimentin and association of full-length IL-1α and Tyr23 phosphorylated AnxA2 with the plasma membrane at peripheral sites depleted of F-actin. Notably, membrane-associated IL-1α and AnxA2 preferentially localize to the outer edges of the MCF10A cell islands, suggesting that the two proteins participate in the communication of these epithelial cells with their neighboring cells. Similarly, in U2OS osteosarcoma cell line both endogenous IL-1α and transiently produced IL-1α/EGFP associate with the plasma membrane. While benign MFC10A cells present membrane-associated IL-1α and AnxA2 at the edges of their cell islands, the aggressive cancerous U2OS cells communicate in such manner also with distant cells.
Collapse
Affiliation(s)
- Josef Novák
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Prague, Czech Republic; Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway.
| | - Václav Vopálenský
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Martin Pospíšek
- Department of Genetics and Microbiology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Anni Vedeler
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
33
|
Kozisek T, Hamann A, Nguyen A, Miller M, Plautz S, Pannier AK. High-throughput screening of clinically approved drugs that prime nonviral gene delivery to human Mesenchymal stem cells. J Biol Eng 2020; 14:16. [PMID: 32467728 PMCID: PMC7238544 DOI: 10.1186/s13036-020-00238-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/04/2020] [Indexed: 01/07/2023] Open
Abstract
Background Human mesenchymal stem cells (hMSCs) are intensely researched for applications in cell therapeutics due to their unique properties, however, intrinsic therapeutic properties of hMSCs could be enhanced by genetic modification. Viral transduction is efficient, but suffers from safety issues. Conversely, nonviral gene delivery, while safer compared to viral, suffers from inefficiency and cytotoxicity, especially in hMSCs. To address the shortcomings of nonviral gene delivery to hMSCs, our lab has previously demonstrated that pharmacological 'priming' of hMSCs with the glucocorticoid dexamethasone can significantly increase transfection in hMSCs by modulating transfection-induced cytotoxicity. This work seeks to establish a library of transfection priming compounds for hMSCs by screening 707 FDA-approved drugs, belonging to diverse drug classes, from the NIH Clinical Collection at four concentrations for their ability to modulate nonviral gene delivery to adipose-derived hMSCs from two human donors. Results Microscope images of cells transfected with a fluorescent transgene were analyzed in order to identify compounds that significantly affected hMSC transfection without significant toxicity. Compound classes that increased transfection across both donors included glucocorticoids, antibiotics, and antihypertensives. Notably, clobetasol propionate, a glucocorticoid, increased transgene production 18-fold over unprimed transfection. Furthermore, compound classes that decreased transfection across both donors included flavonoids, antibiotics, and antihypertensives, with the flavonoid epigallocatechin gallate decreasing transgene production - 41-fold compared to unprimed transfection. Conclusions Our screen of the NCC is the first high-throughput and drug-repurposing approach to identify nonviral gene delivery priming compounds in two donors of hMSCs. Priming compounds and classes identified in this screen suggest that modulation of proliferation, mitochondrial function, and apoptosis is vital for enhancing nonviral gene delivery to hMSCs.
Collapse
Affiliation(s)
- Tyler Kozisek
- 1Department of Biological Systems Engineering, University of Nebraska-Lincoln, 231 L.W. Chase Hall, Lincoln, NE USA
| | - Andrew Hamann
- 1Department of Biological Systems Engineering, University of Nebraska-Lincoln, 231 L.W. Chase Hall, Lincoln, NE USA
| | - Albert Nguyen
- 1Department of Biological Systems Engineering, University of Nebraska-Lincoln, 231 L.W. Chase Hall, Lincoln, NE USA
| | - Michael Miller
- 2Department of Biomedical Engineering, Pennsylvania State University, 122 Chemical and Biomedical Engineering Building, University Park, PA USA
| | - Sarah Plautz
- 1Department of Biological Systems Engineering, University of Nebraska-Lincoln, 231 L.W. Chase Hall, Lincoln, NE USA
| | - Angela K Pannier
- 1Department of Biological Systems Engineering, University of Nebraska-Lincoln, 231 L.W. Chase Hall, Lincoln, NE USA
| |
Collapse
|
34
|
Blackstock DJ, Goh A, Shetty S, Fabozzi G, Yang R, Ivleva VB, Schwartz R, Horwitz J. Comprehensive Flow Cytometry Analysis of PEI-Based Transfections for Virus-Like Particle Production. RESEARCH 2020; 2020:1387402. [PMID: 32259105 PMCID: PMC7094759 DOI: 10.34133/2020/1387402] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 01/28/2020] [Indexed: 11/06/2022]
Abstract
The generation of stable clones for biomolecule production is a common but lengthy and labor-intensive process. For complex molecules, such as viruses or virus-like particles (VLPs), the timeline becomes even more cumbersome. Thus, in the early stages of development, transient production methods serve as a reasonable alternative to stable clone construction. In this work, an investigation of a polyethylenimine- (PEI-) based transfection method for the transient production of Chikungunya (Chik) VLPs, a vaccine candidate molecule, was undertaken. This effort focuses on tracking cell population responses during transfection, understanding how process changes affect these responses, and monitoring patterns in cell performance over the culture duration. Plasmid labeling and VLP staining were employed to comprehensively track cells via flow cytometry and to draw correlations between plasmid DNA (pDNA) uptake and the resulting VLP expression. The method detected high transfection efficiency (≥97%) in all samples tested and demonstrated the capability to track kinetics of plasmid-cell binding. With varied transfection cell concentrations, the pDNA binding kinetics are altered and saturation binding is observed in the lowest cell concentration sample tested in less than 3 hours of incubation. Interestingly, in all samples, the flow cytometry analysis of relative pDNA amount versus VLP expression staining showed that cells which contained fewer pDNA complexes resulted in the highest levels of VLP stain. Finally, to determine the potential breadth of our observations, we compared daily expression patterns of ChikVLP with a reporter, monomeric GFP molecule. The similarities detected suggest the interpretations presented here to likely be more broadly informative and applicable to PEI-based transient production of additional biological products as well.
Collapse
Affiliation(s)
- Daniel J Blackstock
- Vaccine Production Program Lab, Vaccine Research Center, NIAID, NIH, Gaithersburg, MD, USA
| | - Alvenne Goh
- Vaccine Production Program Lab, Vaccine Research Center, NIAID, NIH, Gaithersburg, MD, USA
| | - Shamitha Shetty
- Vaccine Production Program Lab, Vaccine Research Center, NIAID, NIH, Gaithersburg, MD, USA
| | - Giulia Fabozzi
- Vaccine Production Program Lab, Vaccine Research Center, NIAID, NIH, Gaithersburg, MD, USA
| | - Rong Yang
- Vaccine Production Program Lab, Vaccine Research Center, NIAID, NIH, Gaithersburg, MD, USA
| | - Vera B Ivleva
- Vaccine Production Program Lab, Vaccine Research Center, NIAID, NIH, Gaithersburg, MD, USA
| | - Richard Schwartz
- Vaccine Production Program Lab, Vaccine Research Center, NIAID, NIH, Gaithersburg, MD, USA
| | - Joseph Horwitz
- Vaccine Production Program Lab, Vaccine Research Center, NIAID, NIH, Gaithersburg, MD, USA
| |
Collapse
|
35
|
Patiño Vargas MI, Mesa Cadavid M, Arenas Gómez CM, Diosa Arango J, Restrepo Múnera LM, Becerra Colorado NY. Polyplexes System to Enhance the LL-37 Antimicrobial Peptide Expression in Human Skin Cells. Tissue Eng Part A 2020; 26:400-410. [PMID: 31805827 DOI: 10.1089/ten.tea.2019.0196] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Inefficient autologous tissue recovery in diverse skin injuries increases the susceptibility of patients to infections caused by multiresistant microorganisms, resulting in a high mortality rate. Nonviral transfection is an attractive alternative for these patients, where genetically modified cells incorporated into skin substitutes could release additional antimicrobial agents into the native skin. In this work, we have modulated the conditions of using a nonviral system for transfection of primary human keratinocytes and fibroblasts, consisting of a polymer/plasmid DNA (pDNA) complex called polyplex and its effects on the expression of LL-37 antimicrobial peptide. Linear and branched polyethylenimine (PEI) polymers in different weight concentrations were varied for evaluating the formation and colloidal characteristics of the polyplexes. The PEI/pDNA polyplexes with 19 nitrogen/phosphate ratio are nanometric particles (400 and 250 nm with linear and branched PEI, respectively) exhibiting positive surface (+30 ± 2 mV). Both kinds of polyplexes allowed the expression of a reporter gene and increased the human cathelicidin antimicrobial peptide gene expression in transfected keratinocytes and fibroblasts; however, greater cytotoxicity was observed when polyplexes formed with branched PEI were used. Moreover, cell culture supernatants from transfected cells with linear PEI/pDNA polyplexes showed enhanced antimicrobial activity (decrease of bacterial growth in 95.8%) against a Staphylococcus aureus strain in vitro. The study of the PEI/pDNA polyplexes formation allowed us to develop an improved transfection strategy of skin cells, promoting the production of LL-37 antimicrobial peptide. In the future, this strategy could be used for the construction of skin substitutes to prevent, reduce, or eliminate bacterial infections. Impact statement The results of this study contribute to the understanding of the polyplexes system in the genetic modification of skin cells and its effects on the expression of the LL-37 antimicrobial peptide. In the future, three-dimensional skin substitutes built with these cells could be an efficient way to decrease bacterial growth and prevent the infections in skin wounds.
Collapse
Affiliation(s)
- Maria Isabel Patiño Vargas
- Tissue Engineering and Cell Therapy Group, Faculty of Medicine, University of Antioquia, Medellín, Colombia
| | - Mónica Mesa Cadavid
- Materials Science Group, Faculty of Exact and Natural Sciences, The University Research Headquarters (SIU), University of Antioquia, Medellín, Colombia
| | - Claudia Marcela Arenas Gómez
- Marine Biological Laboratory, Eugene Bell Center for Regenerative Biology and Tissue Engineering, Woods Hole, Massachusetts
| | - Johnatan Diosa Arango
- Materials Science Group, Faculty of Exact and Natural Sciences, The University Research Headquarters (SIU), University of Antioquia, Medellín, Colombia
| | - Luz Marina Restrepo Múnera
- Tissue Engineering and Cell Therapy Group, Faculty of Medicine, University of Antioquia, Medellín, Colombia
| | | |
Collapse
|
36
|
Roles of the G protein-coupled receptor kinase 2 and Rab5 in α 1B-adrenergic receptor function and internalization. Eur J Pharmacol 2020; 867:172846. [PMID: 31811856 DOI: 10.1016/j.ejphar.2019.172846] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/21/2019] [Accepted: 12/03/2019] [Indexed: 11/23/2022]
Abstract
Cells expressing eGFP-tagged Rab5 (wild-type or the GDP-Rab5 mutant) and the DsRed-tagged α1B-adrenergic receptors were employed and the roles of GRK2 were studied utilizing paroxetine and the dominant-negative mutant of GRK2 (DN-GRK2). The following parameters were studied: a) FRET (as an index of α1B-adrenergic receptor-Rab5 interaction): b) intracellular accumulation of DsRed fluorescence (receptor internalization); c) α1B-adrenergic receptor phosphorylation, and d) noradrenaline-induced increase in intracellular calcium concentration. Noradrenaline increased α1B-adrenergic receptor-Rab5 interaction, which was blocked by paroxetine and by expression of the dominant-negative GRK2 mutant. Similarly, paroxetine and expression of the DN-GRK2 or the GDP-Rab5 mutants markedly decreased receptor internalization, α1B-adrenergic receptor phosphorylation, and attenuated the ability of the adrenergic agonist to induce homologous desensitization (calcium signaling). The S406, 410,412A α1B-adrenergic receptor mutant did not reproduce the actions of GRK2 inhibition. The data indicate that GRK2 and Rab5 play key roles in α1B-adrenergic receptor phosphorylation, internalization, and desensitization. The possibility that Rab5 might form part of a signaling complex is suggested, as well as that GDP-Rab5 might interfere with the ability of GRK2 to catalyze α1B-adrenergic receptor phosphorylation.
Collapse
|
37
|
Cao Y, Qiu T, Kathayat RS, Azizi SA, Thorne AK, Ahn D, Fukata Y, Fukata M, Rice PA, Dickinson BC. ABHD10 is an S-depalmitoylase affecting redox homeostasis through peroxiredoxin-5. Nat Chem Biol 2019; 15:1232-1240. [PMID: 31740833 PMCID: PMC6871660 DOI: 10.1038/s41589-019-0399-y] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 09/30/2019] [Indexed: 12/12/2022]
Abstract
S-palmitoylation is a reversible lipid post-translational modification that has been observed on mitochondrial proteins, but both the regulation and functional consequences of mitochondrial S-palmitoylation are poorly understood. Here, we show that perturbing the “erasers” of S-palmitoylation, acyl protein thioesterases (APTs), with either pan-active inhibitors or a new mitochondrial-targeted APT inhibitor, diminishes the antioxidant buffering capacity of mitochondria. Surprisingly, this effect was not mediated by the only known mitochondrial APT, but rather by a resident mitochondrial protein with no known endogenous function, ABHD10. We show that ABHD10 is a new member of the APT family of regulatory proteins and identify peroxiredoxin 5 (PRDX5), a key antioxidant protein, as the first target of ABHD10 S-depalmitoylase activity. We then discover that ABHD10 regulates the S-palmitoylation status of the nucleophilic active site residue of PRDX5, providing a direct mechanistic connection between ABHD10-mediated S-depalmitoylation of PRDX5 and its antioxidant capacity.
Collapse
Affiliation(s)
- Yang Cao
- Department of Chemistry, The University of Chicago, Chicago, IL, USA
| | - Tian Qiu
- Department of Chemistry, The University of Chicago, Chicago, IL, USA
| | - Rahul S Kathayat
- Department of Chemistry, The University of Chicago, Chicago, IL, USA
| | - Saara-Anne Azizi
- Department of Chemistry, The University of Chicago, Chicago, IL, USA.,Medical Scientist Training Program, Pritzker School of Medicine, The University of Chicago, Chicago, IL, USA
| | - Anneke K Thorne
- Department of Chemistry, The University of Chicago, Chicago, IL, USA
| | - Daniel Ahn
- Department of Chemistry, The University of Chicago, Chicago, IL, USA
| | - Yuko Fukata
- Division of Membrane Physiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
| | - Masaki Fukata
- Division of Membrane Physiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan
| | - Phoebe A Rice
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA
| | - Bryan C Dickinson
- Department of Chemistry, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
38
|
Herrera LC, Shastri VP. Silencing of GFP expression in human mesenchymal stem cells using quaternary polyplexes of siRNA-PEI with glycosaminoglycans and albumin. Acta Biomater 2019; 99:397-411. [PMID: 31541736 DOI: 10.1016/j.actbio.2019.09.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 08/24/2019] [Accepted: 09/08/2019] [Indexed: 12/19/2022]
Abstract
In recent years evidence has been mounting for a role for mesenchymal stem cells (MSCs) in immunomodulation, anti-inflammatory processes, and paracrine signaling via secreted extracellular vesicles. In order to exploit these biological functions, systems to efficiently deliver genetic material into MSCs would therefore be highly desirable. In this study, efficient silencing of GFP expression by combining high N/P ratio siRNA and branched PEI (bPEI) polyplexes (siRNA-bPEI) polyplexes with glycosaminoglycans (GAGs), namely hyaluronic acid (HA), chondroitin sulfate (CS) and heparin sulfate (HS), and human serum albumin (HSA) is reported. These quaternary systems were characterized using surface charge, size and morphology and applied to MSCs, which represent a challenge due to their typically low transfection efficiency. The quaternary polyplexes promoted efficient charge shielding and release of siRNA in the cytoplasm with reduced toxicity. A high silencing efficiency of >90% (i.e., less than 10% remaining GFP expression) was achieved with noticeably reduced cellular toxicity, especially with siRNA-bPEI polyplexes modified with HA and HA + HSA. In general addition of GAGs led to more compact polyplexes. Endocytosis studies point to improved endosomal escape at high N/P ratios as a reason for high transfection efficiency and a role for hyaluronic acid in the uptake mechanism likely via CD44 interactions. Co-localization studies showed the polyplexes are stable in the cytosol over time, which correlates with a proper disassembly and subsequent silencing of GFP. Furthermore, GAG containing polyplexes were frequently co-localized with the nucleus. These findings in sum suggest that PEI/HSA/GAG based quaternary polyplexes are promising as transfection agents for MSCs. STATEMENT OF SIGNIFICANCE: Since mesenchymal stem cells (MSCs) are recruited to the site of tissue repair and play a role in immunomodulation, anti-inflammatory processes, and paracrine signaling, they present an excellent target for genetic engineering. However, delivery of genetic material into MSCs is challenging. In this study, >97% silencing of constitutive green fluorescent protein expression in human MSCs (hMSCs) using high N/P ratio polyplexes of branched-PEI-siRNA incorporating glycosaminoglycan as a charge neutralizer and human serum albumin as co-complexing agent is demonstrated. In addition to possessing good cytocompatibility and excellent cytosolic stability; polyplexes incorporating GAGs also showed altered endocytic uptake, with incorporation of hyaluronic acid promoting caveolae-mediated entry. Our system highlights the importance of physiologically derived macromolecules in delivery of genetic material into hMSCs.
Collapse
Affiliation(s)
- Laura C Herrera
- Institute for Macromolecular Chemistry, University of Freiburg, 79104 Freiburg, Germany
| | - V Prasad Shastri
- Institute for Macromolecular Chemistry, University of Freiburg, 79104 Freiburg, Germany; BIOSS-Centre for Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany.
| |
Collapse
|
39
|
Xu X, Wan T, Xin H, Li D, Pan H, Wu J, Ping Y. Delivery of CRISPR/Cas9 for therapeutic genome editing. J Gene Med 2019; 21:e3107. [PMID: 31237055 DOI: 10.1002/jgm.3107] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/25/2019] [Accepted: 06/11/2019] [Indexed: 12/15/2022] Open
Abstract
The clustered, regularly-interspaced, short palindromic repeat (CRISPR)-associated nuclease 9 (CRISPR/Cas9) is emerging as a promising genome-editing tool for treating diseases in a precise way, and has been applied to a wide range of research in the areas of biology, genetics, and medicine. Delivery of therapeutic genome-editing agents provides a promising platform for the treatment of genetic disorders. Although viral vectors are widely used to deliver CRISPR/Cas9 elements with high efficiency, they suffer from several drawbacks, such as mutagenesis, immunogenicity, and off-target effects. Recently, non-viral vectors have emerged as another class of delivery carriers in terms of their safety, simplicity, and flexibility. In this review, we discuss the modes of CRISPR/Cas9 delivery, the barriers to the delivery process and the application of CRISPR/Cas9 system for the treatment of genetic disorders. We also highlight several representative types of non-viral vectors, including polymers, liposomes, cell-penetrating peptides, and other synthetic vectors, for the therapeutic delivery of CRISPR/Cas9 system. The applications of CRISPR/Cas9 in treating genetic disorders mediated by the non-viral vectors are also discussed.
Collapse
Affiliation(s)
- Xiaojie Xu
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, China
| | - Tao Wan
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, China
| | - Huhu Xin
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, China
| | - Da Li
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, China
| | - Jun Wu
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yuan Ping
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, China
| |
Collapse
|
40
|
Buck J, Grossen P, Cullis PR, Huwyler J, Witzigmann D. Lipid-Based DNA Therapeutics: Hallmarks of Non-Viral Gene Delivery. ACS NANO 2019; 13:3754-3782. [PMID: 30908008 DOI: 10.1021/acsnano.8b07858] [Citation(s) in RCA: 229] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Gene therapy is a promising strategy for the treatment of monogenic disorders. Non-viral gene delivery systems including lipid-based DNA therapeutics offer the opportunity to deliver an encoding gene sequence specifically to the target tissue and thus enable the expression of therapeutic proteins in diseased cells. Currently, available gene delivery approaches based on DNA are inefficient and require improvements to achieve clinical utility. In this Review, we discuss state-of-the-art lipid-based DNA delivery systems that have been investigated in a preclinical setting. We emphasize factors influencing the delivery and subsequent gene expression in vitro, ex vivo, and in vivo. In addition, we cover aspects of nanoparticle engineering and optimization for DNA therapeutics. Finally, we highlight achievements of lipid-based DNA therapies in clinical trials.
Collapse
Affiliation(s)
- Jonas Buck
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences , University of Basel , Klingelbergstrasse 50 , 4056 Basel , Switzerland
| | - Philip Grossen
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences , University of Basel , Klingelbergstrasse 50 , 4056 Basel , Switzerland
| | - Pieter R Cullis
- Department of Biochemistry and Molecular Biology , University of British Columbia , 2350 Health Sciences Mall , Vancouver , British Columbia V6T 1Z3 , Canada
| | - Jörg Huwyler
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences , University of Basel , Klingelbergstrasse 50 , 4056 Basel , Switzerland
| | - Dominik Witzigmann
- Division of Pharmaceutical Technology, Department of Pharmaceutical Sciences , University of Basel , Klingelbergstrasse 50 , 4056 Basel , Switzerland
- Department of Biochemistry and Molecular Biology , University of British Columbia , 2350 Health Sciences Mall , Vancouver , British Columbia V6T 1Z3 , Canada
| |
Collapse
|
41
|
Thapa B, Remant KC, Uludağ H. siRNA Library Screening to Identify Complementary Therapeutic Pairs in Triple-Negative Breast Cancer Cells. Methods Mol Biol 2019; 1974:1-19. [PMID: 31098991 DOI: 10.1007/978-1-4939-9220-1_1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The existence of tightly integrated cross talk through multiple signaling and effector pathways has been appreciated in malignant cells. The realization of the plasticity of such networks is stimulating the development of combinational therapy to overcome the limitations of one-dimensional therapies. Synergistic pairs of siRNAs or siRNA and drug combinations are the new frontiers in identifying effective therapeutic combinations. To elucidate effective combinations, we developed a versatile protocol to screen siRNA libraries in triple-negative breast cancer cell models. This protocol outlines the steps to identify synergistic combinations of siRNA-siRNA or siRNA-drug combinations using siRNA libraries via a robotic screen. By focusing on smaller functional siRNA libraries, we present methodologies to identify synergistic siRNA pairings against cancerous cell growth and molecular targets to augment the activity of pro-apoptotic TRAIL protein. Here, we summarize the critical steps to undertake such combinational target identification, emphasizing critical factors that affect the outcome of the screens. Our experience suggests that siRNA library screening is an efficient protocol to identify complementary therapeutic pairs of new or already-existing drugs. This protocol is simple, robust and can be completed within a 1-week working period.
Collapse
Affiliation(s)
- Bindu Thapa
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - K C Remant
- Department of Chemical and Material Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada
| | - Hasan Uludağ
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada.
- Department of Chemical and Material Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, Canada.
- Department of Biomedical Engineering, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
42
|
S1P 1 receptor phosphorylation, internalization, and interaction with Rab proteins: effects of sphingosine 1-phosphate, FTY720-P, phorbol esters, and paroxetine. Biosci Rep 2018; 38:BSR20181612. [PMID: 30366961 PMCID: PMC6294635 DOI: 10.1042/bsr20181612] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 10/19/2018] [Accepted: 10/26/2018] [Indexed: 01/04/2023] Open
Abstract
Sphingosine 1-phosphate (S1P) and FTY720-phosphate (FTYp) increased intracellular calcium in cells expressing S1P1 mCherry-tagged receptors; the synthetic agonist was considerably less potent. Activation of protein kinase C by phorbol myristate acetate (PMA) blocked these effects. The three agents induced receptor phosphorylation and internalization, with the action of FTYp being more intense. S1P1 receptor–Rab protein (GFP-tagged) interaction was studied using FRET. The three agents were able to induce S1P1 receptor–Rab5 interaction, although with different time courses. S1P1 receptor–Rab9 interaction was mainly increased by the phorbol ester, whereas S1P1 receptor–Rab7 interaction was only increased by FTYp and after a 30-min incubation. These actions were not observed using dominant negative (GDP-bound) Rab protein mutants. The data suggested that the three agents induce interaction with early endosomes, but that the natural agonist induced rapid receptor recycling, whereas activation of protein kinase C favored interaction with late endosome and slow recycling and FTYp triggered receptor interaction with vesicles associated with proteasomal/lysosomal degradation. The ability of bisindolylmaleimide I and paroxetine to block some of these actions suggested the activation of protein kinase C was associated mainly with the action of PMA, whereas G protein-coupled receptor kinase (GRK) 2 (GRK2) was involved in the action of the three agents.
Collapse
|
43
|
Pan Z, Di S, Shi B, Jiang H, Shi Z, Liu Y, Wang Y, Luo H, Yu M, Wu X, Li Z. Increased antitumor activities of glypican-3-specific chimeric antigen receptor-modified T cells by coexpression of a soluble PD1-CH3 fusion protein. Cancer Immunol Immunother 2018; 67:1621-1634. [PMID: 30078052 PMCID: PMC11028056 DOI: 10.1007/s00262-018-2221-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 07/27/2018] [Indexed: 12/16/2022]
Abstract
Our recent clinical study demonstrated that glypican-3 (GPC3)-specific chimeric antigen receptor-modified T (CAR-T) cells are a promising treatment for hepatocellular carcinoma (HCC). However, the interaction of programmed cell death 1 (PD-1) and PD-L1-mediated T-cell inhibition is involved in immune evasion in a wide range of solid tumors, including HCC. To overcome this problem, we introduced a fusion protein composed of a PD-1 extracellular domain and CH3 from IgG4 into GPC3-specific CAR-T cells (GPC3-28Z) to block the PD-1/PD-L1 pathway. GPC3-specific CAR-T cells carrying the PD-1-CH3 fusion protein (sPD1) specifically recognized and lysed GPC3-positive HCC cells. The proliferation capacity of GPC3-28Z-sPD1 T cells after weekly stimulation with target cells was much higher than that of control GPC3-28Z T cells. Additionally, the coexpression of sPD1 could protect CAR-T cells from exhaustion when incubated with target cells, as phosphorylated AKT and Bcl-xL expression levels were higher in GPC3-28Z-sPD1 T cells than in GPC3-28Z cells. Importantly, in two HCC tumor xenograft models, GPC3-28Z-sPD1 T cells displayed a significantly higher tumor suppression capacity than GPC3-28Z T cells. In addition, an increased number of CD3+ T cells in the circulation and tumors and increased granzyme B levels and decreased Ki67 expression levels in the tumors were observed in the mice treated with GPC3-28Z-sPD1 T cells. Together, these data indicated that GPC3-specific CAR-T cells carrying sPD1 show promise as a treatment for patients with HCC.
Collapse
MESH Headings
- Animals
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/prevention & control
- Cells, Cultured
- Glypicans/immunology
- Glypicans/metabolism
- Humans
- Immunoglobulin G/immunology
- Immunoglobulin G/metabolism
- Liver Neoplasms/immunology
- Liver Neoplasms/metabolism
- Liver Neoplasms/prevention & control
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Programmed Cell Death 1 Receptor/immunology
- Programmed Cell Death 1 Receptor/metabolism
- Protein Domains
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Zeyan Pan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, No. 25/Ln. 2200, XieTu Road, Shanghai, 200032, China
| | - Shengmeng Di
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, No. 25/Ln. 2200, XieTu Road, Shanghai, 200032, China
| | - Bizhi Shi
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, No. 25/Ln. 2200, XieTu Road, Shanghai, 200032, China
| | - Hua Jiang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, No. 25/Ln. 2200, XieTu Road, Shanghai, 200032, China
| | | | - Ying Liu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, No. 25/Ln. 2200, XieTu Road, Shanghai, 200032, China
| | - Yi Wang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, No. 25/Ln. 2200, XieTu Road, Shanghai, 200032, China
| | - Hong Luo
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, No. 25/Ln. 2200, XieTu Road, Shanghai, 200032, China
| | - Min Yu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, No. 25/Ln. 2200, XieTu Road, Shanghai, 200032, China
| | - Xiuqi Wu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, No. 25/Ln. 2200, XieTu Road, Shanghai, 200032, China
| | - Zonghai Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, No. 25/Ln. 2200, XieTu Road, Shanghai, 200032, China.
- CARsgen Therapeutics, Shanghai, China.
| |
Collapse
|
44
|
Comparison between electroporation and polyfection in pig sperm: efficiency and cell viability implications. ZYGOTE 2018; 26:286-293. [DOI: 10.1017/s0967199418000205] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
SummaryThe aim of this study was to optimize protocols for electroporation (EP) and polyfection (PLF) using polyethyleneimine (PEI) for pig sperm transfection and to determine which method was the most efficient. For EP standardization, different voltages, amounts and times of electric pulses were tested using propidium iodide (PI) as reporter. For PLF standardization, different concentrations of fluorescein isothiocyanate (FITC)-labelled PEI (PEI/FITC) were incubated with sperm for different periods of time. Flow cytometry was performed to evaluate the best protocol in terms of cell viability, including cytoplasmic membrane, acrosome, chromatin integrities and mitochondrial potential using the FITC probe, PI, acridine orange (AO) and JC1. Transfections with the plasmid pmhyGENIE-5 were carried out under optimum conditions for each procedure (EP: 500 volts, 500 μs and two pulses; PLF: PEI 0.5 mg/ml and incubation time 10 min). Transfection efficacy was assessed by fluorescence in situ hybridization (FISH). A lower transfection rate was observed for sperm in the control group (17.8%) compared with EP (36.7%), with PLF (76.8%) being the most efficient. These results suggest that the EP and PEI could be an efficient and low cost transfection method for swine sperm. Notably, treated cells showed higher plasmatic the membrane damage (PMD) and/or acrosome damage (AD) indexes, therefore the combination of this procedure with biotechniques that facilitate fecundation (i.e. in vitro fertilization or intracytoplasmic sperm injection) or even inclusion of antioxidant or anti-apoptotic drugs to improve spermatozoa viability would be important.
Collapse
|
45
|
Meizoso‐Huesca A, Villegas‐Comonfort S, Romero‐Ávila MT, García‐Sáinz JA. Free fatty acid receptor 4 agonists induce lysophosphatidic acid receptor 1 (
LPA
1
) desensitization independent of
LPA
1
internalization and heterodimerization. FEBS Lett 2018; 592:2612-2623. [DOI: 10.1002/1873-3468.13179] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 06/09/2018] [Accepted: 06/27/2018] [Indexed: 01/27/2023]
Affiliation(s)
- Aldo Meizoso‐Huesca
- Departamento de Biología Celular y del Desarrollo Instituto de Fisiología Celular Universidad Nacional Autónoma de México Mexico
| | - Sócrates Villegas‐Comonfort
- Departamento de Biología Celular y del Desarrollo Instituto de Fisiología Celular Universidad Nacional Autónoma de México Mexico
| | - M. Teresa Romero‐Ávila
- Departamento de Biología Celular y del Desarrollo Instituto de Fisiología Celular Universidad Nacional Autónoma de México Mexico
| | - J. Adolfo García‐Sáinz
- Departamento de Biología Celular y del Desarrollo Instituto de Fisiología Celular Universidad Nacional Autónoma de México Mexico
| |
Collapse
|
46
|
Hume RD, Berry L, Reichelt S, D'Angelo M, Gomm J, Cameron RE, Watson CJ. An Engineered Human Adipose/Collagen Model for In Vitro Breast Cancer Cell Migration Studies. Tissue Eng Part A 2018; 24:1309-1319. [PMID: 29652604 DOI: 10.1089/ten.tea.2017.0509] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Adipocytes are one of the major stromal cell components of the human breast. These cells play a key role in the development of the gland and are implicated in breast tumorigenesis. Frequently, directional stromal collagen I fibers are found surrounding aggressive breast tumors. These fibers enhance breast cancer cell migration and are associated with poor patient prognosis. We sought to recapitulate these stromal components in vitro to provide a three-dimensional (3D) model comprising human adipose tissue and anisotropic collagen fibers. We developed a human mesenchymal stem cell (hMSC) cell line capable of undergoing differentiation into mature adipocytes by immortalizing hMSCs, isolated from breast reduction mammoplasties, through retroviral transduction. These immortalized hMSCs were seeded in engineered collagen I scaffolds with directional internal architecture, and adipogenesis was chemically induced, resulting in human adipose tissue being synthesized in vitro in an architectural structure associated with breast tumorigenesis. Subsequently, fluorescently labeled cells from an established breast cancer cell line were seeded into this model, cocultured for 7 days and imaged using multiphoton microscopy. Enhanced breast cancer cell migration was observed in the adipose-containing model over empty scaffold controls, demonstrating an adipocyte-mediated influence on breast cancer cell migration. Thus, this 3D in vitro model recapitulates the migratory effects of adipocytes observed on breast cancer cells and suggests that it could have utility with fresh breast tumor biopsies as an assay for cancer therapeutic efficacy in personalized medicine strategies.
Collapse
Affiliation(s)
- Robert D Hume
- 1 Department of Pathology, University of Cambridge , Cambridge, United Kingdom
| | - Lorraine Berry
- 2 Cancer Research UK Cambridge Institute, University of Cambridge , Cambridge, United Kingdom
| | - Stefanie Reichelt
- 2 Cancer Research UK Cambridge Institute, University of Cambridge , Cambridge, United Kingdom
| | - Michael D'Angelo
- 1 Department of Pathology, University of Cambridge , Cambridge, United Kingdom
| | - Jenny Gomm
- 3 Barts Cancer Institute, Queen Mary University of London , London, United Kingdom
| | - Ruth E Cameron
- 4 Department of Materials Science and Metallurgy, University of Cambridge , Cambridge, United Kingdom
| | - Christine J Watson
- 1 Department of Pathology, University of Cambridge , Cambridge, United Kingdom
| |
Collapse
|
47
|
Becerra Colorado NY, Arenas Gómez CM, Patiño Vargas MI, Delgado Charris JP, Muskus López CE, Restrepo Múnera LM. Polyplex System Versus Nucleofection for Human Skin Cell Transfection and Effect of Internal Ribosome Entry Site Sequence. Tissue Eng Part C Methods 2018; 24:233-241. [DOI: 10.1089/ten.tec.2017.0435] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Natalia Yiset Becerra Colorado
- Tissue Engineering and Cell Therapy Group, Faculty of Medicine, Universidad de Antioquia, Medellín, Colombia
- Biobank and Cell Therapy Laboratory, University IPS, Health Services, University of Antioquia, Medellín, Colombia
| | - Claudia Marcela Arenas Gómez
- Genetics, Regeneration and Cancer Group, University of Antioquia, Sede de Investigación Universitaria-SIU, Medellín, Colombia
| | - Maria Isabel Patiño Vargas
- Tissue Engineering and Cell Therapy Group, Faculty of Medicine, Universidad de Antioquia, Medellín, Colombia
| | - Jean Paul Delgado Charris
- Genetics, Regeneration and Cancer Group, University of Antioquia, Sede de Investigación Universitaria-SIU, Medellín, Colombia
| | - Carlos Enrique Muskus López
- The Program for the Study and Control of Tropical Diseases-PECET, University of Antioquia, Sede de Investigación Universitaria-SIU, Medellín, Colombia
| | - Luz Marina Restrepo Múnera
- Tissue Engineering and Cell Therapy Group, Faculty of Medicine, Universidad de Antioquia, Medellín, Colombia
| |
Collapse
|
48
|
Alfonzo-Méndez MA, Carmona-Rosas G, Hernández-Espinosa DA, Romero-Ávila MT, García-Sáinz JA. Different phosphorylation patterns regulate α 1D-adrenoceptor signaling and desensitization. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:842-854. [PMID: 29551601 DOI: 10.1016/j.bbamcr.2018.03.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 03/02/2018] [Accepted: 03/13/2018] [Indexed: 12/14/2022]
Abstract
Human α1D-adrenoceptors (α1D-ARs) are a group of the seven transmembrane-spanning proteins that mediate many of the physiological and pathophysiological actions of adrenaline and noradrenaline. Although it is known that α1D-ARs are phosphoproteins, their specific phosphorylation sites and the kinases involved in their phosphorylation remain largely unknown. Using a combination of in silico analysis, mass spectrometry and site directed mutagenesis, we identified distinct α1D-AR phosphorylation patterns during noradrenaline- or phorbol ester-mediated desensitizations. We found that the G protein coupled receptor kinase, GRK2, and conventional protein kinases C isoforms α/β, phosphorylate α1D-AR during these processes. Furthermore, we showed that the phosphorylated residues are located in the receptor's third intracellular loop (S300, S323, T328, S331, S332, S334) and carboxyl region (S441, T442, T477, S486, S492, T507, S515, S516, S518, S543) and are conserved among orthologues but are not conserved among the other human α1-adrenoceptor subtypes. Additionally, we found that phosphorylation in either the third intracellular loop or carboxyl tail was sufficient to regulate calcium signaling desensitization. By contrast, mutations in either of these two domains significantly altered mitogen activated protein kinase (ERK) pathway and receptor internalization, suggesting that they have differential regulatory mechanisms. Our data provide new insights into the functional repercussions of these posttranslational modifications in signaling outcomes and desensitization.
Collapse
Affiliation(s)
- Marco A Alfonzo-Méndez
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ap. Postal 70-248, Ciudad de México CP 04510, Mexico
| | - Gabriel Carmona-Rosas
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ap. Postal 70-248, Ciudad de México CP 04510, Mexico
| | - David A Hernández-Espinosa
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ap. Postal 70-248, Ciudad de México CP 04510, Mexico
| | - M Teresa Romero-Ávila
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ap. Postal 70-248, Ciudad de México CP 04510, Mexico
| | - J Adolfo García-Sáinz
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ap. Postal 70-248, Ciudad de México CP 04510, Mexico.
| |
Collapse
|
49
|
Ringer KP, Roth MG, Garey MS, Piorczynski TB, Suli A, Hansen JM, Alder JK. Comparative analysis of lipid-mediated CRISPR-Cas9 genome editing techniques. Cell Biol Int 2018; 42:849-858. [PMID: 29457665 DOI: 10.1002/cbin.10952] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 02/12/2018] [Indexed: 12/11/2022]
Abstract
CRISPR-Cas technology has revolutionized genome engineering. While Cas9 was not the first programmable endonuclease identified, its simplicity of use has driven widespread adoption in a short period of time. While CRISPR-Cas genome editing holds enormous potential for clinical applications, its use in laboratory settings for genotype-phenotype studies and genome-wide screens has led to breakthroughs in the understanding of many molecular pathways. Numerous protocols have been described for introducing CRISPR-Cas components into cells, and here we sought to simplify and optimize a protocol for genome editing using readily available and inexpensive tools. We compared plasmid, ribonucleoprotein (RNP), and RNA transfection to determine which was method was most optimal for editing cells in a laboratory setting. We limited our comparison to lipofection-mediated introduction because the reagents are widely available. To facilitate optimization, we developed a novel reporter assay to measure gene disruption and the introduction of a variety of exogenous DNA tags. Each method efficiently disrupted endogenous genes and was able to stimulate the introduction of foreign DNA at specific sites, albeit to varying efficiencies. RNP transfection produced the highest level of gene disruption and was the most rapid and efficient method overall. Finally, we show that very short homology arms of 30 base pairs can mediate site-specific editing. The methods described here should broaden the accessibility of RNP-mediated lipofection for laboratory genome-editing experiments.
Collapse
Affiliation(s)
- Kelsey P Ringer
- Physiology & Developmental Biology, Brigham Young University, Provo, Utah, 84602, USA
| | - Mark G Roth
- Physiology & Developmental Biology, Brigham Young University, Provo, Utah, 84602, USA
| | - Mitchell S Garey
- Physiology & Developmental Biology, Brigham Young University, Provo, Utah, 84602, USA
| | - Ted B Piorczynski
- Physiology & Developmental Biology, Brigham Young University, Provo, Utah, 84602, USA
| | - Arminda Suli
- Physiology & Developmental Biology, Brigham Young University, Provo, Utah, 84602, USA
| | - Jason M Hansen
- Physiology & Developmental Biology, Brigham Young University, Provo, Utah, 84602, USA
| | - Jonathan K Alder
- Physiology & Developmental Biology, Brigham Young University, Provo, Utah, 84602, USA
| |
Collapse
|
50
|
Chimeric antigen receptor T cells secreting anti-PD-L1 antibodies more effectively regress renal cell carcinoma in a humanized mouse model. Oncotarget 2018; 7:34341-55. [PMID: 27145284 PMCID: PMC5085160 DOI: 10.18632/oncotarget.9114] [Citation(s) in RCA: 256] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 04/16/2016] [Indexed: 01/03/2023] Open
Abstract
Advances in the treatment of metastatic clear cell renal cell carcinoma (ccRCC) have led to improved progression-free survival of many patients; however the therapies are toxic, rarely achieve durable long-term complete responses and are not curative. Herein we used a single bicistronic lentiviral vector to develop a new combination immunotherapy that consists of human anti-carbonic anhydrase IX (CAIX)-targeted chimeric antigen receptor (CAR) T cells engineered to secrete human anti-programmed death ligand 1 (PD-L1) antibodies at the tumor site. The local antibody delivery led to marked immune checkpoint blockade. Tumor growth diminished 5 times and tumor weight reduced 50–80% when compared with the anti-CAIX CAR T cells alone in a humanized mice model of ccRCC. The expression of PD-L1 and Ki67 in the tumors decreased and an increase in granzyme B levels was found in CAR T cells. The anti-PD-L1 IgG1 isotype, which is capable of mediating ADCC, was also able to recruit human NK cells to the tumor site in vivo. These armed second-generation CAR T cells empowered to secrete human anti-PD-L1 antibodies in the ccRCC milieu to combat T cell exhaustion is an innovation in this field that should provide renewed potential for CAR T cell immunotherapy of solid tumors where limited efficacy is currently seen.
Collapse
|