Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 58
Filtrer
1.
Acc Chem Res ; 57(16): 2358-2371, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39093824

RÉSUMÉ

ConspectusCells, particularly living cells, serve as natural carriers of bioactive substances. Their inherent low immunogenicity and multifunctionality have garnered significant attention in the realm of disease treatment applications, specifically within the domains of cancer immunotherapy and regenerative tissue repair. Nevertheless, several prominent challenges impede their swift translation into clinical applications, including obstacles related to large-scale production feasibility and high utilization costs. To address these issues comprehensively, researchers have proposed the notion of bionic cells that are synthetically generated through chemical or biosynthetic means to emulate cellular functions and behaviors. However, artificial cell strategies encounter difficulties in fully replicating the intricate functionalities exhibited by living cells while also grappling with the complexities associated with design implementation for clinical translation purposes. The convergence of disciplines has facilitated the reform of living cells through a range of approaches, including chemical-, biological-, genetic-, and materials-based methods. These techniques can be employed to impart specific functions to cells or enhance the efficacy of therapy. For example, cells are engineered through gene transduction, surface modifications, endocytosis of drugs as delivery systems, and membrane fusion. The concept of engineered cells presents a promising avenue for enhancing control over living cells, thereby enhancing therapeutic efficacy while concurrently mitigating toxic side effects and ultimately facilitating the realization of precision medicine.In this Account, we present a comprehensive overview of our recent research advancements in the field of engineered cells. Our work involves the application of biological or chemical engineering techniques to manipulate endogenous cells for therapeutics or drug delivery purposes. For instance, to avoid the laborious process of isolating, modifying, and expanding engineered cells in vitro, we proposed the concept of in situ engineered cells. By applying a hydrogel loaded with nanoparticles carrying edited chimeric antigen receptor (CAR) plasmids within the postoperative cavity of glioma, we successfully targeted tumor-associated macrophages for gene editing, leading to effective tumor recurrence inhibition. Furthermore, leveraging platelet's ability to release microparticles upon activation at injury sites, we modified antiprogrammed death 1 (PD-1) antibodies on their surface to suppress postoperative tumor recurrence and provide immunotherapy for inoperable tumors. Similarly, by exploiting bacteria's active tropism toward sites of inflammation and hypoxia, we delivered protein drugs by engineered bacteria to induce cancer cell death through pyroptosis initiation and immunotherapy strategies. In the final section, we summarize our aforementioned research progress while providing an outlook on cancer therapy and the hurdles for clinical translation with potential solutions or future directions based on the concept of engineered cells.


Sujet(s)
Ingénierie cellulaire , Tumeurs , Humains , Tumeurs/thérapie , Ingénierie cellulaire/méthodes , Animaux , Immunothérapie
2.
Appl Microbiol Biotechnol ; 108(1): 434, 2024 Aug 09.
Article de Anglais | MEDLINE | ID: mdl-39120640

RÉSUMÉ

Chinese hamster ovary (CHO) cells, widely acknowledged as the preferred host system for industrial recombinant protein manufacturing, play a crucial role in developing pharmaceuticals, including anticancer therapeutics. Nevertheless, mammalian cell-based biopharmaceutical production methods are still beset by cellular constraints such as limited growth and poor productivity. MicroRNA-21 (miR-21) has a major impact on a variety of malignancies, including glioblastoma multiforme (GBM). However, reduced productivity and growth rate have been linked to miR-21 overexpression in CHO cells. The current study aimed to engineer a recombinant CHO (rCHO) cell using the CRISPR-mediated precise integration into target chromosome (CRIS-PITCh) system coupled with the Bxb1 recombinase-mediated cassette exchange (RMCE) to express a circular miR-21 decoy (CM21D) with five bulged binding sites for miR-21 sponging. Implementing the ribonucleoprotein (RNP) delivery method, a landing pad was inserted into the genome utilizing the CRIS-PITCh technique. Subsequently, the CM21D cassette flanked by Bxb1 attB was then retargeted into the integrated landing pad using the RMCE/Bxb1 system. This strategy raised the targeting efficiency by 1.7-fold, and off-target effects were decreased. The miR-21 target genes (Pdcd4 and Atp11b) noticed a significant increase in expression upon the miR-21 sponging through CM21D. Following the expression of CM21D, rCHO cells showed a substantial decrease in doubling time and a 1.3-fold increase in growth rate. Further analysis showed an increased yield of hrsACE2, a secretory recombinant protein, by 2.06-fold. Hence, we can conclude that sponging-induced inhibition of miR-21 may lead to a growth rate increase that could be linked to increased CHO cell productivity. For industrial cell lines, including CHO cells, an increase in productivity is crucial. The results of our research indicate that CM21D is an auspicious CHO engineering approach. KEY POINTS: • CHO is an ideal host cell line for producing industrial therapeutics manufacturing, and miR-21 is downregulated in CHO cells, which produce recombinant proteins. • The miR-21 target genes noticed a significant increase in expression upon the miR-21 sponging through CM21D. Additionally, sponging of miR-21 by CM21D enhanced the growth rate of CHO cells. • Productivity and growth rate were increased in CHO cells expressing recombinant hrs-ACE2 protein after CM21D knocking in.


Sujet(s)
Systèmes CRISPR-Cas , Cricetulus , microARN , Cellules CHO , Animaux , microARN/génétique , microARN/métabolisme , Ingénierie cellulaire/méthodes , Édition de gène/méthodes , Protéines recombinantes/génétique , Protéines recombinantes/métabolisme , Recombinases/génétique , Recombinases/métabolisme , Cricetinae
3.
Nature ; 631(8019): 37-48, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38961155

RÉSUMÉ

Living systems contain a vast network of metabolic reactions, providing a wealth of enzymes and cells as potential biocatalysts for chemical processes. The properties of protein and cell biocatalysts-high selectivity, the ability to control reaction sequence and operation in environmentally benign conditions-offer approaches to produce molecules at high efficiency while lowering the cost and environmental impact of industrial chemistry. Furthermore, biocatalysis offers the opportunity to generate chemical structures and functions that may be inaccessible to chemical synthesis. Here we consider developments in enzymes, biosynthetic pathways and cellular engineering that enable their use in catalysis for new chemistry and beyond.


Sujet(s)
Biocatalyse , Voies de biosynthèse , Ingénierie cellulaire , Enzymes , Humains , Ingénierie cellulaire/méthodes , Enzymes/métabolisme , Enzymes/composition chimique , Spécificité du substrat , Techniques de chimie synthétique
4.
Biomed Pharmacother ; 177: 117064, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38964179

RÉSUMÉ

Macrophages play a critical role in the body's defense against cancer by phagocytosing tumor cells, presenting antigens, and activating adaptive T cells. However, macrophages are intrinsically incapable of delivering targeted cancer immunotherapies. Engineered adoptive cell therapy introduces new targeting and antitumor capabilities by modifying macrophages to enhance the innate immune response of cells and improve clinical efficacy. In this study, we developed engineered macrophage cholesterol-AS1411-M1 (CAM1) for cellular immunotherapy. To target macrophages, cholesterol-AS1411 aptamers were anchored to the surface of M1 macrophages to produce CAM1 without genetic modification or cell damage. CAM1 induced significantly higher apoptosis/mortality than unmodified M1 macrophages in murine breast cancer cells. Anchoring AS1411 on the surface of macrophages provided a novel approach to construct engineered macrophages for tumor immunotherapy.


Sujet(s)
Aptamères nucléotidiques , Immunothérapie adoptive , Macrophages , Animaux , Macrophages/immunologie , Macrophages/métabolisme , Immunothérapie adoptive/méthodes , Souris , Lignée cellulaire tumorale , Cholestérol/métabolisme , Femelle , Apoptose , Ingénierie cellulaire/méthodes , Membrane cellulaire/métabolisme , Humains
5.
Methods Mol Biol ; 2844: 85-96, 2024.
Article de Anglais | MEDLINE | ID: mdl-39068333

RÉSUMÉ

Automated high-throughput methods that support tracking of mammalian cell growth are currently needed to advance cell line characterization and identification of desired genetic components required for cell engineering. Here, we describe a high-throughput noninvasive assay based on plate reader measurements. The assay relies on the change in absorbance of the pH indicator phenol red. We show that its basic and acidic absorbance profiles can be converted into a cell growth index consistent with cell count profiles, and that, by adopting a computational pipeline and calibration measurements, it is possible to identify a conversion that enables prediction of cell numbers from plate measurements alone. The assay is suitable for growth characterization of both suspension and adherent cell lines when these are grown under different environmental conditions and treated with chemotherapeutic drugs. The method also supports characterization of stably engineered cell lines and identification of desired promoters based on fluorescence output.


Sujet(s)
Prolifération cellulaire , Régions promotrices (génétique) , Animaux , Humains , Ingénierie cellulaire/méthodes , Phénolsulfonephtaléine , Lignée cellulaire , Tests de criblage à haut débit/méthodes , Techniques de culture cellulaire/méthodes , Concentration en ions d'hydrogène
6.
Nat Commun ; 15(1): 6200, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-39043686

RÉSUMÉ

Cell fate is likely regulated by a common machinery, while components of this machine remain to be identified. Here we report the design and testing of engineered cell fate controller NanogBiD, fusing BiD or BRG1 interacting domain of SS18 with Nanog. NanogBiD promotes mouse somatic cell reprogramming efficiently in contrast to the ineffective native protein under multiple testing conditions. Mechanistic studies further reveal that it facilitates cell fate transition by recruiting the intended Brg/Brahma-associated factor (BAF) complex to modulate chromatin accessibility and reorganize cell state specific enhancers known to be occupied by canonical Nanog, resulting in precocious activation of multiple genes including Sall4, miR-302, Dppa5a and Sox15 towards pluripotency. Although we have yet to test our approach in other species, our findings suggest that engineered chromatin regulators may provide much needed tools to engineer cell fate in the cells as drugs era.


Sujet(s)
Protéine homéotique Nanog , Facteurs de transcription , Animaux , Souris , Facteurs de transcription/métabolisme , Facteurs de transcription/génétique , Protéine homéotique Nanog/métabolisme , Protéine homéotique Nanog/génétique , Reprogrammation cellulaire/génétique , Chromatine/métabolisme , Chromatine/génétique , Helicase/métabolisme , Helicase/génétique , Différenciation cellulaire , Ingénierie cellulaire/méthodes , Protéines nucléaires/métabolisme , Protéines nucléaires/génétique
7.
Biomater Sci ; 12(14): 3500-3521, 2024 Jul 09.
Article de Anglais | MEDLINE | ID: mdl-38828621

RÉSUMÉ

Exosomes exhibit high bioavailability, biological stability, targeted specificity, low toxicity, and low immunogenicity in shuttling various bioactive molecules such as proteins, lipids, RNA, and DNA. Natural exosomes, however, have limited production, targeting abilities, and therapeutic efficacy in clinical trials. On the other hand, engineered exosomes have demonstrated long-term circulation, high stability, targeted delivery, and efficient intracellular drug release, garnering significant attention. The engineered exosomes bring new insights into developing next-generation drug delivery systems and show enormous potential in therapeutic applications, such as tumor therapies, diabetes management, cardiovascular disease, and tissue regeneration and repair. In this review, we provide an overview of recent advancements associated with engineered exosomes by focusing on the state-of-the-art strategies for cell engineering and exosome engineering. Exosome isolation methods, including traditional and emerging approaches, are systematically compared along with advancements in characterization methods. Current challenges and future opportunities are further discussed in terms of the preparation and application of engineered exosomes.


Sujet(s)
Exosomes , Exosomes/composition chimique , Exosomes/métabolisme , Humains , Animaux , Systèmes de délivrance de médicaments , Ingénierie cellulaire
8.
Sci Rep ; 14(1): 14141, 2024 06 19.
Article de Anglais | MEDLINE | ID: mdl-38898154

RÉSUMÉ

Secretion levels required of industrial Chinese hamster ovary (CHO) cell lines can challenge endoplasmic reticulum (ER) homeostasis, and ER stress caused by accumulation of misfolded proteins can be a bottleneck in biomanufacturing. The unfolded protein response (UPR) is initiated to restore homeostasis in response to ER stress, and optimization of the UPR can improve CHO cell production of therapeutic proteins. We compared the fed-batch growth, production characteristics, and transcriptomic response of an immunoglobulin G1 (IgG1) producer to its parental, non-producing host cell line. We conducted differential gene expression analysis using high throughput RNA sequencing (RNASeq) and quantitative polymerase chain reaction (qPCR) to study the ER stress response of each cell line during fed-batch culture. The UPR was activated in the IgG1 producer compared to the host cell line and our analysis of differential expression profiles indicated transient upregulation of ATF6α target mRNAs in the IgG1 producer, suggesting two upstream regulators of the ATF6 arm of the UPR, ATF6ß and WFS1, are rational engineering targets. Although both ATF6ß and WFS1 have been reported to negatively regulate ATF6α, this study shows knockdown of either target elicits different effects in an IgG1-producing CHO cell line. Stable knockdown of ATF6ß decreased cell growth without decreasing titer; however, knockdown of WFS1 decreased titer without affecting growth. Relative expression measured by qPCR indicated no direct relationship between ATF6ß and WFS1 expression, but upregulation of WFS1 in one pool was correlated with decreased growth and upregulation of ER chaperone mRNAs. While knockdown of WFS1 had negative impacts on UPR activation and product mRNA expression, knockdown of ATF6ß improved the UPR specifically later in fed-batch leading to increased overall productivity.


Sujet(s)
Facteur de transcription ATF-6 , Cricetulus , Immunoglobuline G , Réponse aux protéines mal repliées , Animaux , Cellules CHO , Facteur de transcription ATF-6/métabolisme , Facteur de transcription ATF-6/génétique , Immunoglobuline G/génétique , Immunoglobuline G/métabolisme , Réponse aux protéines mal repliées/génétique , Stress du réticulum endoplasmique/génétique , Techniques de knock-down de gènes , Ingénierie cellulaire/méthodes , Techniques de culture cellulaire en batch/méthodes , Protéines membranaires/métabolisme , Protéines membranaires/génétique
9.
Biotechnol Bioeng ; 121(9): 2907-2923, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-38924052

RÉSUMÉ

Continuously secreted by all cell types, extracellular vesicles (EVs) are small membrane-bound structures which shuttle bioactive cargo between cells across their external environment. Their central role as natural molecular messengers and ability to cross biological barriers has garnered significant attention in the use of EVs as therapeutic delivery vehicles. Still, harnessing the potential of EVs is faced with many obstacles. A cell line engineering approach can be used to exploit EVs to encapsulate a bespoke cargo of interest. However, full details regarding native EV-loading mechanisms remain under debate, making this a challenge. While Chinese hamster ovary (CHO) cells are well known to be the preferred host for recombinant therapeutic protein production, their application as an EV producer cell host has been largely overlooked. In this study, we engineered CHO DG44 cells to produce custom EVs with bespoke cargo. To this end, genetic constructs employing split green fluorescent protein technology were designed for tagging both CD81 and protein cargoes to enable EV loading via self-assembling activity. To demonstrate this, NanoLuc and mCherry were used as model reporter cargoes to validate engineered loading into EVs. Experimental findings indicated that our custom EV approach produced vesicles with up to 15-fold greater cargo compared with commonly used passive loading strategies. When applied to recipient cells, we observed a dose-dependent increase in cargo activity, suggesting successful delivery of engineered cargo via our custom CHO EVs.


Sujet(s)
Cricetulus , Vésicules extracellulaires , Animaux , Cellules CHO , Vésicules extracellulaires/métabolisme , Vésicules extracellulaires/composition chimique , Vésicules extracellulaires/génétique , Cricetinae , Ingénierie cellulaire/méthodes , Protéines recombinantes/génétique , Protéines recombinantes/métabolisme , Protéines recombinantes/biosynthèse
10.
Biomaterials ; 311: 122667, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38878480

RÉSUMÉ

Mesenchymal stem cells (MSCs) have garnered attention for their regenerative and immunomodulatory capabilities in clinical trials for various diseases. However, the effectiveness of MSC-based therapies, especially for conditions like graft-versus-host disease (GvHD), remains uncertain. The cytokine interferon (IFN)-γ has been known to enhance the immunosuppressive properties of MSCs through cell-to-cell interactions and soluble factors. In this study, we observed that IFN-γ-treated MSCs upregulated the expression of carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1), associated with immune evasion through the inhibition of natural killer (NK) cell cytotoxicity. To co-opt this immunomodulatory function, we generated MSCs overexpressing CEACAM1 and found that CEACAM1-engineered MSCs significantly reduced NK cell activation and cytotoxicity via cell-to-cell interaction, independent of NKG2D ligand regulation. Furthermore, CEACAM1-engineered MSCs effectively inhibited the proliferation and activation of T cells along with the inflammatory responses of monocytes. In a humanized GvHD mouse model, CEACAM1-MSCs, particularly CEACAM1-4S-MSCs, demonstrated therapeutic potential by improving survival and alleviating symptoms. These findings suggest that CEACAM1 expression on MSCs contributes to MSC-mediated regulation of immune responses and that CEACAM1-engineered MSC could have therapeutic potential in conditions involving immune dysregulation.


Sujet(s)
Antigènes CD , Molécules d'adhérence cellulaire , Communication cellulaire , Maladie du greffon contre l'hôte , Cellules tueuses naturelles , Cellules souches mésenchymateuses , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/cytologie , Animaux , Antigènes CD/métabolisme , Humains , Maladie du greffon contre l'hôte/immunologie , Maladie du greffon contre l'hôte/thérapie , Molécules d'adhérence cellulaire/métabolisme , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Immunomodulation , Souris , Interféron gamma/métabolisme , Transplantation de cellules souches mésenchymateuses , Prolifération cellulaire/effets des médicaments et des substances chimiques , Ingénierie cellulaire/méthodes
11.
Mol Cell Proteomics ; 23(7): 100796, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38851451

RÉSUMÉ

Protein O-linked mannose (O-Man) glycosylation is an evolutionary conserved posttranslational modification that fulfills important biological roles during embryonic development. Three nonredundant enzyme families, POMT1/POMT2, TMTC1-4, and TMEM260, selectively coordinate the initiation of protein O-Man glycosylation on distinct classes of transmembrane proteins, including α-dystroglycan, cadherins, and plexin receptors. However, a systematic investigation of their substrate specificities is lacking, in part due to the ubiquitous expression of O-Man glycosyltransferases in cells, which precludes analysis of pathway-specific O-Man glycosylation on a proteome-wide scale. Here, we apply a targeted workflow for membrane glycoproteomics across five human cell lines to extensively map O-Man substrates and genetically deconstruct O-Man initiation by individual and combinatorial knockout of O-Man glycosyltransferase genes. We established a human cell library for the analysis of substrate specificities of individual O-Man initiation pathways by quantitative glycoproteomics. Our results identify 180 O-Man glycoproteins, demonstrate new protein targets for the POMT1/POMT2 pathway, and show that TMTC1-4 and TMEM260 pathways widely target distinct Ig-like protein domains of plasma membrane proteins involved in cell-cell and cell-extracellular matrix interactions. The identification of O-Man on Ig-like folds adds further knowledge on the emerging concept of domain-specific O-Man glycosylation which opens for functional studies of O-Man-glycosylated adhesion molecules and receptors.


Sujet(s)
Mannose , Humains , Glycosylation , Mannose/métabolisme , Spécificité du substrat , Glycoprotéines/métabolisme , Protéomique/méthodes , Lignée cellulaire , Glycosyltransferase/métabolisme , Glycosyltransferase/génétique , Maturation post-traductionnelle des protéines , Ingénierie cellulaire/méthodes
12.
Sci Transl Med ; 16(749): eadg9814, 2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38809963

RÉSUMÉ

T cell-based cancer immunotherapy has typically relied on membrane-bound cytotoxicity enhancers such as chimeric antigen receptors expressed in autologous αß T cells. These approaches are limited by tonic signaling of synthetic constructs and costs associated with manufacturing. γδ T cells are an emerging alternative for cellular therapy, having innate antitumor activity, potent antibody-dependent cellular cytotoxicity, and minimal alloreactivity. We present an immunotherapeutic platform technology built around the innate properties of the Vγ9Vδ2 T cell, harnessing specific characteristics of this cell type and offering an allocompatible cellular therapy that recruits bystander immunity. We engineered γδ T cells to secrete synthetic tumor-targeting opsonins in the form of an scFv-Fc fusion protein and a mitogenic IL-15Rα-IL-15 fusion protein (stIL15). Using GD2 as a model antigen, we show that GD2-specific opsonin-secreting Vγ9Vδ2 T cells (stIL15-OPS-γδ T cells) have enhanced cytotoxicity and promote bystander activity of other lymphoid and myeloid cells. Secretion of stIL-15 abrogated the need for exogenous cytokine supplementation and further mediated activation of bystander natural killer cells. Compared with unmodified γδ T cells, stIL15-OPS-γδ T cells exhibited superior in vivo control of subcutaneous tumors and persistence in the blood. Moreover, stIL15-OPS-γδ T cells were efficacious against patient-derived osteosarcomas in animal models and in vitro, where efficacy could be boosted with the addition of zoledronic acid. Together, the data identify stIL15-OPS-γδ T cells as a candidate allogeneic cell therapy platform combining direct cytolysis with bystander activation to promote tumor control.


Sujet(s)
Ostéosarcome , Récepteur lymphocytaire T antigène, gamma-delta , Animaux , Ostéosarcome/thérapie , Ostéosarcome/immunologie , Ostéosarcome/anatomopathologie , Humains , Récepteur lymphocytaire T antigène, gamma-delta/métabolisme , Récepteur lymphocytaire T antigène, gamma-delta/immunologie , Lignée cellulaire tumorale , Cytotoxicité immunologique , Souris , Lymphocytes T/immunologie , Acide zolédronique/pharmacologie , Effet bystander , Interleukine-15 , Ingénierie cellulaire
13.
Acc Chem Res ; 57(12): 1722-1735, 2024 Jun 18.
Article de Anglais | MEDLINE | ID: mdl-38819691

RÉSUMÉ

ConspectusIn human cells, intracellular access and therapeutic cargo transport, including gene-editing tools (e.g., CRISPR-Cas9 and transposons), nucleic acids (e.g., DNA, mRNA, and siRNA), peptides, and proteins (e.g., enzymes and antibodies), are tightly constrained to ensure healthy cell function and behavior. This principle is exemplified in the delivery mechanisms of chimeric antigen receptor (CAR)-T cells for ex-vivo immunotherapy. In particular, the clinical success of CAR-T cells has established a new standard of care by curing previously incurable blood cancers. The approach involves the delivery, typically via the use of electroporation (EP) and lentivirus, of therapeutic CAR genes into a patient's own T cells, which are then engineered to express CARs that target and combat their blood cancer. But the key difficulty lies in genetically manipulating these cells without causing irreversible damage or loss of function─all the while minimizing complexities of manufacturing, safety concerns, and costs, and ensuring the efficacy of the final CAR-T cell product.Nanoinjection─the process of intracellular delivery using nanoneedles (NNs)─is an emerging physical delivery route that efficiently negotiates the plasma membrane of many cell types, including primary human T cells. It occurs with minimal perturbation, invasiveness, and toxicity, with high efficiency and throughput at high spatial and temporal resolutions. Nanoinjection promises greatly improved delivery of a broad range of therapeutic cargos with little or no damage to those cargos. A nanoinjection platform allows these cargos to function in the intracellular space as desired. The adaptability of nanoinjection platforms is now bringing major advantages in immunomodulation, mechanotransduction, sampling of cell states (nanobiopsy), controlled intracellular interrogation, and the primary focus of this account─intracellular delivery and its applications in ex vivo cell engineering.Mechanical nanoinjection typically exerts direct mechanical force on the cell membrane, offering a straightforward route to improve membrane perturbation by the NNs and subsequent transport of genetic cargo into targeted cell type (adherent or suspension cells). By contrast, electroactive nanoinjection is controlled by coupling NNs with an electric field─a new route for activating electroporation (EP) at the nanoscale─allowing a dramatic reduction of the applied voltage to a cell and so minimizing post-EP damage to cells and cargo, and overcoming many of the limitations of conventional bulk EP. Nanoinjection transcends mere technique; it is an approach to cell engineering ex vivo, offering the potential to endow cells with new, powerful features such as generating chimeric antigen receptor (CAR)-T cells for future CAR-T cell technologies.We first discuss the manufacturing of NN devices (Section 2), then delve into nanoinjection-mediated cell engineering (Section 3), nanoinjection mechanisms and interfacing methodologies (Section 4), and emerging applications in using nanoinjection to create functional CAR-T cells (Section 5).


Sujet(s)
Ingénierie cellulaire , Humains , Ingénierie cellulaire/méthodes , Récepteurs chimériques pour l'antigène/métabolisme , Nanotechnologie/méthodes , Lymphocytes T/cytologie , Lymphocytes T/métabolisme , Électroporation/méthodes , Injections
14.
Immunol Cell Biol ; 102(6): 444-447, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38693888

RÉSUMÉ

Natural killer (NK) cells possess potent cytotoxicity against infected and cancerous cells and hold promise in the development of new immunotherapies. This article for the Highlights of 2023 Series focuses on current advances in NK cell biology in cancerous and infectious settings and highlights opportunities for therapeutic interventions, including engineered NK cell therapies and advancements in feeder cell technologies.


Sujet(s)
Ingénierie cellulaire , Immunothérapie , Cellules tueuses naturelles , Tumeurs , Animaux , Humains , Cytotoxicité immunologique , Immunothérapie/méthodes , Immunothérapie adoptive/méthodes , Cellules tueuses naturelles/immunologie , Tumeurs/thérapie , Tumeurs/immunologie
15.
Acta Biochim Pol ; 71: 12185, 2024.
Article de Anglais | MEDLINE | ID: mdl-38721308

RÉSUMÉ

Human chemokine receptor 8 (CCR8) is a promising drug target for immunotherapy of cancer and autoimmune diseases. Monoclonal antibody-based CCR8 targeted treatment shows significant inhibition in tumor growth. The inhibition of CCR8 results in the improvement of antitumor immunity and patient survival rates by regulating tumor-resident regulatory T cells. Recently monoclonal antibody drug development targeting CCR8 has become a research hotspot, which also promotes the advancement of antibody evaluation methods. Therefore, we constructed a novel engineered customized cell line HEK293-cAMP-biosensor-CCR8 combined with CCR8 and a cAMP-biosensor reporter. It can be used for the detection of anti-CCR8 antibody functions like specificity and biological activity, in addition to the detection of antibody-dependent cell-mediated cytotoxicity and antibody-dependent-cellular-phagocytosis. We obtained a new CCR8 mAb 22H9 and successfully verified its biological activities with HEK293-cAMP-biosensor-CCR8. Our reporter cell line has high sensitivity and specificity, and also offers a rapid kinetic detection platform for evaluating anti-CCR8 antibody functions.


Sujet(s)
Anticorps monoclonaux , Techniques de biocapteur , AMP cyclique , Récepteurs CCR8 , Humains , Cellules HEK293 , Anticorps monoclonaux/immunologie , Anticorps monoclonaux/pharmacologie , Récepteurs CCR8/immunologie , Récepteurs CCR8/métabolisme , AMP cyclique/métabolisme , Techniques de biocapteur/méthodes , Cytotoxicité à médiation cellulaire dépendante des anticorps/immunologie , Ingénierie cellulaire/méthodes
16.
Adv Protein Chem Struct Biol ; 140: 91-156, 2024.
Article de Anglais | MEDLINE | ID: mdl-38762281

RÉSUMÉ

This book chapter highlights a comprehensive exploration of the transformative innovations in the field of cancer immunotherapy. CAR (Chimeric Antigen Receptor) T-cell therapy represents a groundbreaking approach to treat cancer by reprogramming a patient immune cells to recognize and destroy cancer cells. This chapter underscores the critical role of synthetic biology in enhancing the safety and effectiveness of CAR T-cell therapies. It begins by emphasizing the growing importance of personalized medicine in cancer treatment, emphasizing the shift from one-size-fits-all approaches to patient-specific solutions. Synthetic biology, a multidisciplinary field, has been instrumental in customizing CAR T-cell therapies, allowing for fine-tuned precision and minimizing unwanted side effects. The chapter highlights recent advances in gene editing, synthetic gene circuits, and molecular engineering, showcasing how these technologies are optimizing CAR T-cell function. In summary, this book chapter sheds light on the remarkable progress made in the development of CAR T-cell therapies using synthetic biology, providing hope for cancer patients and hinting at a future where highly personalized and effective cancer treatments are the norm.


Sujet(s)
Tumeurs , Récepteurs chimériques pour l'antigène , Biologie synthétique , Humains , Tumeurs/thérapie , Tumeurs/immunologie , Tumeurs/génétique , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/génétique , Immunothérapie adoptive/méthodes , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Édition de gène , Ingénierie cellulaire
17.
Mol Ther ; 32(7): 2357-2372, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38751112

RÉSUMÉ

Natural killer (NK) cells have high intrinsic cytotoxic capacity, and clinical trials have demonstrated their safety and efficacy for adoptive cancer therapy. Expression of chimeric antigen receptors (CARs) enhances NK cell target specificity, with these cells applicable as off-the-shelf products generated from allogeneic donors. Here, we present for the first time an innovative approach for CAR NK cell engineering employing a non-viral Sleeping Beauty (SB) transposon/transposase-based system and minimized DNA vectors termed minicircles. SB-modified peripheral blood-derived primary NK cells displayed high and stable CAR expression and more frequent vector integration into genomic safe harbors than lentiviral vectors. Importantly, SB-generated CAR NK cells demonstrated enhanced cytotoxicity compared with non-transfected NK cells. A strong antileukemic potential was confirmed using established acute lymphocytic leukemia cells and patient-derived primary acute B cell leukemia and lymphoma samples as targets in vitro and in vivo in a xenograft leukemia mouse model. Our data suggest that the SB-transposon system is an efficient, safe, and cost-effective approach to non-viral engineering of highly functional CAR NK cells, which may be suitable for cancer immunotherapy of leukemia as well as many other malignancies.


Sujet(s)
Vecteurs génétiques , Immunothérapie adoptive , Cellules tueuses naturelles , Récepteurs chimériques pour l'antigène , Humains , Cellules tueuses naturelles/immunologie , Cellules tueuses naturelles/métabolisme , Animaux , Souris , Vecteurs génétiques/génétique , Récepteurs chimériques pour l'antigène/génétique , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/métabolisme , Immunothérapie adoptive/méthodes , Tests d'activité antitumorale sur modèle de xénogreffe , Transposases/génétique , Transposases/métabolisme , Lignée cellulaire tumorale , Éléments transposables d'ADN , Cytotoxicité immunologique , Leucémie-lymphome lymphoblastique à précurseurs B et T/thérapie , Leucémie-lymphome lymphoblastique à précurseurs B et T/génétique , Leucémie-lymphome lymphoblastique à précurseurs B et T/immunologie , Ingénierie cellulaire/méthodes
18.
Front Immunol ; 15: 1360237, 2024.
Article de Anglais | MEDLINE | ID: mdl-38576617

RÉSUMÉ

Comprising only 1-10% of the circulating T cell population, γδT cells play a pivotal role in cancer immunotherapy due to their unique amalgamation of innate and adaptive immune features. These cells can secrete cytokines, including interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α), and can directly eliminate tumor cells through mechanisms like Fas/FasL and antibody-dependent cell-mediated cytotoxicity (ADCC). Unlike conventional αßT cells, γδT cells can target a wide variety of cancer cells independently of major histocompatibility complex (MHC) presentation and function as antigen-presenting cells (APCs). Their ability of recognizing antigens in a non-MHC restricted manner makes them an ideal candidate for allogeneic immunotherapy. Additionally, γδT cells exhibit specific tissue tropism, and rapid responsiveness upon reaching cellular targets, indicating a high level of cellular precision and adaptability. Despite these capabilities, the therapeutic potential of γδT cells has been hindered by some limitations, including their restricted abundance, unsatisfactory expansion, limited persistence, and complex biology and plasticity. To address these issues, gene-engineering strategies like the use of chimeric antigen receptor (CAR) T therapy, T cell receptor (TCR) gene transfer, and the combination with γδT cell engagers are being explored. This review will outline the progress in various engineering strategies, discuss their implications and challenges that lie ahead, and the future directions for engineered γδT cells in both monotherapy and combination immunotherapy.


Sujet(s)
Tumeurs , Récepteur lymphocytaire T antigène, gamma-delta , Récepteur lymphocytaire T antigène, gamma-delta/génétique , Lymphocytes T , Immunothérapie , Immunothérapie adoptive , Ingénierie cellulaire , Tumeurs/thérapie
19.
Trends Pharmacol Sci ; 45(5): 406-418, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38614815

RÉSUMÉ

T cells modified to express intelligently designed chimeric antigen receptors (CARs) are exceptionally powerful therapeutic agents for relapsed and refractory blood cancers and have the potential to revolutionize therapy for many other diseases. To circumvent the complexity and cost associated with broad-scale implementation of ex vivo manufactured adoptive cell therapy products, alternative strategies to generate CAR T cells in vivo by direct infusion of nanoparticle-formulated nucleic acids or engineered viral vectors under development have received a great deal of attention in the past few years. Here, we outline the ex vivo manufacturing process as a motivating framework for direct in vivo strategies and discuss emerging data from preclinical models to highlight the potency of the in vivo approach, the applicability for new disease indications, and the remaining challenges associated with clinical readiness, including delivery specificity, long term efficacy, and safety.


Sujet(s)
Immunothérapie adoptive , Récepteurs chimériques pour l'antigène , Lymphocytes T , Humains , Animaux , Lymphocytes T/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Immunothérapie adoptive/méthodes , Ingénierie cellulaire/méthodes , Récepteurs aux antigènes des cellules T/immunologie , Tumeurs/thérapie , Tumeurs/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE