Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Mol Ther ; 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39169622

ABSTRACT

Adoptive cell therapy using chimeric antigen receptor (CAR) T cells has proven to be lifesaving for many cancer patients. However, its therapeutic efficacy has been limited in solid tumors. One key factor for this is cancer-associated fibroblasts (CAFs) that modulate the tumor microenvironment (TME) to inhibit T cell infiltration and induce "T cell dysfunction." Additionally, the sparsity of tumor-specific antigens (TSA) and expression of CAR-directed tumor-associated antigens (TAA) on normal tissues often results in "on-target off-tumor" cytotoxicity, raising safety concerns. Using TALEN-mediated gene editing, we present here an innovative CAR T cell engineering strategy to overcome these challenges. Our allogeneic "Smart CAR T cells" are designed to express a constitutive CAR, targeting FAP+ CAFs in solid tumors. Additionally, a second CAR targeting a TAA such as mesothelin is specifically integrated at a TCR signaling-inducible locus like PDCD1. FAPCAR-mediated CAF targeting induces expression of the mesothelin CAR, establishing an IF/THEN-gated circuit sensitive to dual antigen sensing. Using this approach, we observe enhanced anti-tumor cytotoxicity, while limiting "on-target off-tumor" toxicity. Our study thus demonstrates TALEN-mediated gene editing capabilities for design of allogeneic IF/THEN-gated dual CAR T cells that efficiently target immunotherapy-recalcitrant solid tumors while mitigating potential safety risks, encouraging clinical development of this strategy.

2.
Mol Ther ; 32(6): 1643-1657, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38582963

ABSTRACT

Gene therapy in hematopoietic stem and progenitor cells (HSPCs) shows great potential for the treatment of inborn metabolic diseases. Typical HSPC gene therapy approaches rely on constitutive promoters to express a therapeutic transgene, which is associated with multiple disadvantages. Here, we propose a novel promoterless intronic gene editing approach that triggers transgene expression only after cellular differentiation into the myeloid lineage. We integrated a splicing-competent eGFP cassette into the first intron of CD11b and observed expression of eGFP in the myeloid lineage but minimal to no expression in HSPCs or differentiated non-myeloid lineages. In vivo, edited HSPCs successfully engrafted in immunodeficient mice and displayed transgene expression in the myeloid compartment of multiple tissues. Using the same approach, we expressed alpha-L-iduronidase (IDUA), the defective enzyme in Mucopolysaccharidosis type I, and observed a 10-fold supraendogenous IDUA expression exclusively after myeloid differentiation. Edited cells efficiently populated bone marrow, blood, and spleen of immunodeficient mice, and retained the capacity to secrete IDUA ex vivo. Importantly, cells edited with the eGFP and IDUA transgenes were also found in the brain. This approach may unlock new therapeutic strategies for inborn metabolic and neurological diseases that require the delivery of therapeutics in brain.


Subject(s)
Gene Editing , Hematopoietic Stem Cells , Introns , Myeloid Cells , Transcription Activator-Like Effector Nucleases , Transgenes , Animals , Gene Editing/methods , Mice , Hematopoietic Stem Cells/metabolism , Humans , Myeloid Cells/metabolism , Transcription Activator-Like Effector Nucleases/genetics , Transcription Activator-Like Effector Nucleases/metabolism , Cell Differentiation/genetics , Genetic Therapy/methods , Iduronidase/genetics , Iduronidase/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Gene Expression , Cell Lineage/genetics , CD11b Antigen/genetics , CD11b Antigen/metabolism , Hematopoietic Stem Cell Transplantation/methods , Mucopolysaccharidosis I/therapy , Mucopolysaccharidosis I/genetics
3.
Mol Ther ; 27(6): 1126-1138, 2019 06 05.
Article in English | MEDLINE | ID: mdl-31005597

ABSTRACT

Clinical success of autologous CD19-directed chimeric antigen receptor T cells (CAR Ts) in acute lymphoblastic leukemia and non-Hodgkin lymphoma suggests that CAR Ts may be a promising therapy for hematological malignancies, including multiple myeloma. However, autologous CAR T therapies have limitations that may impact clinical use, including lengthy vein-to-vein time and manufacturing constraints. Allogeneic CAR T (AlloCAR T) therapies may overcome these innate limitations of autologous CAR T therapies. Unlike autologous cell therapies, AlloCAR T therapies employ healthy donor T cells that are isolated in a manufacturing facility, engineered to express CARs with specificity for a tumor-associated antigen, and modified using gene-editing technology to limit T cell receptor (TCR)-mediated immune responses. Here, transcription activator-like effector nuclease (TALEN) gene editing of B cell maturation antigen (BCMA) CAR Ts was used to confer lymphodepletion resistance and reduced graft-versus-host disease (GvHD) potential. The safety profile of allogeneic BCMA CAR Ts was further enhanced by incorporating a CD20 mimotope-based intra-CAR off switch enabling effective CAR T elimination in the presence of rituximab. Allogeneic BCMA CAR Ts induced sustained antitumor responses in mice supplemented with human cytokines, and, most importantly, maintained their phenotype and potency after scale-up manufacturing. This novel off-the-shelf allogeneic BCMA CAR T product is a promising candidate for clinical evaluation.


Subject(s)
B-Cell Maturation Antigen/immunology , Cell Transplantation/methods , Immunotherapy, Adoptive/methods , Multiple Myeloma/therapy , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Animals , Antineoplastic Agents, Immunological/therapeutic use , B-Cell Maturation Antigen/genetics , Blood Donors , Cell Line, Tumor , Cell Transplantation/adverse effects , Cytotoxicity, Immunologic/genetics , Gene Editing , Genetic Vectors , Graft vs Host Disease/therapy , Humans , Immunotherapy, Adoptive/adverse effects , Mice , Mice, Inbred NOD , Mice, SCID , Multiple Myeloma/pathology , Progression-Free Survival , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Rituximab/therapeutic use , T-Lymphocytes/metabolism , Transcription Activator-Like Effector Nucleases/genetics , Transduction, Genetic , Transplantation, Homologous/methods
4.
BMC Biotechnol ; 19(1): 44, 2019 07 03.
Article in English | MEDLINE | ID: mdl-31269942

ABSTRACT

BACKGROUND: Engineered therapeutic cells have attracted a great deal of interest due to their potential applications in treating a wide range of diseases, including cancer and autoimmunity. Chimeric antigen receptor (CAR) T-cells are designed to detect and kill tumor cells that present a specific, predefined antigen. The rapid expansion of targeted antigen beyond CD19, has highlighted new challenges, such as autoactivation and T-cell fratricide, that could impact the capacity to manufacture engineered CAR T-cells. Therefore, the development of strategies to control CAR expression at the surface of T-cells and their functions is under intense investigations. RESULTS: Here, we report the development and evaluation of an off-switch directly embedded within a CAR construct (SWIFF-CAR). The incorporation of a self-cleaving degradation moiety controlled by a protease/protease inhibitor pair allowed the ex vivo tight and reversible control of the CAR surface presentation and the subsequent CAR-induced signaling and cytolytic functions of the engineered T-cells using the cell permeable Asunaprevir (ASN) small molecule. CONCLUSIONS: The strategy described in this study could, in principle, be broadly adapted to CAR T-cells development to circumvent some of the possible hurdle of CAR T-cell manufacturing. This system essentially creates a CAR T-cell with an integrated functional rheostat.


Subject(s)
Antigens, CD19/immunology , Gene Expression/immunology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Antigens, CD19/genetics , Antigens, CD19/metabolism , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Gene Expression/drug effects , Gene Expression/genetics , Humans , Isoquinolines/pharmacology , Protease Inhibitors/pharmacology , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Sulfonamides/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism
5.
Malar J ; 15: 28, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26772184

ABSTRACT

BACKGROUND: Rosetting, namely the capacity of the Plasmodium falciparum-infected red blood cells to bind uninfected RBCs, is commonly observed in African children with severe malaria. Rosetting results from specific interactions between a subset of variant P. falciparum erythrocyte membrane protein 1 (PfEMP1) adhesins encoded by var genes, serum components and RBC receptors. Rosette formation is a redundant phenotype, as there exists more than one var gene encoding a rosette-mediating PfEMP1 in each genome and hence a diverse array of underlying interactions. Moreover, field diversity creates a large panel of rosetting-associated serotypes and studies with human immune sera indicate that surface-reacting antibodies are essentially variant-specific. To gain better insight into the interactions involved in rosetting and map surface epitopes, a panel of monoclonal antibodies (mAbs) was investigated. METHODS: Monoclonal antibodies were isolated from mice immunized with PfEMP1-VarO recombinant domains. They were characterized using ELISA and reactivity with the native PfEMP1-VarO adhesin on immunoblots of reduced and unreduced extracts, as well as SDS-extracts of Palo Alto 89F5 VarO schizonts. Functionality was assessed using inhibition of Palo Alto 89F5 VarO rosette formation and disruption of Palo Alto 89F5 VarO rosettes. Competition ELISAs were performed with biotinylated antibodies against DBL1 to identify reactivity groups. Specificity of mAbs reacting with the DBL1 adhesion domain was explored using recombinant proteins carrying mutations abolishing RBC binding or binding to heparin, a potent inhibitor of rosette formation. RESULTS: Domain-specific, surface-reacting mAbs were obtained for four individual domains (DBL1, CIDR1, DBL2, DBL4). Monoclonal antibodies reacting with DBL1 potently inhibited the formation of rosettes and disrupted Palo Alto 89F5 VarO rosettes. Most surface-reactive mAbs and all mAbs interfering with rosetting reacted on parasite immunoblots with disulfide bond-dependent PfEMP1 epitopes. Based on competition ELISA and binding to mutant DBL1 domains, two distinct binding sites for rosette-disrupting mAbs were identified in close proximity to the RBC-binding site. CONCLUSIONS: Rosette-inhibitory antibodies bind to conformation-dependent epitopes located close to the RBC-binding site and distant from the heparin-binding site. These results provide novel clues for a rational intervention strategy that targets rosetting.


Subject(s)
Antibodies, Monoclonal/metabolism , Cell Adhesion Molecules/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Enzyme-Linked Immunosorbent Assay , Mice , Plasmodium falciparum/drug effects , Protein Binding
6.
Mol Ther ; 23(9): 1507-18, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26061646

ABSTRACT

The adoptive transfer of chimeric antigen receptor (CAR) T cell represents a highly promising strategy to fight against multiple cancers. The clinical outcome of such therapies is intimately linked to the ability of effector cells to engraft, proliferate, and specifically kill tumor cells within patients. When allogeneic CAR T-cell infusion is considered, host versus graft and graft versus host reactions must be avoided to prevent rejection of adoptively transferred cells, host tissue damages and to elicit significant antitumoral outcome. This work proposes to address these three requirements through the development of multidrug-resistant T cell receptor αß-deficient CAR T cells. We demonstrate that these engineered T cells displayed efficient antitumor activity and proliferated in the presence of purine and pyrimidine nucleoside analogues, currently used in clinic as preconditioning lymphodepleting regimens. The absence of TCRαß at their cell surface along with their purine nucleotide analogues-resistance properties could prevent their alloreactivity and enable them to resist to lymphodepleting regimens that may be required to avoid their ablation via HvG reaction. By providing a basic framework to develop a universal T cell compatible with allogeneic adoptive transfer, this work is laying the foundation stone of the large-scale utilization of CAR T-cell immunotherapies.


Subject(s)
Cell- and Tissue-Based Therapy , Drug Resistance, Multiple/genetics , Immunotherapy, Adoptive , Receptors, Antigen, T-Cell/genetics , Recombinant Fusion Proteins/genetics , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Antigens, CD19/genetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Cell- and Tissue-Based Therapy/methods , Combined Modality Therapy , Cytotoxicity, Immunologic , Deoxycytidine Kinase/deficiency , Deoxycytidine Kinase/genetics , Gene Expression , Gene Silencing , Humans , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Inhibitory Concentration 50 , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphocyte Culture Test, Mixed , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/therapy , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell, alpha-beta/deficiency , Receptors, Antigen, T-Cell, alpha-beta/genetics , Recombinant Fusion Proteins/metabolism , T-Lymphocytes/drug effects , Transplantation, Homologous
7.
Nucleic Acids Res ; 42(8): 5390-402, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24569350

ABSTRACT

A key issue when designing and using DNA-targeting nucleases is specificity. Ideally, an optimal DNA-targeting tool has only one recognition site within a genomic sequence. In practice, however, almost all designer nucleases available today can accommodate one to several mutations within their target site. The ability to predict the specificity of targeting is thus highly desirable. Here, we describe the first comprehensive experimental study focused on the specificity of the four commonly used repeat variable diresidues (RVDs; NI:A, HD:C, NN:G and NG:T) incorporated in transcription activator-like effector nucleases (TALEN). The analysis of >15 500 unique TALEN/DNA cleavage profiles allowed us to monitor the specificity gradient of the RVDs along a TALEN/DNA binding array and to present a specificity scoring matrix for RVD/nucleotide association. Furthermore, we report that TALEN can only accommodate a relatively small number of position-dependent mismatches while maintaining a detectable activity at endogenous loci in vivo, demonstrating the high specificity of these molecular tools. We thus envision that the results we provide will allow for more deliberate choices of DNA binding arrays and/or DNA targets, extending our engineering capabilities.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Deoxyribonucleases/chemistry , Deoxyribonucleases/metabolism , Amino Acids/chemistry , Animals , Base Sequence , CHO Cells , Cricetinae , Cricetulus , DNA/chemistry , DNA/metabolism , DNA Cleavage , Mutation , Protein Array Analysis , Protein Engineering , Yeasts/genetics
8.
Methods ; 69(2): 151-70, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25047178

ABSTRACT

TALEN is one of the most widely used tools in the field of genome editing. It enables gene integration and gene inactivation in a highly efficient and specific fashion. Although very attractive, the apparent simplicity and high success rate of TALEN could be misleading for novices in the field of gene editing. Depending on the application, specific TALEN designs, activity assessments and screening strategies need to be adopted. Here we report different methods to efficiently perform TALEN-mediated gene integration and inactivation in different mammalian cell systems including induced pluripotent stem cells and delineate experimental examples associated with these approaches.


Subject(s)
Gene Targeting/methods , Genome/genetics , Transcriptional Activation/genetics , Transfection/methods , Animals , Base Sequence , Cell Line , DNA-Binding Proteins/genetics , HCT116 Cells , Humans , Molecular Sequence Data
9.
Acta Crystallogr D Biol Crystallogr ; 70(Pt 7): 2042-52, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25004980

ABSTRACT

DNA editing offers new possibilities in synthetic biology and biomedicine for modulation or modification of cellular functions to organisms. However, inaccuracy in this process may lead to genome damage. To address this important problem, a strategy allowing specific gene modification has been achieved through the addition, removal or exchange of DNA sequences using customized proteins and the endogenous DNA-repair machinery. Therefore, the engineering of specific protein-DNA interactions in protein scaffolds is key to providing `toolkits' for precise genome modification or regulation of gene expression. In a search for putative DNA-binding domains, BurrH, a protein that recognizes a 19 bp DNA target, was identified. Here, its apo and DNA-bound crystal structures are reported, revealing a central region containing 19 repeats of a helix-loop-helix modular domain (BurrH domain; BuD), which identifies the DNA target by a single residue-to-nucleotide code, thus facilitating its redesign for gene targeting. New DNA-binding specificities have been engineered in this template, showing that BuD-derived nucleases (BuDNs) induce high levels of gene targeting in a locus of the human haemoglobin ß (HBB) gene close to mutations responsible for sickle-cell anaemia. Hence, the unique combination of high efficiency and specificity of the BuD arrays can push forward diverse genome-modification approaches for cell or organism redesign, opening new avenues for gene editing.


Subject(s)
DNA/chemistry , Genome , Helix-Loop-Helix Motifs , Calorimetry , Crystallography, X-Ray , Humans
10.
BMC Mol Biol ; 15: 13, 2014 Jul 05.
Article in English | MEDLINE | ID: mdl-24997498

ABSTRACT

BACKGROUND: The past decade has seen the emergence of several molecular tools that render possible modification of cellular functions through accurate and easy addition, removal, or exchange of genomic DNA sequences. Among these technologies, transcription activator-like effectors (TALE) has turned out to be one of the most versatile and incredibly robust platform for generating targeted molecular tools as demonstrated by fusion to various domains such as transcription activator, repressor and nucleases. RESULTS: In this study, we generated a novel nuclease architecture based on the transcription activator-like effector scaffold. In contrast to the existing Tail to Tail (TtT) and head to Head (HtH) nuclease architectures based on the symmetrical association of two TALE DNA binding domains fused to the C-terminal (TtT) or N-terminal (HtH) end of FokI, this novel architecture consists of the asymmetrical association of two different engineered TALE DNA binding domains fused to the N- and C-terminal ends of FokI (TALE::FokI and FokI::TALE scaffolds respectively). The characterization of this novel Tail to Head (TtH) architecture in yeast enabled us to demonstrate its nuclease activity and define its optimal target configuration. We further showed that this architecture was able to promote substantial level of targeted mutagenesis at three endogenous loci present in two different mammalian cell lines. CONCLUSION: Our results demonstrated that this novel functional TtH architecture which requires binding to only one DNA strand of a given endogenous locus has the potential to extend the targeting possibility of FokI-based TALE nucleases.


Subject(s)
Deoxyribonucleases, Type II Site-Specific/metabolism , Fungal Proteins/metabolism , Protein Engineering/methods , Recombinant Fusion Proteins/metabolism , Transcription Factors/metabolism , Yeasts/metabolism , Animals , Base Sequence , Binding Sites , Cell Line , DNA/metabolism , Deoxyribonucleases, Type II Site-Specific/chemistry , Deoxyribonucleases, Type II Site-Specific/genetics , Fungal Proteins/chemistry , Fungal Proteins/genetics , Gene Targeting/methods , Genetic Loci , Humans , Molecular Sequence Data , Mutagenesis , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Sequence Alignment , Transcription Factors/chemistry , Transcription Factors/genetics , Yeasts/genetics
11.
Plant Biotechnol J ; 12(7): 934-40, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24851712

ABSTRACT

Soybean oil is high in polyunsaturated fats and is often partially hydrogenated to increase its shelf life and improve oxidative stability. The trans-fatty acids produced through hydrogenation pose a health threat. Soybean lines that are low in polyunsaturated fats were generated by introducing mutations in two fatty acid desaturase 2 genes (FAD2-1A and FAD2-1B), which in the seed convert the monounsaturated fat, oleic acid, to the polyunsaturated fat, linoleic acid. Transcription activator-like effector nucleases (TALENs) were engineered to recognize and cleave conserved DNA sequences in both genes. In four of 19 transgenic soybean lines expressing the TALENs, mutations in FAD2-1A and FAD2-1B were observed in DNA extracted from leaf tissue; three of the four lines transmitted heritable FAD2-1 mutations to the next generation. The fatty acid profile of the seed was dramatically changed in plants homozygous for mutations in both FAD2-1A and FAD2-1B: oleic acid increased from 20% to 80% and linoleic acid decreased from 50% to under 4%. Further, mutant plants were identified that lacked the TALEN transgene and only carried the targeted mutations. The ability to create a valuable trait in a single generation through targeted modification of a gene family demonstrates the power of TALENs for genome engineering and crop improvement.


Subject(s)
Fatty Acid Desaturases/genetics , Glycine max/genetics , Plant Proteins/genetics , Soybean Oil/chemistry , Base Sequence , Fatty Acids/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Nutritive Value/genetics , Oleic Acid/metabolism , Plants, Genetically Modified/chemistry , Plants, Genetically Modified/metabolism , Seeds/enzymology , Seeds/genetics , Seeds/metabolism , Sequence Alignment , Glycine max/enzymology , Glycine max/metabolism
12.
PLoS Pathog ; 8(7): e1002781, 2012.
Article in English | MEDLINE | ID: mdl-22807674

ABSTRACT

The ABO blood group influences susceptibility to severe Plasmodium falciparum malaria. Recent evidence indicates that the protective effect of group O operates by virtue of reduced rosetting of infected red blood cells (iRBCs) with uninfected RBCs. Rosetting is mediated by a subgroup of PfEMP1 adhesins, with RBC binding being assigned to the N-terminal DBL1α1 domain. Here, we identify the ABO blood group as the main receptor for VarO rosetting, with a marked preference for group A over group B, which in turn is preferred to group O RBCs. We show that recombinant NTS-DBL1α1 and NTS-DBL1α1-CIDR1γ reproduce the VarO-iRBC blood group preference and document direct binding to blood group trisaccharides by surface plasmon resonance. More detailed RBC subgroup analysis showed preferred binding to group A1, weaker binding to groups A2 and B, and least binding to groups A(x) and O. The 2.8 Å resolution crystal structure of the PfEMP1-VarO Head region, NTS-DBL1α1-CIDR1γ, reveals extensive contacts between the DBL1α1 and CIDR1γ and shows that the NTS-DBL1α1 hinge region is essential for RBC binding. Computer docking of the blood group trisaccharides and subsequent site-directed mutagenesis localized the RBC-binding site to the face opposite to the heparin-binding site of NTS-DBLα1. RBC binding involves residues that are conserved between rosette-forming PfEMP1 adhesins, opening novel opportunities for intervention against severe malaria. By deciphering the structural basis of blood group preferences in rosetting, we provide a link between ABO blood grouppolymorphisms and rosette-forming adhesins, consistent with the selective role of falciparum malaria on human genetic makeup.


Subject(s)
ABO Blood-Group System/metabolism , Plasmodium falciparum/metabolism , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Rosette Formation , ABO Blood-Group System/immunology , Amino Acid Sequence , Antibodies, Protozoan/immunology , Binding Sites , Crystallography, X-Ray , Erythrocytes/immunology , Erythrocytes/metabolism , Humans , Immune Adherence Reaction , Malaria, Falciparum/blood , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Molecular Sequence Data , Mutagenesis, Site-Directed , Plasmodium falciparum/genetics , Plasmodium falciparum/ultrastructure , Protein Structure, Secondary , Protein Structure, Tertiary , Protozoan Proteins/genetics , Protozoan Proteins/immunology
13.
Proc Natl Acad Sci U S A ; 108(13): 5243-8, 2011 Mar 29.
Article in English | MEDLINE | ID: mdl-21402930

ABSTRACT

The human malaria parasite Plasmodium falciparum can cause infected red blood cells (iRBC) to form rosettes with uninfected RBC, a phenotype associated with severe malaria. Rosetting is mediated by a subset of the Plasmodium falciparum membrane protein 1 (PfEMP1) variant adhesins expressed on the infected host-cell surface. Heparin and other sulfated oligosaccharides, however, can disrupt rosettes, suggesting that therapeutic approaches to this form of severe malaria are feasible. We present a structural and functional study of the N-terminal domain of PfEMP1 from the VarO variant comprising the N-terminal segment (NTS) and the first DBL domain (DBL1α(1)), which is directly implicated in rosetting. We demonstrate that NTS-DBL1α(1)-VarO binds to RBC and that heparin inhibits this interaction in a dose-dependent manner, thus mimicking heparin-mediated rosette disruption. We have determined the crystal structure of NTS-DBL1α(1), showing that NTS, previously thought to be a structurally independent component of PfEMP1, forms an integral part of the DBL1α domain. Using mutagenesis and docking studies, we have located the heparin-binding site, which includes NTS. NTS, unique to the DBL α-class domain, is thus an intrinsic structural and functional component of the N-terminal VarO domain. The specific interaction observed with heparin opens the way for developing antirosetting therapeutic strategies.


Subject(s)
Erythrocytes/parasitology , Heparin/metabolism , Plasmodium falciparum/metabolism , Protein Structure, Tertiary , Protozoan Proteins/chemistry , Rosette Formation , Amino Acid Sequence , Crystallography, X-Ray , Humans , Models, Molecular , Molecular Sequence Data , Mutation , Plasmodium falciparum/pathogenicity , Protozoan Proteins/genetics , Protozoan Proteins/metabolism
14.
Sci Rep ; 14(1): 12870, 2024 06 04.
Article in English | MEDLINE | ID: mdl-38834632

ABSTRACT

One of the most recent advances in the genome editing field has been the addition of "TALE Base Editors", an innovative platform for cell therapy that relies on the deamination of cytidines within double strand DNA, leading to the formation of an uracil (U) intermediate. These molecular tools are fusions of transcription activator-like effector domains (TALE) for specific DNA sequence binding, split-DddA deaminase halves that will, upon catalytic domain reconstitution, initiate the conversion of a cytosine (C) to a thymine (T), and an uracil glycosylase inhibitor (UGI). We developed a high throughput screening strategy capable to probe key editing parameters in a precisely defined genomic context in cellulo, excluding or minimizing biases arising from different microenvironmental and/or epigenetic contexts. Here we aimed to further explore how target composition and TALEB architecture will impact the editing outcomes. We demonstrated how the nature of the linker between TALE array and split DddAtox head allows us to fine tune the editing window, also controlling possible bystander activity. Furthermore, we showed that both the TALEB architecture and spacer length separating the two TALE DNA binding regions impact the target TC editing dependence by the surrounding bases, leading to more restrictive or permissive editing profiles.


Subject(s)
Cytosine , Gene Editing , Thymine , Gene Editing/methods , Humans , Cytosine/metabolism , Cytosine/chemistry , Thymine/metabolism , Thymine/chemistry , Transcription Activator-Like Effectors/metabolism , Transcription Activator-Like Effectors/genetics , DNA/metabolism , DNA/genetics , HEK293 Cells
15.
Sci Adv ; 10(35): eadn9857, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39213364

ABSTRACT

Solid tumors, such as triple-negative breast cancer (TNBC), are biologically complex due to cellular heterogeneity, lack of tumor-specific antigens, and an immunosuppressive tumor microenvironment (TME). These challenges restrain chimeric antigen receptor (CAR) T cell efficacy, underlining the importance of armoring. In solid cancers, a localized tumor mass allows alternative administration routes, such as intratumoral delivery with the potential to improve efficacy and safety but may compromise metastatic-site treatment. Using a multi-layered CAR T cell engineering strategy that allowed a synergy between attributes, we show enhanced cytotoxic activity of MUC1 CAR T cells armored with PD1KO, tumor-specific interleukin-12 release, and TGFBR2KO attributes catered towards the TNBC TME. Intratumoral treatment effectively reduced distant tumors, suggesting retention of antigen-recognition benefits at metastatic sites. Overall, we provide preclinical evidence of armored non-alloreactive MUC1 CAR T cells greatly reducing high TNBC tumor burden in a TGFB1- and PD-L1-rich TME both at local and distant sites while preserving safety.


Subject(s)
Immunotherapy, Adoptive , Mucin-1 , Receptors, Chimeric Antigen , Triple Negative Breast Neoplasms , Tumor Microenvironment , Mucin-1/metabolism , Mucin-1/genetics , Mucin-1/immunology , Triple Negative Breast Neoplasms/therapy , Triple Negative Breast Neoplasms/pathology , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/metabolism , Humans , Female , Animals , Receptors, Chimeric Antigen/immunology , Receptors, Chimeric Antigen/metabolism , Receptors, Chimeric Antigen/genetics , Mice , Immunotherapy, Adoptive/methods , Cell Line, Tumor , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Xenograft Model Antitumor Assays
16.
Cell Rep Methods ; 4(9): 100857, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39260365

ABSTRACT

We present a TALEN-based workflow to generate and maintain dual-edited (IL-15+/+/TGFßR2-/-) iPSCs that produce enhanced iPSC-derived natural killer (iNK) cells for cancer immunotherapy. It involves using a cell lineage promoter for knocking in (KI) gene(s) to minimize the potential effects of expression of any exogenous genes on iPSCs. As a proof-of-principle, we KI IL-15 under the endogenous B2M promoter and show that it results in high expression of the sIL-15 in iNK cells but minimal expression in iPSCs. Furthermore, given that it is known that knockout (KO) of TGFßR2 in immune cells can enhance resistance to the suppressive TGF-ß signaling in the tumor microenvironment, we develop a customized medium containing Nodal that can maintain the pluripotency of iPSCs with TGFßR2 KO, enabling banking of these iPSC clones. Ultimately, we show that the dual-edited IL-15+/+/TGFßR2-/- iPSCs can be efficiently differentiated into NK cells that show enhanced autonomous growth and are resistant to the suppressive TGF-ß signaling.


Subject(s)
Induced Pluripotent Stem Cells , Interleukin-15 , Killer Cells, Natural , Receptor, Transforming Growth Factor-beta Type II , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/cytology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Interleukin-15/genetics , Interleukin-15/metabolism , Humans , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism , Cell Differentiation , Transcription Activator-Like Effector Nucleases/metabolism , Transcription Activator-Like Effector Nucleases/genetics , Gene Editing/methods
17.
Nat Commun ; 15(1): 4965, 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38862518

ABSTRACT

Sickle cell disease is a devastating blood disorder that originates from a single point mutation in the HBB gene coding for hemoglobin. Here, we develop a GMP-compatible TALEN-mediated gene editing process enabling efficient HBB correction via a DNA repair template while minimizing risks associated with HBB inactivation. Comparing viral versus non-viral DNA repair template delivery in hematopoietic stem and progenitor cells in vitro, both strategies achieve comparable HBB correction and result in over 50% expression of normal adult hemoglobin in red blood cells without inducing ß-thalassemic phenotype. In an immunodeficient female mouse model, transplanted cells edited with the non-viral strategy exhibit higher engraftment and gene correction levels compared to those edited with the viral strategy. Transcriptomic analysis reveals that non-viral DNA repair template delivery mitigates P53-mediated toxicity and preserves high levels of long-term hematopoietic stem cells. This work paves the way for TALEN-based autologous gene therapy for sickle cell disease.


Subject(s)
Anemia, Sickle Cell , Gene Editing , Genetic Therapy , Hematopoietic Stem Cells , Transcription Activator-Like Effector Nucleases , Anemia, Sickle Cell/therapy , Anemia, Sickle Cell/genetics , Gene Editing/methods , Animals , Hematopoietic Stem Cells/metabolism , Humans , Female , Mice , Genetic Therapy/methods , Transcription Activator-Like Effector Nucleases/metabolism , Transcription Activator-Like Effector Nucleases/genetics , Hematopoietic Stem Cell Transplantation , beta-Globins/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , DNA Repair , Mutation , beta-Thalassemia/therapy , beta-Thalassemia/genetics , Disease Models, Animal , Gene Transfer Techniques
18.
J Biol Chem ; 287(46): 38427-32, 2012 Nov 09.
Article in English | MEDLINE | ID: mdl-23019344

ABSTRACT

Within the past 2 years, transcription activator-like effector (TALE) DNA binding domains have emerged as the new generation of engineerable platform for production of custom DNA binding domains. However, their recently described sensitivity to cytosine methylation represents a major bottleneck for genome engineering applications. Using a combination of biochemical, structural, and cellular approaches, we were able to identify the molecular basis of such sensitivity and propose a simple, drug-free, and universal method to overcome it.


Subject(s)
Cytosine/chemistry , DNA Methylation , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Repressor Proteins/genetics , Repressor Proteins/physiology , Animals , Base Sequence , CHO Cells , Cricetinae , DNA/genetics , Epigenesis, Genetic , Gene Silencing , Genetic Engineering/methods , Genetic Therapy/methods , HEK293 Cells , Humans , Molecular Sequence Data , Mutagenesis , Protein Binding , Protein Engineering/methods , Protein Structure, Tertiary , Recombinant Proteins/chemistry
19.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 9): 1707-16, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23999294

ABSTRACT

Transcription activator-like effectors contain a DNA-binding domain organized in tandem repeats. The repeats include two adjacent residues known as the repeat variable di-residue, which recognize a single base pair, establishing a direct code between the dipeptides and the target DNA. This feature suggests this scaffold as an excellent candidate to generate new protein-DNA specificities for biotechnological applications. Here, the crystal structure of AvrBs3 (residues 152-895, molecular mass 82 kDa) in complex with its target DNA sequence is presented, revealing a new mode of interaction with the initial thymine of the target sequence, together with an analysis of both the binding specificity and the thermodynamic properties of AvrBs3. This study quantifies the affinity and the specificity between AvrBs3 and its target DNA. Moreover, in vitro and in vivo analyses reveal that AvrBs3 does not show a strict nucleotide-binding preference for the nucleotide at the zero position of the DNA, widening the number of possible sequences that could be targeted by this scaffold.


Subject(s)
Bacterial Proteins/chemistry , Bacterial Proteins/genetics , DNA, Bacterial/chemistry , Gene Targeting/methods , Tandem Repeat Sequences/genetics , Thymine/chemistry , Crystallization , Crystallography, X-Ray , DNA, Antisense/chemistry , DNA, Antisense/genetics , DNA, Bacterial/genetics , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Dipeptides/chemistry , Dipeptides/genetics , Nucleic Acid Heteroduplexes/chemistry , Nucleic Acid Heteroduplexes/genetics , Protein Interaction Mapping/methods , Thermodynamics , Transcription Activator-Like Effectors , Transcriptional Activation/genetics
20.
Proc Natl Acad Sci U S A ; 107(11): 4884-9, 2010 Mar 16.
Article in English | MEDLINE | ID: mdl-20194779

ABSTRACT

Pregnancy-associated malaria (PAM) is a serious consequence of sequestration of Plasmodium falciparum-parasitized erythrocytes (PE) in the placenta through adhesion to chondroitin sulfate A (CSA) present on placental proteoglycans. Recent work implicates var2CSA, a member of the PfEMP1 family, as the mediator of placental sequestration and as a key target for PAM vaccine development. Var2CSA is a 350 kDa transmembrane protein, whose extracellular region includes six Duffy-binding-like (DBL) domains. Due to its size and high cysteine content, the full-length var2CSA extracellular region has not hitherto been expressed in heterologous systems, thus limiting investigations to individual recombinant domains. Here we report for the first time the expression of the full-length var2CSA extracellular region (domains DBL1X to DBL6epsilon) from the 3D7 parasite strain using the human embryonic kidney 293 cell line. We show that the recombinant extracellular var2CSA region is correctly folded and that, unlike the individual DBL domains, it binds with high affinity and specificity to CSA (K(D) = 61 nM) and efficiently inhibits PE from binding to CSA. Structural characterization by analytical ultracentrifugation and small-angle x-ray scattering reveals a compact organization of the full-length protein, most likely governed by specific interdomain interactions, rather than an extended structure. Collectively, these data suggest that a high-affinity, CSA-specific binding site is formed by the higher-order structure of the var2CSA extracellular region. These results have important consequences for the development of an effective vaccine and therapeutic inhibitors.


Subject(s)
Antigens, Protozoan/chemistry , Antigens, Protozoan/metabolism , Chondroitin Sulfates/metabolism , Extracellular Space/chemistry , Plasmodium falciparum/metabolism , Animals , Cell Line , Chondroitin Sulfate Proteoglycans/metabolism , Circular Dichroism , Decorin , Erythrocytes/metabolism , Erythrocytes/parasitology , Extracellular Matrix Proteins/metabolism , Female , Humans , Kinetics , Models, Molecular , Parasites/metabolism , Placenta/metabolism , Pregnancy , Protein Binding , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Proteoglycans/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL