Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.685
Filter
Add more filters

Publication year range
1.
Cell ; 186(10): 2062-2077.e17, 2023 05 11.
Article in English | MEDLINE | ID: mdl-37075755

ABSTRACT

Entry of enveloped viruses into cells is mediated by viral fusogenic proteins that drive membrane rearrangements needed for fusion between viral and target membranes. Skeletal muscle development also requires membrane fusion events between progenitor cells to form multinucleated myofibers. Myomaker and Myomerger are muscle-specific cell fusogens but do not structurally or functionally resemble classical viral fusogens. We asked whether the muscle fusogens could functionally substitute for viral fusogens, despite their structural distinctiveness, and fuse viruses to cells. We report that engineering of Myomaker and Myomerger on the membrane of enveloped viruses leads to specific transduction of skeletal muscle. We also demonstrate that locally and systemically injected virions pseudotyped with the muscle fusogens can deliver µDystrophin to skeletal muscle of a mouse model of Duchenne muscular dystrophy and alleviate pathology. Through harnessing the intrinsic properties of myogenic membranes, we establish a platform for delivery of therapeutic material to skeletal muscle.


Subject(s)
Bioengineering , Lentivirus , Membrane Proteins , Muscle, Skeletal , Muscular Dystrophy, Duchenne , Animals , Mice , Cell Fusion , Membrane Fusion , Membrane Proteins/genetics , Membrane Proteins/metabolism , Muscle Development , Muscle, Skeletal/metabolism , Muscle, Skeletal/virology , Bioengineering/methods , Muscular Dystrophy, Duchenne/therapy , Disease Models, Animal , Viral Tropism , Lentivirus/genetics
2.
Cell ; 173(3): 665-676.e14, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29551272

ABSTRACT

Class 2 CRISPR-Cas systems endow microbes with diverse mechanisms for adaptive immunity. Here, we analyzed prokaryotic genome and metagenome sequences to identify an uncharacterized family of RNA-guided, RNA-targeting CRISPR systems that we classify as type VI-D. Biochemical characterization and protein engineering of seven distinct orthologs generated a ribonuclease effector derived from Ruminococcus flavefaciens XPD3002 (CasRx) with robust activity in human cells. CasRx-mediated knockdown exhibits high efficiency and specificity relative to RNA interference across diverse endogenous transcripts. As one of the most compact single-effector Cas enzymes, CasRx can also be flexibly packaged into adeno-associated virus. We target virally encoded, catalytically inactive CasRx to cis elements of pre-mRNA to manipulate alternative splicing, alleviating dysregulated tau isoform ratios in a neuronal model of frontotemporal dementia. Our results present CasRx as a programmable RNA-binding module for efficient targeting of cellular RNA, enabling a general platform for transcriptome engineering and future therapeutic development.


Subject(s)
CRISPR-Cas Systems , Computational Biology/methods , Genetic Engineering/methods , Protein Engineering/methods , RNA/analysis , Alternative Splicing , Animals , Bacterial Proteins/metabolism , Cell Differentiation , Escherichia coli/metabolism , Gene Expression Profiling , HEK293 Cells , Humans , Induced Pluripotent Stem Cells/cytology , Lentivirus/genetics , Mice , RNA Interference , RNA, Guide, Kinetoplastida/genetics , Ruminococcus , Sequence Analysis, RNA , Transcriptome
3.
Cell ; 173(7): 1622-1635.e14, 2018 06 14.
Article in English | MEDLINE | ID: mdl-29779948

ABSTRACT

Degrons are minimal elements that mediate the interaction of proteins with degradation machineries to promote proteolysis. Despite their central role in proteostasis, the number of known degrons remains small, and a facile technology to characterize them is lacking. Using a strategy combining global protein stability (GPS) profiling with a synthetic human peptidome, we identify thousands of peptides containing degron activity. Employing CRISPR screening, we establish that the stability of many proteins is regulated through degrons located at their C terminus. We characterize eight Cullin-RING E3 ubiquitin ligase (CRL) complex adaptors that regulate C-terminal degrons, including six CRL2 and two CRL4 complexes, and computationally implicate multiple non-CRLs in end recognition. Proteome analysis revealed that the C termini of eukaryotic proteins are depleted for C-terminal degrons, suggesting an E3-ligase-dependent modulation of proteome composition. Thus, we propose that a series of "C-end rules" operate to govern protein stability and shape the eukaryotic proteome.


Subject(s)
Proteome/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Motifs , Animals , Antigens, Neoplasm/metabolism , CRISPR-Cas Systems/genetics , Computational Biology/methods , Genetic Vectors/genetics , Genetic Vectors/metabolism , HEK293 Cells , Humans , Lentivirus/genetics , Leupeptins/pharmacology , Open Reading Frames/genetics , Peptides/metabolism , Proteasome Endopeptidase Complex/chemistry , Proteasome Endopeptidase Complex/metabolism , Protein Stability/drug effects , Protein Subunits/metabolism , Proteolysis , Proteome/genetics , Receptors, Cytokine/genetics , Receptors, Cytokine/metabolism
4.
Immunity ; 54(3): 571-585.e6, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33497609

ABSTRACT

CRISPR-Cas9 genome engineering has increased the pace of discovery for immunology and cancer biology, revealing potential therapeutic targets and providing insight into mechanisms underlying resistance to immunotherapy. However, endogenous immune recognition of Cas9 has limited the applicability of CRISPR technologies in vivo. Here, we characterized immune responses against Cas9 and other expressed CRISPR vector components that cause antigen-specific tumor rejection in several mouse cancer models. To avoid unwanted immune recognition, we designed a lentiviral vector system that allowed selective CRISPR antigen removal (SCAR) from tumor cells. The SCAR system reversed immune-mediated rejection of CRISPR-modified tumor cells in vivo and enabled high-throughput genetic screens in previously intractable models. A pooled in vivo screen using SCAR in a CRISPR-antigen-sensitive renal cell carcinoma revealed resistance pathways associated with autophagy and major histocompatibility complex class I (MHC class I) expression. Thus, SCAR presents a resource that enables CRISPR-based studies of tumor-immune interactions and prevents unwanted immune recognition of genetically engineered cells, with implications for clinical applications.


Subject(s)
Carcinoma, Renal Cell/immunology , Genetic Testing/methods , Genetic Vectors/genetics , Immunotherapy/methods , Kidney Neoplasms/immunology , Killer Cells, Natural/immunology , Lentivirus/genetics , Animals , Antigen Presentation , Autophagy , Carcinoma, Renal Cell/therapy , Cells, Cultured , Clustered Regularly Interspaced Short Palindromic Repeats , Genetic Engineering , Histocompatibility Antigens Class I/metabolism , Kidney Neoplasms/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Targeted Therapy
5.
Mol Cell ; 82(2): 479-491.e7, 2022 01 20.
Article in English | MEDLINE | ID: mdl-34963054

ABSTRACT

Genetically encoded biosensors are powerful tools to monitor cellular behavior, but the difficulty in generating appropriate reporters for chromatin factors hampers our ability to dissect epigenetic pathways. Here, we present TRACE (transgene reporters across chromatin environments), a high-throughput, genome-wide technique to generate fluorescent human reporter cell lines responsive to manipulation of epigenetic factors. By profiling GFP expression from a large pool of individually barcoded lentiviral integrants in the presence and absence of a perturbation, we identify reporters responsive to pharmacological inhibition of the histone lysine demethylase LSD1 and genetic ablation of the PRC2 subunit SUZ12. Furthermore, by manipulating the HIV-1 host factor LEDGF through targeted deletion or fusion to chromatin reader domains, we alter lentiviral integration site preferences, thus broadening the types of chromatin examined by TRACE. The phenotypic reporters generated through TRACE will allow the genetic interrogation of a broad range of epigenetic pathways, furthering our mechanistic understanding of chromatin biology.


Subject(s)
Biosensing Techniques , Epigenesis, Genetic , Genes, Reporter , Genetic Vectors , Green Fluorescent Proteins/genetics , Lentivirus/genetics , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Chromatin Assembly and Disassembly , Epigenome , Green Fluorescent Proteins/metabolism , HEK293 Cells , HeLa Cells , Histone Demethylases/genetics , Histone Demethylases/metabolism , Humans , Lentivirus/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , THP-1 Cells , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Mol Cell ; 81(22): 4692-4708.e9, 2021 11 18.
Article in English | MEDLINE | ID: mdl-34555355

ABSTRACT

Inhibitors of poly(ADP-ribose) (PAR) polymerase (PARPi) have entered the clinic for the treatment of homologous recombination (HR)-deficient cancers. Despite the success of this approach, preclinical and clinical research with PARPi has revealed multiple resistance mechanisms, highlighting the need for identification of novel functional biomarkers and combination treatment strategies. Functional genetic screens performed in cells and organoids that acquired resistance to PARPi by loss of 53BP1 identified loss of LIG3 as an enhancer of PARPi toxicity in BRCA1-deficient cells. Enhancement of PARPi toxicity by LIG3 depletion is dependent on BRCA1 deficiency but independent of the loss of 53BP1 pathway. Mechanistically, we show that LIG3 loss promotes formation of MRE11-mediated post-replicative ssDNA gaps in BRCA1-deficient and BRCA1/53BP1 double-deficient cells exposed to PARPi, leading to an accumulation of chromosomal abnormalities. LIG3 depletion also enhances efficacy of PARPi against BRCA1-deficient mammary tumors in mice, suggesting LIG3 as a potential therapeutic target.


Subject(s)
BRCA1 Protein/genetics , DNA Ligase ATP/genetics , DNA, Single-Stranded , MRE11 Homologue Protein/genetics , Ovarian Neoplasms/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly-ADP-Ribose Binding Proteins/genetics , Triple Negative Breast Neoplasms/metabolism , Tumor Suppressor p53-Binding Protein 1/genetics , Animals , Biopsy , CRISPR-Cas Systems , Cell Line , Cell Nucleus/metabolism , Cell Proliferation , Chromosome Aberrations , DNA Damage , DNA Ligase ATP/metabolism , Female , Humans , Lentivirus/genetics , Mammary Neoplasms, Animal , Mice , Mutation , Poly-ADP-Ribose Binding Proteins/metabolism , RNA, Small Interfering/metabolism , Transgenes
7.
Nature ; 596(7873): 576-582, 2021 08.
Article in English | MEDLINE | ID: mdl-34381210

ABSTRACT

Non-genetic mechanisms have recently emerged as important drivers of cancer therapy failure1, where some cancer cells can enter a reversible drug-tolerant persister state in response to treatment2. Although most cancer persisters remain arrested in the presence of the drug, a rare subset can re-enter the cell cycle under constitutive drug treatment. Little is known about the non-genetic mechanisms that enable cancer persisters to maintain proliferative capacity in the presence of drugs. To study this rare, transiently resistant, proliferative persister population, we developed Watermelon, a high-complexity expressed barcode lentiviral library for simultaneous tracing of each cell's clonal origin and proliferative and transcriptional states. Here we show that cycling and non-cycling persisters arise from different cell lineages with distinct transcriptional and metabolic programs. Upregulation of antioxidant gene programs and a metabolic shift to fatty acid oxidation are associated with persister proliferative capacity across multiple cancer types. Impeding oxidative stress or metabolic reprogramming alters the fraction of cycling persisters. In human tumours, programs associated with cycling persisters are induced in minimal residual disease in response to multiple targeted therapies. The Watermelon system enabled the identification of rare persister lineages that are preferentially poised to proliferate under drug pressure, thus exposing new vulnerabilities that can be targeted to delay or even prevent disease recurrence.


Subject(s)
Cell Cycle , Cell Lineage , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Neoplasms/drug therapy , Neoplasms/pathology , Antioxidants/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Lineage/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Clone Cells/drug effects , Clone Cells/metabolism , Clone Cells/pathology , DNA Barcoding, Taxonomic , Fatty Acids/metabolism , Gene Expression Regulation, Neoplastic , Humans , Lentivirus/genetics , Neoplasm Recurrence, Local/genetics , Neoplasms/genetics , Neoplasms/metabolism , Oncogene Proteins/antagonists & inhibitors , Oxidation-Reduction , Oxidative Stress , Reactive Oxygen Species/metabolism , Transcription, Genetic/drug effects
8.
N Engl J Med ; 386(5): 415-427, 2022 02 03.
Article in English | MEDLINE | ID: mdl-34891223

ABSTRACT

BACKGROUND: Betibeglogene autotemcel (beti-cel) gene therapy for transfusion-dependent ß-thalassemia contains autologous CD34+ hematopoietic stem cells and progenitor cells transduced with the BB305 lentiviral vector encoding the ß-globin (ßA-T87Q) gene. METHODS: In this open-label, phase 3 study, we evaluated the efficacy and safety of beti-cel in adult and pediatric patients with transfusion-dependent ß-thalassemia and a non-ß0/ß0 genotype. Patients underwent myeloablation with busulfan (with doses adjusted on the basis of pharmacokinetic analysis) and received beti-cel intravenously. The primary end point was transfusion independence (i.e., a weighted average hemoglobin level of ≥9 g per deciliter without red-cell transfusions for ≥12 months). RESULTS: A total of 23 patients were enrolled and received treatment, with a median follow-up of 29.5 months (range, 13.0 to 48.2). Transfusion independence occurred in 20 of 22 patients who could be evaluated (91%), including 6 of 7 patients (86%) who were younger than 12 years of age. The average hemoglobin level during transfusion independence was 11.7 g per deciliter (range, 9.5 to 12.8). Twelve months after beti-cel infusion, the median level of gene therapy-derived adult hemoglobin (HbA) with a T87Q amino acid substitution (HbAT87Q) was 8.7 g per deciliter (range, 5.2 to 10.6) in patients who had transfusion independence. The safety profile of beti-cel was consistent with that of busulfan-based myeloablation. Four patients had at least one adverse event that was considered by the investigators to be related or possibly related to beti-cel; all events were nonserious except for thrombocytopenia (in 1 patient). No cases of cancer were observed. CONCLUSIONS: Treatment with beti-cel resulted in a sustained HbAT87Q level and a total hemoglobin level that was high enough to enable transfusion independence in most patients with a non-ß0/ß0 genotype, including those younger than 12 years of age. (Funded by Bluebird Bio; HGB-207 ClinicalTrials.gov number, NCT02906202.).


Subject(s)
Biological Products/therapeutic use , Genetic Therapy/methods , beta-Globins/genetics , beta-Thalassemia/therapy , Adolescent , Adult , Biological Products/adverse effects , Busulfan/therapeutic use , Child , Erythrocyte Transfusion/adverse effects , Erythropoiesis , Female , Genetic Vectors , Genotype , Hemoglobins/analysis , Humans , Iron Overload/prevention & control , Lentivirus/genetics , Male , Middle Aged , Myeloablative Agonists/therapeutic use , beta-Thalassemia/blood , beta-Thalassemia/genetics
9.
Cell ; 141(3): 392-8, 2010 Apr 30.
Article in English | MEDLINE | ID: mdl-20434978

ABSTRACT

Mammalian cells are equipped with so-called "restriction factors" that suppress virus replication and help to prevent virus transmission from one species to another. This Essay discusses the host restriction factor tetherin, which blocks the release of enveloped viruses like HIV-1, and the factors evolved by primate lentiviruses, such as Vpu and Nef, that antagonize tetherin's action.


Subject(s)
Antigens, CD/metabolism , HIV-1/metabolism , Membrane Glycoproteins/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/immunology , GPI-Linked Proteins , Gene Products, nef/metabolism , Human Immunodeficiency Virus Proteins/metabolism , Humans , Lentivirus/genetics , Lentivirus Infections/drug therapy , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Viral Regulatory and Accessory Proteins/metabolism
10.
Mol Ther ; 32(1): 124-139, 2024 Jan 03.
Article in English | MEDLINE | ID: mdl-37990494

ABSTRACT

Quiescent human hematopoietic stem cells (HSC) are ideal targets for gene therapy applications due to their preserved stemness and repopulation capacities; however, they have not been exploited extensively because of their resistance to genetic manipulation. We report here the development of a lentiviral transduction protocol that overcomes this resistance in long-term repopulating quiescent HSC, allowing their efficient genetic manipulation. Mechanistically, lentiviral vector transduction of quiescent HSC was found to be restricted at the level of vector entry and by limited pyrimidine pools. These restrictions were overcome by the combined addition of cyclosporin H (CsH) and deoxynucleosides (dNs) during lentiviral vector transduction. Clinically relevant transduction levels were paired with higher polyclonal engraftment of long-term repopulating HSC as compared with standard ex vivo cultured controls. These findings identify the cell-intrinsic barriers that restrict the transduction of quiescent HSC and provide a means to overcome them, paving the way for the genetic engineering of unstimulated HSC.


Subject(s)
Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells , Humans , Transduction, Genetic , Lentivirus/genetics , Genetic Therapy/methods , Immunity, Innate , Genetic Vectors/genetics , Antigens, CD34
11.
Mol Ther ; 32(5): 1497-1509, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38429928

ABSTRACT

The hallmark of epidermolysis bullosa (EB) is fragile attachment of epithelia due to genetic variants in cell adhesion genes. We describe 16 EB patients treated in the ear, nose, and throat department of a tertiary pediatric hospital linked to the United Kingdom's national EB unit between 1992 and 2023. Patients suffered a high degree of morbidity and mortality from laryngotracheal stenosis. Variants in laminin subunit alpha-3 (LAMA3) were found in 10/15 patients where genotype was available. LAMA3 encodes a subunit of the laminin-332 heterotrimeric extracellular matrix protein complex and is expressed by airway epithelial basal stem cells. We investigated the benefit of restoring wild-type LAMA3 expression in primary EB patient-derived basal cell cultures. EB basal cells demonstrated weak adhesion to cell culture substrates, but could otherwise be expanded similarly to non-EB basal cells. In vitro lentiviral overexpression of LAMA3A in EB basal cells enabled them to differentiate in air-liquid interface cultures, producing cilia with normal ciliary beat frequency. Moreover, transduction restored cell adhesion to levels comparable to a non-EB donor culture. These data provide proof of concept for a combined cell and gene therapy approach to treat airway disease in LAMA3-affected EB.


Subject(s)
Cell Adhesion , Epidermolysis Bullosa , Laminin , Lentivirus , Humans , Laminin/metabolism , Laminin/genetics , Epidermolysis Bullosa/genetics , Epidermolysis Bullosa/metabolism , Epidermolysis Bullosa/therapy , Epidermolysis Bullosa/pathology , Child , Lentivirus/genetics , Male , Female , Child, Preschool , Genetic Therapy/methods , Genetic Vectors/genetics , Epithelial Cells/metabolism , Cells, Cultured , Gene Expression , Adolescent , Infant
12.
Mol Ther ; 32(3): 619-636, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38310355

ABSTRACT

Mucopolysaccharidosis type II (MPS II), or Hunter syndrome, is a rare X-linked recessive lysosomal storage disorder due to a mutation in the lysosomal enzyme iduronate-2-sulfatase (IDS) gene. IDS deficiency leads to a progressive, multisystem accumulation of glycosaminoglycans (GAGs) and results in central nervous system (CNS) manifestations in the severe form. We developed up to clinical readiness a new hematopoietic stem cell (HSC) gene therapy approach for MPS II that benefits from a novel highly effective transduction protocol. We first provided proof of concept of efficacy of our approach aimed at enhanced IDS enzyme delivery to the CNS in a murine study of immediate translational value, employing a lentiviral vector (LV) encoding a codon-optimized human IDS cDNA. Then the therapeutic LV was tested for its ability to efficiently and safely transduce bona fide human HSCs in clinically relevant conditions according to a standard vs. a novel protocol that demonstrated superior ability to transduce bona fide long-term repopulating HSCs. Overall, these results provide strong proof of concept for the clinical translation of this approach for the treatment of Hunter syndrome.


Subject(s)
Iduronate Sulfatase , Mucopolysaccharidosis II , Humans , Animals , Mice , Mucopolysaccharidosis II/therapy , Mucopolysaccharidosis II/drug therapy , Iduronate Sulfatase/genetics , Iduronate Sulfatase/metabolism , Genetic Therapy , Central Nervous System/metabolism , Lentivirus/genetics , Lentivirus/metabolism , Hematopoietic Stem Cells/metabolism
13.
Mol Ther ; 32(5): 1202-1218, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38454604

ABSTRACT

In recent years, a growing number of clinical trials have been initiated to evaluate gene therapy approaches for the treatment of patients with transfusion-dependent ß-thalassemia and sickle cell disease (SCD). Therapeutic modalities being assessed in these trials utilize different molecular techniques, including lentiviral vectors to add functional copies of the gene encoding the hemoglobin ß subunit in defective cells and CRISPR-Cas9, transcription activator-like effector protein nuclease, and zinc finger nuclease gene editing strategies to either directly address the underlying genetic cause of disease or induce fetal hemoglobin production by gene disruption. Here, we review the mechanisms of action of these various gene addition and gene editing approaches and describe the status of clinical trials designed to evaluate the potentially for these approaches to provide one-time functional cures to patients with transfusion-dependent ß-thalassemia and SCD.


Subject(s)
Genetic Therapy , Hemoglobinopathies , Animals , Humans , Anemia, Sickle Cell/therapy , Anemia, Sickle Cell/genetics , beta-Thalassemia/therapy , beta-Thalassemia/genetics , Clinical Trials as Topic , CRISPR-Cas Systems , Gene Editing/methods , Genetic Therapy/methods , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Hemoglobinopathies/therapy , Hemoglobinopathies/genetics , Lentivirus/genetics
14.
Mol Ther ; 32(5): 1311-1327, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38449314

ABSTRACT

While studying transgene expression after systemic administration of lentiviral vectors, we found that splenic B cells are robustly transduced, regardless of the types of pseudotyped envelope proteins. However, the administration of two different pseudotypes resulted in transduction of two distinct B cell populations, suggesting that each pseudotype uses unique and specific receptors for its attachment and entry into splenic B cells. Single-cell RNA sequencing analysis of the transduced cells demonstrated that different pseudotypes transduce distinct B cell subpopulations characterized by specific B cell receptor (BCR) genotypes. Functional analysis of the BCRs of the transduced cells demonstrated that BCRs specific to the pseudotyping envelope proteins mediate viral entry, enabling the vectors to selectively transduce the B cell populations that are capable of producing antibodies specific to their envelope proteins. Lentiviral vector entry via the BCR activated the transduced B cells and induced proliferation and differentiation into mature effectors, such as memory B and plasma cells. BCR-mediated viral entry into clonally specific B cell subpopulations raises new concepts for understanding the biodistribution of transgene expression after systemic administration of lentiviral vectors and offers new opportunities for BCR-targeted gene delivery by pseudotyped lentiviral vectors.


Subject(s)
B-Lymphocytes , Genetic Vectors , Lentivirus , Receptors, Antigen, B-Cell , Transduction, Genetic , Transgenes , Viral Envelope Proteins , Lentivirus/genetics , Receptors, Antigen, B-Cell/metabolism , Receptors, Antigen, B-Cell/genetics , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Animals , Mice , B-Lymphocytes/metabolism , B-Lymphocytes/immunology , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Viral Tropism , Humans , Virus Internalization
15.
Mol Ther ; 32(7): 2223-2231, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38796702

ABSTRACT

Positron emission tomography (PET) reporter systems are a valuable means of estimating the level of expression of a transgene in vivo. For example, the safety and efficacy of gene therapy approaches for the treatment of neurological and neuropsychiatric disorders could be enhanced via the monitoring of exogenous gene expression levels in the brain. The present study evaluated the ability of a newly developed PET reporter system [18F]fluoroestradiol ([18F]FES) and the estrogen receptor-based PET reporter ChRERα, to monitor expression levels of a small hairpin RNA (shRNA) designed to suppress choline acetyltransferase (ChAT) expression in rhesus monkey brain. The ChRERα gene and shRNA were expressed from the same transcript via lentivirus injected into monkey striatum. In two monkeys that received injections of viral vector, [18F]FES binding increased by 70% and 86% at the target sites compared with pre-injection, demonstrating that ChRERα expression could be visualized in vivo with PET imaging. Post-mortem immunohistochemistry confirmed that ChAT expression was significantly suppressed in regions in which [18F]FES uptake was increased. The consistency between PET imaging and immunohistochemical results suggests that [18F]FES and ChRERα can serve as a PET reporter system in rhesus monkey brain for in vivo evaluation of the expression of potential therapeutic agents, such as shRNAs.


Subject(s)
Brain , Estradiol , Genes, Reporter , Macaca mulatta , Positron-Emission Tomography , Animals , Positron-Emission Tomography/methods , Estradiol/analogs & derivatives , Estradiol/pharmacology , Brain/metabolism , Brain/diagnostic imaging , Fluorine Radioisotopes , Receptors, Estrogen/metabolism , Receptors, Estrogen/genetics , Genetic Vectors/genetics , Genetic Vectors/administration & dosage , Gene Expression , RNA, Small Interfering/genetics , Lentivirus/genetics , Humans
16.
Mol Cell ; 68(1): 210-223.e6, 2017 Oct 05.
Article in English | MEDLINE | ID: mdl-28985505

ABSTRACT

Chemical libraries paired with phenotypic screens can now readily identify compounds with therapeutic potential. A central limitation to exploiting these compounds, however, has been in identifying their relevant cellular targets. Here, we present a two-tiered CRISPR-mediated chemical-genetic strategy for target identification: combined genome-wide knockdown and overexpression screening as well as focused, comparative chemical-genetic profiling. Application of these strategies to rigosertib, a drug in phase 3 clinical trials for high-risk myelodysplastic syndrome whose molecular target had remained controversial, pointed singularly to microtubules as rigosertib's target. We showed that rigosertib indeed directly binds to and destabilizes microtubules using cell biological, in vitro, and structural approaches. Finally, expression of tubulin with a structure-guided mutation in the rigosertib-binding pocket conferred resistance to rigosertib, establishing that rigosertib kills cancer cells by destabilizing microtubules. These results demonstrate the power of our chemical-genetic screening strategies for pinpointing the physiologically relevant targets of chemical agents.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic , Genetic Testing/methods , Glycine/analogs & derivatives , Microtubules/drug effects , Sulfones/pharmacology , Tubulin Modulators/pharmacology , Tubulin/genetics , Antineoplastic Agents/chemistry , CRISPR-Cas Systems , Colchicine/pharmacology , Drug Resistance, Neoplasm , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Glycine/chemistry , Glycine/pharmacology , HeLa Cells , Humans , K562 Cells , Kinesins/genetics , Kinesins/metabolism , Lentivirus/genetics , Lentivirus/metabolism , Microtubules/metabolism , Microtubules/ultrastructure , Mutation , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/metabolism , Myelodysplastic Syndromes/pathology , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Small Molecule Libraries/pharmacology , Sulfones/chemistry , Tubulin/chemistry , Tubulin/metabolism , Tubulin Modulators/chemistry , Vinblastine/pharmacology
17.
Cell Mol Life Sci ; 81(1): 305, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39012348

ABSTRACT

Lentiviral vectors have markedly enhanced gene therapy efficiency in treating congenital diseases, but their long-term safety remains controversial. Most gene therapies for congenital eye diseases need to be carried out at early ages, yet the assessment of related risks to ocular development posed by lentiviral vectors is challenging. Utilizing single-cell transcriptomic profiling on human retinal organoids, this study explored the impact of lentiviral vectors on the retinal development and found that lentiviral vectors can cause retinal precursor cells to shift toward photoreceptor fate through the up-regulation of key fate-determining genes such as PRDM1. Further investigation demonstrated that the intron and intergenic region of PRDM1 was bound by PHLDA1, which was also up-regulated by lentiviral vectors exposure. Importantly, knockdown of PHLDA1 successfully suppressed the lentivirus-induced differentiation bias of photoreceptor cells. The findings also suggest that while lentiviral vectors may disrupt the fate determination of retinal precursor cells, posing risks in early-stage retinal gene therapy, these risks could potentially be reduced by inhibiting the PHLDA1-PRDM1 axis.


Subject(s)
Cell Differentiation , Genetic Vectors , Lentivirus , Retina , Stem Cells , Transcription Factors , Humans , Retina/metabolism , Retina/cytology , Lentivirus/genetics , Transcription Factors/metabolism , Transcription Factors/genetics , Genetic Vectors/metabolism , Genetic Vectors/genetics , Cell Differentiation/genetics , Stem Cells/metabolism , Stem Cells/cytology , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/metabolism , Organoids/metabolism , Organoids/cytology , Nuclear Proteins/metabolism , Nuclear Proteins/genetics , Genetic Therapy/methods
18.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Article in English | MEDLINE | ID: mdl-35042811

ABSTRACT

T cells promote our body's ability to battle cancers and infectious diseases but can act pathologically in autoimmunity. The recognition of peptides presented by major histocompatibility complex (pMHC) molecules by T cell receptors (TCRs) enables T cell-mediated responses. To modify disease-relevant T cells, new tools to genetically modify T cells and decode their antigen recognition are needed. Here, we present an approach using viruses pseudotyped with peptides loaded on MHC called V-CARMA (Viral ChimAeric Receptor MHC-Antigen) to specifically target T cells expressing cognate TCRs for antigen discovery and T cell engineering. We show that lentiviruses displaying antigens on human leukocyte antigen (HLA) class I and class II molecules can robustly infect CD8+ and CD4+ T cells expressing cognate TCRs, respectively. The infection rates of the pseudotyped lentiviruses (PLVs) are correlated with the binding affinity of the TCR to its cognate antigen. Furthermore, peptide-HLA pseudotyped lentivirus V-CARMA constructs can identify target cells from a mixed T cell population, suppress PD-1 expression on CD8+ T cells via PDCD1 shRNA delivery, and induce apoptosis in autoreactive CD4+ T cells. Thus, V-CARMA is a versatile tool for TCR ligand identification and selective T cell manipulation.


Subject(s)
Genetic Engineering/methods , Immunotherapy/methods , Lymphokines/metabolism , Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Histocompatibility Antigens Class I/immunology , Humans , Lentivirus/genetics , Lentivirus/immunology , Lymphocyte Activation , Lymphokines/physiology , Major Histocompatibility Complex , Peptides/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Chimeric Antigen/genetics
19.
Genes Dev ; 31(6): 537-552, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28404629

ABSTRACT

Rapid advances in genetics are linking mutations on genes to diseases at an exponential rate, yet characterizing the gene-mutation-cell-behavior relationships essential for precision medicine remains a daunting task. More than 350 mutations on small GTPase BRaf are associated with various tumors, and ∼40 mutations are associated with the neurodevelopmental disorder cardio-facio-cutaneous syndrome (CFC). We developed a fast cost-effective lentivirus-based rapid gene replacement method to interrogate the physiopathology of BRaf and ∼50 disease-linked BRaf mutants, including all CFC-linked mutants. Analysis of simultaneous multiple patch-clamp recordings from 6068 pairs of rat neurons with validation in additional mouse and human neurons and multiple learning tests from 1486 rats identified BRaf as the key missing signaling effector in the common synaptic NMDA-R-CaMKII-SynGap-Ras-BRaf-MEK-ERK transduction cascade. Moreover, the analysis creates the original big data unveiling three general features of BRaf signaling. This study establishes the first efficient procedure that permits large-scale functional analysis of human disease-linked mutations essential for precision medicine.


Subject(s)
MAP Kinase Signaling System/genetics , Mutation , Proto-Oncogene Proteins B-raf/genetics , Synaptic Transmission/genetics , Animals , Cells, Cultured , Disease/genetics , Female , Gene Transfer Techniques , Humans , Lentivirus/genetics , Male , Mice, Inbred C57BL , Neurons/physiology , Rats, Sprague-Dawley , Tissue Culture Techniques
20.
J Hepatol ; 80(1): 31-40, 2024 01.
Article in English | MEDLINE | ID: mdl-37827470

ABSTRACT

BACKGROUND & AIMS: Immunotherapy for chronic hepatitis B virus (HBV) infection has not yet demonstrated sufficient efficacy. We developed a non-integrative lentiviral-vectored therapeutic vaccine for chronic hepatitis B and tested its antiviral effects in HBV-persistent mice and two inactive HBsAg carriers. METHODS: Lentiviral vectors (LVs) encoding the core, preS1, or large HBsAg (LHBs) proteins of HBV were evaluated for immunogenicity in HBV-naïve mice and therapeutic efficacy in a murine model of chronic HBV infection. In addition, two inactive HBsAg carriers each received two doses of 5×107 transduction units (TU) or 1×108 TU of lentiviral-vectored LHBs (LV-LHBs), respectively. The endpoints were safety, LHBs-specific T-cell responses, and serum HBsAg levels during a 24-week follow-up. RESULTS: In the mouse models, LV-LHBs was the most promising in eliciting robust antigen-specific T cells and in reducing the levels of serum HBsAg and viral load. By the end of the 34-week observation period, six out of ten (60%) HBV-persistent mice vaccinated with LV-LHBs achieved serum HBsAg loss and significant depletion of HBV-positive hepatocytes in the liver. In the two inactive HBsAg carriers, vaccination with LV-LHBs induced a considerable increase in the number of peripheral LHBs-specific T cells in one patient, and a weak but detectable response in the other, accompanied by a sustained reduction of HBsAg (-0.31 log10 IU/ml and -0.46 log10 IU/ml, respectively) from baseline to nadir. CONCLUSIONS: A lentiviral-vectored therapeutic vaccine for chronic HBV infection demonstrated the potential to improve HBV-specific T-cell responses and deplete HBV-positive hepatocytes, leading to a sustained loss or reduction of serum HBsAg. IMPACT AND IMPLICATIONS: Chronic HBV infection is characterized by an extremely low number and profound hypo-responsiveness of HBV-specific T cells. Therapeutic vaccines are designed to improve HBV-specific T-cell responses. We show that immunization with a lentiviral-vectored therapeutic HBV vaccine was able to expand HBV-specific T cells in vivo, leading to reductions of HBV-positive hepatocytes and serum HBsAg.


Subject(s)
Hepatitis B, Chronic , Humans , Mice , Animals , Hepatitis B, Chronic/prevention & control , Hepatitis B, Chronic/drug therapy , Hepatitis B virus , Hepatitis B Surface Antigens , Lentivirus/genetics , Hepatitis B Vaccines/therapeutic use , Vaccination
SELECTION OF CITATIONS
SEARCH DETAIL