Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 137
Filter
Add more filters

Publication year range
1.
Cell ; 186(2): 305-326.e27, 2023 01 19.
Article in English | MEDLINE | ID: mdl-36638792

ABSTRACT

All living things experience an increase in entropy, manifested as a loss of genetic and epigenetic information. In yeast, epigenetic information is lost over time due to the relocalization of chromatin-modifying proteins to DNA breaks, causing cells to lose their identity, a hallmark of yeast aging. Using a system called "ICE" (inducible changes to the epigenome), we find that the act of faithful DNA repair advances aging at physiological, cognitive, and molecular levels, including erosion of the epigenetic landscape, cellular exdifferentiation, senescence, and advancement of the DNA methylation clock, which can be reversed by OSK-mediated rejuvenation. These data are consistent with the information theory of aging, which states that a loss of epigenetic information is a reversible cause of aging.


Subject(s)
Aging , Epigenesis, Genetic , Animals , Aging/genetics , DNA Methylation , Epigenome , Mammals/genetics , Nucleoproteins , Saccharomyces cerevisiae/genetics
3.
Nature ; 632(8023): 192-200, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39020181

ABSTRACT

Gene replacement using adeno-associated virus (AAV) vectors is a promising therapeutic approach for many diseases1,2. However, this therapeutic modality is challenged by the packaging capacity of AAVs (approximately 4.7 kilobases)3, limiting its application for disorders involving large coding sequences, such as Duchenne muscular dystrophy, with a 14 kilobase messenger RNA. Here we developed a new method for expressing large dystrophins by utilizing the protein trans-splicing mechanism mediated by split inteins. We identified several split intein pairs that efficiently join two or three fragments to generate a large midi-dystrophin or the full-length protein. We show that delivery of two or three AAVs into dystrophic mice results in robust expression of large dystrophins and significant physiological improvements compared with micro-dystrophins. Moreover, using the potent myotropic AAVMYO4, we demonstrate that low total doses (2 × 1013 viral genomes per kg) are sufficient to express large dystrophins in striated muscles body-wide with significant physiological corrections in dystrophic mice. Our data show a clear functional superiority of large dystrophins over micro-dystrophins that are being tested in clinical trials. This method could benefit many patients with Duchenne or Becker muscular dystrophy, regardless of genotype, and could be adapted to numerous other disorders caused by mutations in large genes that exceed the AAV capacity.


Subject(s)
Dystrophin , Genetic Therapy , Inteins , Muscular Dystrophy, Duchenne , Protein Splicing , Animals , Humans , Male , Mice , Dependovirus/genetics , Dependovirus/metabolism , Disease Models, Animal , Dystrophin/biosynthesis , Dystrophin/deficiency , Dystrophin/genetics , Dystrophin/metabolism , Genetic Therapy/methods , Genetic Vectors/genetics , Genetic Vectors/metabolism , Inteins/genetics , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Muscular Dystrophy, Duchenne/metabolism , Protein Splicing/genetics
4.
Nature ; 603(7901): 482-487, 2022 03.
Article in English | MEDLINE | ID: mdl-35264795

ABSTRACT

Methane (CH4), the most abundant hydrocarbon in the atmosphere, originates largely from biogenic sources1 linked to an increasing number of organisms occurring in oxic and anoxic environments. Traditionally, biogenic CH4 has been regarded as the final product of anoxic decomposition of organic matter by methanogenic archaea. However, plants2,3, fungi4, algae5 and cyanobacteria6 can produce CH4 in the presence of oxygen. Although methanogens are known to produce CH4 enzymatically during anaerobic energy metabolism7, the requirements and pathways for CH4 production by non-methanogenic cells are poorly understood. Here, we demonstrate that CH4 formation by Bacillus subtilis and Escherichia coli is triggered by free iron and reactive oxygen species (ROS), which are generated by metabolic activity and enhanced by oxidative stress. ROS-induced methyl radicals, which are derived from organic compounds containing sulfur- or nitrogen-bonded methyl groups, are key intermediates that ultimately lead to CH4 production. We further show CH4 production by many other model organisms from the Bacteria, Archaea and Eukarya domains, including in several human cell lines. All these organisms respond to inducers of oxidative stress by enhanced CH4 formation. Our results imply that all living cells probably possess a common mechanism of CH4 formation that is based on interactions among ROS, iron and methyl donors, opening new perspectives for understanding biochemical CH4 formation and cycling.


Subject(s)
Archaea , Euryarchaeota , Methane , Archaea/metabolism , Cell Line , Cell Physiological Phenomena , Humans , Iron/metabolism , Methane/chemistry , Methane/metabolism , Nitrogen/metabolism , Reactive Oxygen Species/metabolism , Sulfur/metabolism
5.
PLoS Biol ; 22(7): e3002637, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39018342

ABSTRACT

A new study characterizes and improves a novel small Cas12a variant before adapting it for in vivo genome editing by delivery via adeno-associated virus (AAV) vectors, showcasing the potential of small CRISPR systems and their compatibility with viral vectors.


Subject(s)
CRISPR-Cas Systems , Dependovirus , Gene Editing , Genetic Vectors , Gene Editing/methods , Dependovirus/genetics , CRISPR-Cas Systems/genetics , Genetic Vectors/genetics , Animals , CRISPR-Associated Proteins/genetics , CRISPR-Associated Proteins/metabolism , Humans , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Endodeoxyribonucleases/genetics , Endodeoxyribonucleases/metabolism
6.
EMBO Rep ; 25(10): 4311-4336, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39232200

ABSTRACT

Current culture systems available for studying hepatitis D virus (HDV) are suboptimal. In this study, we demonstrate that hepatocyte-like cells (HLCs) derived from human pluripotent stem cells (hPSCs) are fully permissive to HDV infection across various tested genotypes. When co-infected with the helper hepatitis B virus (HBV) or transduced to express the HBV envelope protein HBsAg, HLCs effectively release infectious progeny virions. We also show that HBsAg-expressing HLCs support the extracellular spread of HDV, thus providing a valuable platform for testing available anti-HDV regimens. By challenging the cells along the differentiation with HDV infection, we have identified CD63 as a potential HDV co-entry factor that was rate-limiting for HDV infection in immature hepatocytes. Given their renewable source and the potential to derive hPSCs from individual patients, we propose HLCs as a promising model for investigating HDV biology. Our findings offer new insights into HDV infection and expand the repertoire of research tools available for the development of therapeutic interventions.


Subject(s)
Cell Differentiation , Hepatitis B virus , Hepatitis B , Hepatitis Delta Virus , Hepatocytes , Pluripotent Stem Cells , Humans , Hepatitis Delta Virus/physiology , Hepatitis Delta Virus/genetics , Hepatocytes/virology , Hepatocytes/metabolism , Pluripotent Stem Cells/virology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Hepatitis B virus/physiology , Hepatitis B virus/genetics , Hepatitis B/virology , Hepatitis D/virology , Virus Replication , Hepatitis B Surface Antigens/metabolism , Hepatitis B Surface Antigens/genetics , Virus Internalization
7.
J Cell Sci ; 136(12)2023 06 15.
Article in English | MEDLINE | ID: mdl-37288813

ABSTRACT

The axon initial segment (AIS) is a highly specialized neuronal compartment that regulates the generation of action potentials and maintenance of neuronal polarity. Live imaging of the AIS is challenging due to the limited number of suitable labeling methods. To overcome this limitation, we established a novel approach for live labeling of the AIS using unnatural amino acids (UAAs) and click chemistry. The small size of UAAs and the possibility of introducing them virtually anywhere into target proteins make this method particularly suitable for labeling of complex and spatially restricted proteins. Using this approach, we labeled two large AIS components, the 186 kDa isoform of neurofascin (NF186; encoded by Nfasc) and the 260 kDa voltage-gated Na+ channel (NaV1.6, encoded by Scn8a) in primary neurons and performed conventional and super-resolution microscopy. We also studied the localization of epilepsy-causing NaV1.6 variants with a loss-of-function effect. Finally, to improve the efficiency of UAA incorporation, we developed adeno-associated viral (AAV) vectors for click labeling in neurons, an achievement that could be transferred to more complex systems such as organotypic slice cultures, organoids, and animal models.


Subject(s)
Axon Initial Segment , Click Chemistry , Animals , Action Potentials/physiology , Amino Acids/metabolism , Axon Initial Segment/metabolism , Neurons , Mice , Rats
8.
Development ; 149(8)2022 04 15.
Article in English | MEDLINE | ID: mdl-35420133

ABSTRACT

The ectopic expression of the transcription factors OCT4, SOX2, KLF4 and MYC (OSKM) enables reprogramming of differentiated cells into pluripotent embryonic stem cells. Methods based on partial and reversible in vivo reprogramming are a promising strategy for tissue regeneration and rejuvenation. However, little is known about the barriers that impair reprogramming in an in vivo context. We report that natural killer (NK) cells significantly limit reprogramming, both in vitro and in vivo. Cells and tissues in the intermediate states of reprogramming upregulate the expression of NK-activating ligands, such as MULT1 and ICAM1. NK cells recognize and kill partially reprogrammed cells in a degranulation-dependent manner. Importantly, in vivo partial reprogramming is strongly reduced by adoptive transfer of NK cells, whereas it is significantly increased by their depletion. Notably, in the absence of NK cells, the pancreatic organoids derived from OSKM-expressing mice are remarkably large, suggesting that ablating NK surveillance favours the acquisition of progenitor-like properties. We conclude that NK cells pose an important barrier for in vivo reprogramming, and speculate that this concept may apply to other contexts of transient cellular plasticity.


Subject(s)
Cellular Reprogramming , Pluripotent Stem Cells , Animals , Cell Differentiation , Cellular Reprogramming/genetics , Embryonic Stem Cells/metabolism , Fibroblasts/metabolism , Killer Cells, Natural/metabolism , Kruppel-Like Factor 4/metabolism , Mice , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/cytology , SOXB1 Transcription Factors/metabolism
9.
Mol Ther ; 32(10): 3470-3484, 2024 Oct 02.
Article in English | MEDLINE | ID: mdl-39113357

ABSTRACT

One of the biggest challenges for in vivo gene therapy are vectors mediating highly selective gene transfer into a defined population of therapy-relevant cells. Here we present DARPin-targeted AAVs (DART-AAVs) displaying DARPins specific for human and murine CD8. Insertion of DARPins into the GH2/GH3 loop of the capsid protein 1 (VP1) of AAV2 and AAV6 resulted in high selectivity for CD8-positive T cells with unimpaired gene delivery activity. Remarkably, the capsid core structure was unaltered with protruding DARPins detectable. In complex primary cell mixtures, including donor blood or systemic injections into mice, the CD8-targeted AAVs were by far superior to unmodified AAV2 and AAV6 in terms of selectivity, target cell viability, and gene transfer rates. In vivo, up to 80% of activated CD8+ T cells were hit upon a single vector injection into conditioned humanized or immunocompetent mice. While gene transfer rates decreased significantly under non-activated conditions, genomic modification selectively in CD8+ T cells was still detectable upon Cre delivery into indicator mice. In both mouse models, selectivity for CD8+ T cells was close to absolute with exceptional detargeting from liver. The CD8-AAVs described here expand strategies for immunological research and in vivo gene therapy options.


Subject(s)
CD8-Positive T-Lymphocytes , Dependovirus , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors , Animals , Dependovirus/genetics , Mice , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Humans , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Genetic Therapy/methods , Transduction, Genetic , Capsid Proteins/genetics
10.
Basic Res Cardiol ; 118(1): 13, 2023 03 29.
Article in English | MEDLINE | ID: mdl-36988697

ABSTRACT

The prospective use of human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CM) for cardiac regenerative medicine strongly depends on the electro-mechanical properties of these cells, especially regarding the Ca2+-dependent excitation-contraction (EC) coupling mechanism. Currently, the immature structural and functional features of hiPSC-CM limit the progression towards clinical applications. Here, we show that a specific microarchitecture is essential for functional maturation of hiPSC-CM. Structural remodelling towards a cuboid cell shape and induction of BIN1, a facilitator of membrane invaginations, lead to transverse (t)-tubule-like structures. This transformation brings two Ca2+ channels critical for EC coupling in close proximity, the L-type Ca2+ channel at the sarcolemma and the ryanodine receptor at the sarcoplasmic reticulum. Consequently, the Ca2+-dependent functional interaction of these channels becomes more efficient, leading to improved spatio-temporal synchronisation of Ca2+ transients and higher EC coupling gain. Thus, functional maturation of hiPSC-cardiomyocytes by optimised cell microarchitecture needs to be considered for future cardiac regenerative approaches.


Subject(s)
Induced Pluripotent Stem Cells , Myocytes, Cardiac , Humans , Myocytes, Cardiac/metabolism , Induced Pluripotent Stem Cells/metabolism , Excitation Contraction Coupling , Calcium Signaling , Ryanodine Receptor Calcium Release Channel/metabolism , Calcium/metabolism
11.
Cytotherapy ; 25(3): 254-260, 2023 03.
Article in English | MEDLINE | ID: mdl-35999132

ABSTRACT

Adeno-associated virus (AAV) is one of the most exciting and most versatile templates for engineering of gene-delivery vectors for use in human gene therapy, owing to the existence of numerous naturally occurring capsid variants and their amenability to directed molecular evolution. As a result, the field has witnessed an explosion of novel "designer" AAV capsids and ensuing vectors over the last two decades, which have been isolated from comprehensive capsid libraries generated through technologies such as DNA shuffling or peptide display, and stratified under stringent positive and/or negative selection pressures. Here, we briefly highlight a panel of recent, innovative and transformative methodologies that we consider to have exceptional potential to advance directed AAV capsid evolution and to thereby accelerate AAV vector revolution. These avenues comprise original technologies for (i) barcoding and high-throughput screening of individual AAV variants or entire capsid libraries, (ii) selection of transduction-competent AAV vectors on the DNA level, (iii) enrichment of expression-competent AAV variants on the RNA level, as well as (iv) high-resolution stratification of focused AAV capsid libraries on the single-cell level. Together with other emerging AAV engineering stratagems, such as rational design or machine learning, these pioneering techniques promise to provide an urgently needed booster for AAV (r)evolution.


Subject(s)
Dependovirus , Genetic Therapy , Humans , Dependovirus/genetics , Genetic Therapy/methods , Gene Transfer Techniques , Capsid Proteins/chemistry , Capsid Proteins/genetics , Capsid , Genetic Vectors/genetics
12.
Mol Ther ; 30(5): 2005-2023, 2022 05 04.
Article in English | MEDLINE | ID: mdl-35038579

ABSTRACT

Despite rapid development and deployment of vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), clinically relevant modalities to curb the pandemic by directly attacking the virus on a genetic level remain highly desirable and are urgently needed. Here we comprehensively illustrate the capacity of adeno-associated virus (AAV) vectors co-expressing a cocktail of three short hairpin RNAs (shRNAs; RNAi triggers) directed against the SARS-CoV-2 RdRp and N genes as versatile and effective antiviral agents. In cultured monkey cells and human gut organoids, our most potent vector, SAVIOR (SARS virus repressor), suppressed SARS-CoV-2 infection to background levels. Strikingly, in control experiments using single shRNAs, multiple SARS-CoV-2 escape mutants quickly emerged from infected cells within 24-48 h. Importantly, such adverse viral adaptation was fully prevented with the triple-shRNA AAV vector even during long-term cultivation. In addition, AAV-SAVIOR efficiently purged SARS-CoV-2 in a new model of chronically infected human intestinal cells. Finally, intranasal AAV-SAVIOR delivery using an AAV9 capsid moderately diminished viral loads and/or alleviated disease symptoms in hACE2-transgenic or wild-type mice infected with human or mouse SARS-CoV-2 strains, respectively. Our combinatorial and customizable AAV/RNAi vector complements ongoing global efforts to control the coronavirus disease 2019 (COVID-19) pandemic and holds great potential for clinical translation as an original and flexible preventive or therapeutic antiviral measure.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Antiviral Agents , COVID-19/prevention & control , Dependovirus , Mice , Pandemics , RNA Interference , RNA, Small Interfering/genetics , SARS-CoV-2/genetics
13.
Nucleic Acids Res ; 49(5): e29, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33330940

ABSTRACT

Optogenetic control of CRISPR-Cas9 systems has significantly improved our ability to perform genome perturbations in living cells with high precision in time and space. As new Cas orthologues with advantageous properties are rapidly being discovered and engineered, the need for straightforward strategies to control their activity via exogenous stimuli persists. The Cas9 from Neisseria meningitidis (Nme) is a particularly small and target-specific Cas9 orthologue, and thus of high interest for in vivo genome editing applications. Here, we report the first optogenetic tool to control NmeCas9 activity in mammalian cells via an engineered, light-dependent anti-CRISPR (Acr) protein. Building on our previous Acr engineering work, we created hybrids between the NmeCas9 inhibitor AcrIIC3 and the LOV2 blue light sensory domain from Avena sativa. Two AcrIIC3-LOV2 hybrids from our collection potently blocked NmeCas9 activity in the dark, while permitting robust genome editing at various endogenous loci upon blue light irradiation. Structural analysis revealed that, within these hybrids, the LOV2 domain is located in striking proximity to the Cas9 binding surface. Together, our work demonstrates optogenetic regulation of a type II-C CRISPR effector and might suggest a new route for the design of optogenetic Acrs.


Subject(s)
CRISPR-Associated Protein 9/antagonists & inhibitors , CRISPR-Associated Protein 9/chemistry , CRISPR-Cas Systems , Gene Editing/methods , Neisseria meningitidis/enzymology , Optogenetics/methods , Cell Line , HEK293 Cells , Humans , Light , Models, Molecular , Protein Engineering , Proteins/chemistry , Proteins/radiation effects
14.
Nat Chem Biol ; 16(7): 725-730, 2020 07.
Article in English | MEDLINE | ID: mdl-32284602

ABSTRACT

Anti-CRISPR (Acr) proteins are powerful tools to control CRISPR-Cas technologies. However, the available Acr repertoire is limited to naturally occurring variants. Here, we applied structure-based design on AcrIIC1, a broad-spectrum CRISPR-Cas9 inhibitor, to improve its efficacy on different targets. We first show that inserting exogenous protein domains into a selected AcrIIC1 surface site dramatically enhances inhibition of Neisseria meningitidis (Nme)Cas9. Then, applying structure-guided design to the Cas9-binding surface, we converted AcrIIC1 into AcrIIC1X, a potent inhibitor of the Staphylococcus aureus (Sau)Cas9, an orthologue widely applied for in vivo genome editing. Finally, to demonstrate the utility of AcrIIC1X for genome engineering applications, we implemented a hepatocyte-specific SauCas9 ON-switch by placing AcrIIC1X expression under regulation of microRNA-122. Our work introduces designer Acrs as important biotechnological tools and provides an innovative strategy to safeguard CRISPR technologies.


Subject(s)
CRISPR-Associated Protein 9/genetics , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Gene Editing/methods , MicroRNAs/genetics , Protein Engineering/methods , Amino Acid Sequence , CRISPR-Associated Protein 9/metabolism , Cell Line, Tumor , Genome, Human , HEK293 Cells , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , MicroRNAs/metabolism , Models, Molecular , Mutagenesis, Insertional , Neisseria meningitidis/enzymology , Neisseria meningitidis/genetics , Plasmids/chemistry , Plasmids/metabolism , Protein Domains , Protein Structure, Secondary , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism , Staphylococcus aureus/enzymology , Staphylococcus aureus/genetics
15.
Mol Ther ; 29(12): 3359-3382, 2021 12 01.
Article in English | MEDLINE | ID: mdl-33831556

ABSTRACT

Parvoviruses and especially the adeno-associated virus (AAV) species provide an exciting and versatile platform for the rational design or molecular evolution of human gene-therapy vectors, documented by literature from over half a century, hundreds of clinical trials, and the recent commercialization of multiple AAV gene therapeutics. For the last three decades, the power of these vectors has been further potentiated through various types of hybrid vectors created by intra- or inter-genus juxtaposition of viral DNA and protein cis elements or by synergistic complementation of parvoviral features with those of heterologous, prokaryotic, or eukaryotic viruses. Here, we provide an overview of the history and promise of this rapidly expanding field of hybrid parvoviral gene-therapy vectors, starting with early generations of chimeric particles composed of a recombinant AAV genome encapsidated in shells of synthetic AAVs or of adeno-, herpes-, baculo-, or protoparvoviruses. We then dedicate our attention to two newer, highly promising types of hybrid vectors created via (1) pseudotyping of AAV genomes with bocaviral serotypes and capsid mutants or (2) packaging of AAV DNA into, or tethering of entire vector particles to, bacteriophages. Finally, we conclude with an outlook summarizing critical requirements and improvements toward clinical translation of these original concepts.


Subject(s)
Genetic Vectors , Parvovirus , DNA, Viral , Dependovirus/genetics , Dependovirus/metabolism , Genetic Therapy , Genetic Vectors/genetics , Humans , Parvovirus/genetics
16.
Nucleic Acids Res ; 48(1): e2, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31680162

ABSTRACT

The lack of endogenous RNAi machinery in the malaria parasite Plasmodium hampers gene annotation and hence antimalarial drug and vaccine development. Here, we engineered rodent Plasmodium berghei to express a minimal, non-canonical RNAi machinery that solely requires Argonaute 2 (Ago2) and a modified short hairpin RNA, so-called AgoshRNA. Using this strategy, we achieved robust and specific gene knockdown throughout the entire parasite life cycle. We also successfully silenced the endogenous gene perforin-like protein 2, phenocopying a full gene knockout. Transcriptionally restricting Ago2 expression to the liver stage further enabled us to perform a stage-specific gene knockout. The RNAi-competent Plasmodium lines reported here will be a valuable resource for loss-of-function phenotyping of the many uncharacterized genes of Plasmodium in low or high throughput, without the need to engineer the target gene locus. Thereby, our new strategy and transgenic Plasmodium lines will ultimately benefit the discovery of urgently needed antimalarial drug and vaccine candidates. Generally, the ability to render RNAi-negative organisms RNAi-competent by mere introduction of two components, Ago2 and AgoshRNA, is a unique paradigm that should find broad applicability in other species.


Subject(s)
Argonaute Proteins/genetics , Genetic Engineering/methods , Plasmodium berghei/genetics , Protozoan Proteins/genetics , RNA Interference , RNA, Small Interfering/genetics , Animals , Anopheles/parasitology , Argonaute Proteins/metabolism , Female , Genes, Reporter , Green Fluorescent Proteins/antagonists & inhibitors , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Life Cycle Stages/genetics , Mice , Mice, Inbred C57BL , Mosquito Vectors/parasitology , Organisms, Genetically Modified , Perforin/genetics , Perforin/metabolism , Plasmodium berghei/growth & development , Plasmodium berghei/metabolism , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/metabolism , RNA, Small Interfering/metabolism , Transgenes
17.
Pflugers Arch ; 473(3): 533-546, 2021 03.
Article in English | MEDLINE | ID: mdl-33580817

ABSTRACT

The cation channel transient receptor potential melastatin 4 (TRPM4) is a calcium-activated non-selective cation channel and acts in cardiomyocytes as a negative modulator of the L-type Ca2+ influx. Global deletion of TRPM4 in the mouse led to increased cardiac contractility under ß-adrenergic stimulation. Consequently, cardiomyocyte-specific inactivation of the TRPM4 function appears to be a promising strategy to improve cardiac contractility in heart failure patients. The aim of this study was to develop a gene therapy approach in mice that specifically silences the expression of TRPM4 in cardiomyocytes. First, short hairpin RNAmiR30 (shRNAmiR30) sequences against the TRPM4 mRNA were screened in vitro using lentiviral transduction for a stable expression of the shRNA cassettes. Western blot analysis identified three efficient shRNAmiR30 sequences out of six, which reduced the endogenous TRPM4 protein level by up to 90 ± 6%. Subsequently, the most efficient shRNAmiR30 sequences were delivered into cardiomyocytes of adult mice using adeno-associated virus serotype 9 (AAV9)-mediated gene transfer. Initially, the AAV9 vector particles were administered via the lateral tail vein, which resulted in a downregulation of TRPM4 by 46 ± 2%. Next, various optimization steps were carried out to improve knockdown efficiency in vivo. First, the design of the expression cassette was streamlined for integration in a self-complementary AAV vector backbone for a faster expression. Compared to the application via the lateral tail vein, intravenous application via the retro-orbital sinus has the advantage that the vector solution reaches the heart directly and in a high concentration, and eventually a TRPM4 knockdown efficiency of 90 ± 7% in the heart was accomplished by this approach. By optimization of the shRNAmiR30 constructs and expression cassette as well as the route of AAV9 vector application, a 90% reduction of TRPM4 expression was achieved in the adult mouse heart. In the future, AAV9-RNAi-mediated inactivation of TRPM4 could be a promising strategy to increase cardiac contractility in preclinical animal models of acute and chronic forms of cardiac contractile failure.


Subject(s)
Gene Transfer Techniques , Myocytes, Cardiac/metabolism , TRPM Cation Channels , Animals , Dependovirus , Genetic Vectors , Male , Mice , RNA Interference , RNA, Small Interfering , Transduction, Genetic/methods
18.
Hum Mol Genet ; 28(R1): R3-R14, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31261383

ABSTRACT

Recombinant adeno-associated viruses (AAV) are under intensive investigation in numerous clinical trials after they have emerged as a highly promising vector for human gene therapy. Best exemplifying their power and potential is the authorization of three gene therapy products based on wild-type AAV serotypes, comprising Glybera (AAV1), Luxturna (AAV2) and, most recently, Zolgensma (AAV9). Nonetheless, it has also become evident that the current AAV vector generation will require improvements in transduction potency, antibody evasion and cell/tissue specificity to allow the use of lower and safer vector doses. To this end, others and we devoted substantial previous research to the implementation and application of key technologies for engineering of next-generation viral capsids in a high-throughput 'top-down' or (semi-)rational 'bottom-up' approach. Here, we describe a set of recent complementary strategies to enhance features of AAV vectors that act on the level of the recombinant cargo. As examples that illustrate the innovative and synergistic concepts that have been reported lately, we highlight (i) novel synthetic enhancers/promoters that provide an unprecedented degree of AAV tissue specificity, (ii) pioneering genetic circuit designs that harness biological (microRNAs) or physical (light) triggers as regulators of AAV gene expression and (iii) new insights into the role of AAV DNA structures on vector genome stability, integrity and functionality. Combined with ongoing capsid engineering and selection efforts, these and other state-of-the-art innovations and investigations promise to accelerate the arrival of the next generation of AAV vectors and to solidify the unique role of this exciting virus in human gene therapy.


Subject(s)
Dependovirus/genetics , Genetic Vectors/genetics , Animals , Enhancer Elements, Genetic , Gene Expression Regulation , Genetic Engineering , Genetic Therapy , Humans , Promoter Regions, Genetic , Transduction, Genetic , Transgenes
19.
Gastroenterology ; 158(6): 1762-1775.e9, 2020 05.
Article in English | MEDLINE | ID: mdl-32001321

ABSTRACT

BACKGROUND & AIMS: Hepatitis B virus (HBV) infection persists because the virus-specific immune response is dysfunctional. Therapeutic vaccines might be used to end immune tolerance to the virus in patients with chronic infection, but these have not been effective in patients so far. In patients with chronic HBV infection, high levels of virus antigens might prevent induction of HBV-specific immune responses. We investigated whether knocking down expression levels of HBV antigens in liver might increase the efficacy of HBV vaccines in mice. METHODS: We performed studies with male C57BL/6 mice that persistently replicate HBV (genotype D, serotype ayw)-either from a transgene or after infection with an adeno-associated virus that transferred an overlength HBV genome-and expressed HB surface antigen at levels relevant to patients. Small hairpin or small interfering (si)RNAs against the common 3'-end of all HBV transcripts were used to knock down antigen expression in mouse hepatocytes. siRNAs were chemically stabilized and conjugated to N-acetylgalactosamine to increase liver uptake. Control mice were given either entecavir or non-HBV-specific siRNAs and vaccine components. Eight to 12 weeks later, mice were immunized twice with a mixture of adjuvanted HBV S and core antigen, followed by a modified Vaccinia virus Ankara vector to induce HBV-specific B- and T-cell responses. Serum and liver samples were collected and analyzed for HBV-specific immune responses, liver damage, and viral parameters. RESULTS: In both models of HBV infection, mice that express hepatocyte-specific small hairpin RNAs or that were given subcutaneous injections of siRNAs had reduced levels of HBV antigens, HBV replication, and viremia (1-3 log10 reduction) compared to mice given control RNAs. Vaccination induced production of HBV-neutralizing antibodies and increased numbers and functionality of HBV-specific, CD8+ T cells in mice with low, but not in mice with high, levels of HBV antigen. Mice with initially high titers of HBV and knockdown of HBV antigen expression, but not mice with reduced viremia after administration of entecavir, developed polyfunctional, HBV-specific CD8+ T cells, and HBV was eliminated. CONCLUSIONS: In mice with high levels of HBV replication, knockdown of HBV antigen expression along with a therapeutic vaccination strategy, but not knockdown alone, increased numbers of effector T cells and eliminated the virus. These findings indicate that high titers of virus antigens reduce the efficacy of therapeutic vaccination. Anti-HBV siRNAs and therapeutic vaccines are each being tested in clinical trials-their combination might cure chronic HBV infection.


Subject(s)
Hepatitis B Surface Antigens/genetics , Hepatitis B Vaccines/immunology , Hepatitis B virus/immunology , Hepatitis B, Chronic/therapy , RNA, Small Interfering/administration & dosage , Animals , B-Lymphocytes/immunology , Carrier State/immunology , Carrier State/virology , Combined Modality Therapy/methods , Disease Models, Animal , Female , Gene Knockdown Techniques , Hepatitis B Surface Antigens/immunology , Hepatitis B Vaccines/administration & dosage , Hepatitis B virus/genetics , Hepatitis B virus/isolation & purification , Hepatitis B, Chronic/diagnosis , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/virology , Hepatocytes/virology , Humans , Immunization, Secondary , Immunogenicity, Vaccine , Male , Mice , T-Lymphocytes, Cytotoxic/immunology , Virus Replication/genetics , Virus Replication/immunology
20.
Nat Methods ; 15(11): 924-927, 2018 11.
Article in English | MEDLINE | ID: mdl-30377362

ABSTRACT

Anti-CRISPR proteins are powerful tools for CRISPR-Cas9 regulation; the ability to precisely modulate their activity could facilitate spatiotemporally confined genome perturbations and uncover fundamental aspects of CRISPR biology. We engineered optogenetic anti-CRISPR variants comprising hybrids of AcrIIA4, a potent Streptococcus pyogenes Cas9 inhibitor, and the LOV2 photosensor from Avena sativa. Coexpression of these proteins with CRISPR-Cas9 effectors enabled light-mediated genome and epigenome editing, and revealed rapid Cas9 genome targeting in human cells.


Subject(s)
Biosensing Techniques , CRISPR-Associated Proteins/antagonists & inhibitors , CRISPR-Cas Systems , Gene Editing , Optogenetics , Phototropins/chemistry , Protein Engineering , Epigenomics , Genome , HEK293 Cells , Humans , Light , Streptococcus pyogenes/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL