Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Nat Nanotechnol ; 18(10): 1241-1251, 2023 10.
Article in English | MEDLINE | ID: mdl-37430038

ABSTRACT

Crossing the blood-brain barrier in primates is a major obstacle for gene delivery to the brain. Adeno-associated viruses (AAVs) promise robust, non-invasive gene delivery from the bloodstream to the brain. However, unlike in rodents, few neurotropic AAVs efficiently cross the blood-brain barrier in non-human primates. Here we report on AAV.CAP-Mac, an engineered variant identified by screening in adult marmosets and newborn macaques, which has improved delivery efficiency in the brains of multiple non-human primate species: marmoset, rhesus macaque and green monkey. CAP-Mac is neuron biased in infant Old World primates, exhibits broad tropism in adult rhesus macaques and is vasculature biased in adult marmosets. We demonstrate applications of a single, intravenous dose of CAP-Mac to deliver functional GCaMP for ex vivo calcium imaging across multiple brain areas, or a cocktail of fluorescent reporters for Brainbow-like labelling throughout the macaque brain, circumventing the need for germline manipulations in Old World primates. As such, CAP-Mac is shown to have potential for non-invasive systemic gene transfer in the brains of non-human primates.


Subject(s)
Brain , Callithrix , Humans , Animals , Infant, Newborn , Chlorocebus aethiops , Macaca mulatta/genetics , Callithrix/genetics , Brain/physiology , Gene Transfer Techniques , Neurons , Genetic Vectors/genetics
2.
Res Sq ; 2023 Jan 13.
Article in English | MEDLINE | ID: mdl-36789432

ABSTRACT

Adeno-associated viruses (AAVs) promise robust gene delivery to the brain through non-invasive, intravenous delivery. However, unlike in rodents, few neurotropic AAVs efficiently cross the blood-brain barrier in non-human primates (NHPs). Here we describe AAV.CAP-Mac, an engineered variant identified by screening in adult marmosets and newborn macaques with improved efficiency in the brain of multiple NHP species: marmoset, rhesus macaque, and green monkey. CAP-Mac is neuron-biased in infant Old World primates, exhibits broad tropism in adult rhesus macaques, and is vasculature-biased in adult marmosets. We demonstrate applications of a single, intravenous dose of CAP-Mac to deliver (1) functional GCaMP for ex vivo calcium imaging across multiple brain areas, and (2) a cocktail of fluorescent reporters for Brainbow-like labeling throughout the macaque brain, circumventing the need for germline manipulations in Old World primates. Given its capabilities for systemic gene transfer in NHPs, CAP-Mac promises to help unlock non-invasive access to the brain.

3.
Mol Ther Methods Clin Dev ; 26: 331-342, 2022 Sep 08.
Article in English | MEDLINE | ID: mdl-35990749

ABSTRACT

Advances in adeno-associated virus (AAV) engineering have provided exciting new tools for research and potential solutions for gene therapy. However, the lung has not received the same tailored engineering as other major targets of debilitating genetic disorders. To address this, here we engineered the surface-exposed residues AA452-458 of AAV9 capsid proteins at the three-fold axis of symmetry and employed a Cre-transgenic-based screening platform to identify AAV capsids targeted to the lung after intravenous delivery in mice. Using a custom image processing pipeline to quantify transgene expression across whole tissue images, we found that one engineered variant, AAV9.452sub.LUNG1, displays dramatically improved transgene expression in lung tissue after systemic delivery in mice. This improved transduction extends to alveolar epithelial type II cells, expanding the toolbox for gene therapy research for diseases specific to the lung.

4.
Nat Neurosci ; 25(1): 106-115, 2022 01.
Article in English | MEDLINE | ID: mdl-34887588

ABSTRACT

Genetic intervention is increasingly being explored as a therapeutic option for debilitating disorders of the central nervous system. The safety and efficacy of gene therapies rely upon expressing a transgene in affected cells while minimizing off-target expression. Here we show organ-specific targeting of adeno-associated virus (AAV) capsids after intravenous delivery, which we achieved by employing a Cre-transgenic-based screening platform and sequential engineering of AAV-PHP.eB between the surface-exposed AA452 and AA460 of VP3. From this selection, we identified capsid variants that were enriched in the brain and targeted away from the liver in C57BL/6J mice. This tropism extends to marmoset (Callithrix jacchus), enabling robust, non-invasive gene delivery to the marmoset brain after intravenous administration. Notably, the capsids identified result in distinct transgene expression profiles within the brain, with one exhibiting high specificity to neurons. The ability to cross the blood-brain barrier with neuronal specificity in rodents and non-human primates enables new avenues for basic research and therapeutic possibilities unattainable with naturally occurring serotypes.


Subject(s)
Capsid , Dependovirus , Administration, Intravenous , Animals , Brain/metabolism , Callithrix/genetics , Dependovirus/genetics , Genetic Vectors , Liver , Mice , Mice, Inbred C57BL , Transduction, Genetic , Transgenes
5.
Nat Commun ; 11(1): 2102, 2020 04 30.
Article in English | MEDLINE | ID: mdl-32355221

ABSTRACT

Adeno-associated viruses (AAVs) are typically single-stranded deoxyribonucleic acid (ssDNA) encapsulated within 25-nm protein capsids. Recently, tissue-specific AAV capsids (e.g. PHP.eB) have been shown to enhance brain delivery in rodents via the LY6A receptor on brain endothelial cells. Here, we create a non-invasive positron emission tomography (PET) methodology to track viruses. To provide the sensitivity required to track AAVs injected at picomolar levels, a unique multichelator construct labeled with a positron emitter (Cu-64, t1/2 = 12.7 h) is coupled to the viral capsid. We find that brain accumulation of the PHP.eB capsid 1) exceeds that reported in any previous PET study of brain uptake of targeted therapies and 2) is correlated with optical reporter gene transduction of the brain. The PHP.eB capsid brain endothelial receptor affinity is nearly 20-fold greater than that of AAV9. The results suggest that novel PET imaging techniques can be applied to inform and optimize capsid design.


Subject(s)
Brain/diagnostic imaging , Dependovirus/isolation & purification , Positron-Emission Tomography , Animals , Capsid , Chelating Agents/pharmacokinetics , Copper Radioisotopes/pharmacokinetics , Female , Genetic Vectors , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Transduction, Genetic
7.
Nat Protoc ; 10(11): 1860-1896, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26492141

ABSTRACT

To facilitate fine-scale phenotyping of whole specimens, we describe here a set of tissue fixation-embedding, detergent-clearing and staining protocols that can be used to transform excised organs and whole organisms into optically transparent samples within 1-2 weeks without compromising their cellular architecture or endogenous fluorescence. PACT (passive CLARITY technique) and PARS (perfusion-assisted agent release in situ) use tissue-hydrogel hybrids to stabilize tissue biomolecules during selective lipid extraction, resulting in enhanced clearing efficiency and sample integrity. Furthermore, the macromolecule permeability of PACT- and PARS-processed tissue hybrids supports the diffusion of immunolabels throughout intact tissue, whereas RIMS (refractive index matching solution) grants high-resolution imaging at depth by further reducing light scattering in cleared and uncleared samples alike. These methods are adaptable to difficult-to-image tissues, such as bone (PACT-deCAL), and to magnified single-cell visualization (ePACT). Together, these protocols and solutions enable phenotyping of subcellular components and tracing cellular connectivity in intact biological networks.


Subject(s)
Histocytochemistry/methods , Optical Imaging/methods , Pathology/methods , Specimen Handling/methods , Animals , Detergents/isolation & purification , Lipids/isolation & purification , Mice , Rats , Staining and Labeling/methods , Time Factors , Tissue Embedding/methods , Tissue Fixation/methods
8.
J Mol Biol ; 427(1): 205-20, 2015 Jan 16.
Article in English | MEDLINE | ID: mdl-24979679

ABSTRACT

Proton-pumping rhodopsins (PPRs) are photoactive retinal-binding proteins that transport ions across biological membranes in response to light. These proteins are interesting for light-harvesting applications in bioenergy production, in optogenetics applications in neuroscience, and as fluorescent sensors of membrane potential. Little is known, however, about how the protein sequence determines the considerable variation in spectral properties of PPRs from different biological niches or how to engineer these properties in a given PPR. Here we report a comprehensive study of amino acid substitutions in the retinal-binding pocket of Gloeobacter violaceus rhodopsin (GR) that tune its spectral properties. Directed evolution generated 70 GR variants with absorption maxima shifted by up to ±80nm, extending the protein's light absorption significantly beyond the range of known natural PPRs. While proton-pumping activity was disrupted in many of the spectrally shifted variants, we identified single tuning mutations that incurred blue and red shifts of 42nm and 22nm, respectively, that did not disrupt proton pumping. Blue-shifting mutations were distributed evenly along the retinal molecule while red-shifting mutations were clustered near the residue K257, which forms a covalent bond with retinal through a Schiff base linkage. Thirty eight of the identified tuning mutations are not found in known microbial rhodopsins. We discovered a subset of red-shifted GRs that exhibit high levels of fluorescence relative to the WT (wild-type) protein.


Subject(s)
Cyanobacteria/metabolism , Directed Molecular Evolution , Proton Pumps/metabolism , Rhodopsins, Microbial/chemistry , Rhodopsins, Microbial/metabolism , Amino Acid Sequence , Amino Acid Substitution , Cyanobacteria/genetics , Cyanobacteria/growth & development , Fluorescence , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation/genetics , Rhodopsins, Microbial/genetics , Sequence Homology, Amino Acid
9.
Nat Commun ; 5: 4894, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25222271

ABSTRACT

Probing the neural circuit dynamics underlying behaviour would benefit greatly from improved genetically encoded voltage indicators. The proton pump Archaerhodopsin-3 (Arch), an optogenetic tool commonly used for neuronal inhibition, has been shown to emit voltage-sensitive fluorescence. Here we report two Arch variants with enhanced radiance (Archers) that in response to 655 nm light have 3-5 times increased fluorescence and 55-99 times reduced photocurrents compared with Arch WT. The most fluorescent variant, Archer1, has 25-40% fluorescence change in response to action potentials while using 9 times lower light intensity compared with other Arch-based voltage sensors. Archer1 is capable of wavelength-specific functionality as a voltage sensor under red light and as an inhibitory actuator under green light. As a proof-of-concept for the application of Arch-based sensors in vivo, we show fluorescence voltage sensing in behaving Caenorhabditis elegans. Archer1's characteristics contribute to the goal of all-optical detection and modulation of activity in neuronal networks in vivo.


Subject(s)
Action Potentials/physiology , Archaeal Proteins/chemistry , Helminth Proteins/chemistry , Nerve Tissue Proteins/chemistry , Neurons/chemistry , Animals , Archaeal Proteins/genetics , Caenorhabditis elegans/chemistry , Caenorhabditis elegans/metabolism , Gene Expression , Helminth Proteins/genetics , Hippocampus/chemistry , Hippocampus/metabolism , Light , Nerve Tissue Proteins/genetics , Neurons/metabolism , Optogenetics/methods , Patch-Clamp Techniques , Primary Cell Culture , Protein Isoforms/chemistry , Protein Isoforms/genetics , Rats , Rats, Wistar , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Spectrometry, Fluorescence
10.
Proc Natl Acad Sci U S A ; 111(36): 13034-9, 2014 Sep 09.
Article in English | MEDLINE | ID: mdl-25157169

ABSTRACT

Microbial rhodopsins are a diverse group of photoactive transmembrane proteins found in all three domains of life. A member of this protein family, Archaerhodopsin-3 (Arch) of halobacterium Halorubrum sodomense, was recently shown to function as a fluorescent indicator of membrane potential when expressed in mammalian neurons. Arch fluorescence, however, is very dim and is not optimal for applications in live-cell imaging. We used directed evolution to identify mutations that dramatically improve the absolute brightness of Arch, as confirmed biochemically and with live-cell imaging (in Escherichia coli and human embryonic kidney 293 cells). In some fluorescent Arch variants, the pK(a) of the protonated Schiff-base linkage to retinal is near neutral pH, a useful feature for voltage-sensing applications. These bright Arch variants enable labeling of biological membranes in the far-red/infrared and exhibit the furthest red-shifted fluorescence emission thus far reported for a fluorescent protein (maximal excitation/emission at ∼ 620 nm/730 nm).


Subject(s)
Archaeal Proteins/metabolism , Directed Molecular Evolution , Binding Sites , Cell Survival , Escherichia coli/metabolism , Fluorescence , Green Fluorescent Proteins/metabolism , HEK293 Cells , Halorubrum/metabolism , Humans , Mutant Proteins/metabolism , Mutation , Structural Homology, Protein
11.
PLoS Genet ; 7(8): e1002218, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21876675

ABSTRACT

Epithelial-mesenchymal transition (EMT), a mechanism important for embryonic development, plays a critical role during malignant transformation. While much is known about transcriptional regulation of EMT, alternative splicing of several genes has also been correlated with EMT progression, but the extent of splicing changes and their contributions to the morphological conversion accompanying EMT have not been investigated comprehensively. Using an established cell culture model and RNA-Seq analyses, we determined an alternative splicing signature for EMT. Genes encoding key drivers of EMT-dependent changes in cell phenotype, such as actin cytoskeleton remodeling, regulation of cell-cell junction formation, and regulation of cell migration, were enriched among EMT-associated alternatively splicing events. Our analysis suggested that most EMT-associated alternative splicing events are regulated by one or more members of the RBFOX, MBNL, CELF, hnRNP, or ESRP classes of splicing factors. The EMT alternative splicing signature was confirmed in human breast cancer cell lines, which could be classified into basal and luminal subtypes based exclusively on their EMT-associated splicing pattern. Expression of EMT-associated alternative mRNA transcripts was also observed in primary breast cancer samples, indicating that EMT-dependent splicing changes occur commonly in human tumors. The functional significance of EMT-associated alternative splicing was tested by expression of the epithelial-specific splicing factor ESRP1 or by depletion of RBFOX2 in mesenchymal cells, both of which elicited significant changes in cell morphology and motility towards an epithelial phenotype, suggesting that splicing regulation alone can drive critical aspects of EMT-associated phenotypic changes. The molecular description obtained here may aid in the development of new diagnostic and prognostic markers for analysis of breast cancer progression.


Subject(s)
Alternative Splicing , Breast Neoplasms/genetics , Epithelial-Mesenchymal Transition/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement/genetics , Female , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Phenotype , RNA Splicing Factors , RNA-Binding Proteins/genetics , Repressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...