Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
NPJ Parkinsons Dis ; 9(1): 91, 2023 Jun 15.
Article in English | MEDLINE | ID: mdl-37322068

ABSTRACT

Prion-like transmission of pathology in α-synucleinopathies like Parkinson's disease or multiple system atrophy is increasingly recognized as one potential mechanism to address disease progression. Active and passive immunotherapies targeting insoluble, aggregated α-synuclein are already being actively explored in the clinic with mixed outcomes so far. Here, we report the identification of 306C7B3, a highly selective, aggregate-specific α-synuclein antibody with picomolar affinity devoid of binding to the monomeric, physiologic protein. 306C7B3 binding is Ser129-phosphorylation independent and shows high affinity to several different aggregated α-synuclein polymorphs, increasing the likelihood that it can also bind to the pathological seeds assumed to drive disease progression in patients. In support of this, highly selective binding to pathological aggregates in postmortem brains of MSA patients was demonstrated, with no staining in samples from other human neurodegenerative diseases. To achieve CNS exposure of 306C7B3, an adeno-associated virus (AAV) based approach driving expression of the secreted antibody within the brain of (Thy-1)-[A30P]-hα-synuclein mice was used. Widespread central transduction after intrastriatal inoculation was ensured by using the AAV2HBKO serotype, with transduction being spread to areas far away from the inoculation site. Treatment of (Thy-1)-[A30P]-hα-synuclein mice at the age of 12 months demonstrated significantly increased survival, with 306C7B3 concentration reaching 3.9 nM in the cerebrospinal fluid. These results suggest that AAV-mediated expression of 306C7B3, targeting extracellular, presumably disease-propagating aggregates of α-synuclein, has great potential as a disease-modifying therapy for α-synucleinopathies as it ensures CNS exposure of the antibody, thereby mitigating the selective permeability of the blood-brain barrier.

2.
Mol Ther Methods Clin Dev ; 25: 360-369, 2022 Jun 09.
Article in English | MEDLINE | ID: mdl-35573045

ABSTRACT

Adeno-associated virus (AAV) vector applications are often limited by capsid-directed humoral immune responses, mainly through neutralizing antibodies (NAbs), which are present throughout the human population due to natural AAV infections. Currently, antibody levels are often quantified via ELISA-based protocols or by cellular NAb assays and less frequently by in vivo NAb assays in mice. These methods need optimization for each serotype and are often not applicable to AAV variants with poor in vitro transduction. To tackle these limitations, we have established Meso Scale Discovery (MSD)-based assays for the quantification of binding antibodies (BAbs) and NAbs against the three most commonly used AAV serotypes, AAV2, AAV8, and AAV9. Both assays detect anti-AAV-IgG1-3 with high sensitivity and consistency as shown in a screen of sera from 40 healthy human donors. Subsequently, BAb and NAb titers were determined for identification of seronegative animals in a non-human primate (NHP) cohort. Moreover, the MSD-based BAb assay protocol was extended to a panel of 14 different AAV serotypes. In summary, our platform allows a rapid and quantitative assessment of the immunological properties of any natural or engineered AAV variant irrespective of transduction efficiency and enables high-throughput screens.

3.
Hum Mutat ; 32(10): E2283-93, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21913284

ABSTRACT

Benign familial neonatal seizures (BFNS) are a dominant epilepsy syndrome caused by mutations in the voltage-gated potassium channels K(V) 7.2 and K(V) 7.3. We examined the molecular pathomechanism of a BFNS-causing mutation (p.N258S) in the extracellular S5-H5 loop of K(V) 7.2. Wild type (WT) and mutant channels, expressed in both Xenopus laevis oocytes and CHO cells, were studied using electrophysiological techniques. The results revealed a pronounced loss-of-function with a dominant-negative effect of the mutant on WT K(V) 7.2 and K(V) 7.3 channels. Since single-channel recordings of K(V) 7.3-K(V) 7.2 and K(V) 7.3-N285S concatemers showed similar properties for both constructs, we hypothesized that the observed reduction in current amplitude was due to a folding and trafficking defect, which was confirmed by biochemical and immunocytochemical experiments revealing a reduced number of mutant channels in the surface membrane. Furthermore, rescuing experiments revealed that upon specific incubation of transfected CHO cells-either at lower temperatures of <30°C or in presence of the agonist retigabine (RTG)-the N258S-derived currents increased fivefold in contrast to the WT. The obtained results represent a first example of temperature and pharmacological rescue of a K(V) 7 mutation and suggest a folding and trafficking deficiency as the cause of reduced current amplitudes with a dominant-negative effect of N258S mutant proteins.


Subject(s)
Epilepsy, Benign Neonatal/genetics , KCNQ2 Potassium Channel/genetics , Mutation , Temperature , Animals , Anthracenes/pharmacology , CHO Cells , Cricetinae , Gene Expression , Humans , KCNQ2 Potassium Channel/antagonists & inhibitors , KCNQ2 Potassium Channel/chemistry , Oocytes , Potassium Channel Blockers/pharmacology , Protein Folding/drug effects , Protein Transport/genetics
4.
J Clin Invest ; 118(6): 2157-68, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18451999

ABSTRACT

Paroxysmal dyskinesias are episodic movement disorders that can be inherited or are sporadic in nature. The pathophysiology underlying these disorders remains largely unknown but may involve disrupted ion homeostasis due to defects in cell-surface channels or nutrient transporters. In this study, we describe a family with paroxysmal exertion-induced dyskinesia (PED) over 3 generations. Their PED was accompanied by epilepsy, mild developmental delay, reduced CSF glucose levels, hemolytic anemia with echinocytosis, and altered erythrocyte ion concentrations. Using a candidate gene approach, we identified a causative deletion of 4 highly conserved amino acids (Q282_S285del) in the pore region of the glucose transporter 1 (GLUT1). Functional studies in Xenopus oocytes and human erythrocytes revealed that this mutation decreased glucose transport and caused a cation leak that alters intracellular concentrations of sodium, potassium, and calcium. We screened 4 additional families, in which PED is combined with epilepsy, developmental delay, or migraine, but not with hemolysis or echinocytosis, and identified 2 additional GLUT1 mutations (A275T, G314S) that decreased glucose transport but did not affect cation permeability. Combining these data with brain imaging studies, we propose that the dyskinesias result from an exertion-induced energy deficit that may cause episodic dysfunction of the basal ganglia, and that the hemolysis with echinocytosis may result from alterations in intracellular electrolytes caused by a cation leak through mutant GLUT1.


Subject(s)
Anemia, Hemolytic/etiology , Anemia, Hemolytic/genetics , Cations , Chorea/genetics , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/physiology , Glucose/metabolism , Adult , Amino Acid Sequence , Animals , Chorea/pathology , Erythrocytes/metabolism , Female , Humans , Male , Models, Biological , Molecular Sequence Data , Physical Exertion , Xenopus
5.
Brain ; 133(Pt 5): 1403-14, 2010 May.
Article in English | MEDLINE | ID: mdl-20371507

ABSTRACT

Many idiopathic epilepsy syndromes have a characteristic age dependence, the underlying molecular mechanisms of which are largely unknown. Here we propose a mechanism that can explain that epileptic spells in benign familial neonatal-infantile seizures occur almost exclusively during the first days to months of life. Benign familial neonatal-infantile seizures are caused by mutations in the gene SCN2A encoding the voltage-gated Na(+) channel Na(V)1.2. We identified two novel SCN2A mutations causing benign familial neonatal-infantile seizures and analysed the functional consequences of these mutations in a neonatal and an adult splice variant of the human Na(+) channel Na(V)1.2 expressed heterologously in tsA201 cells together with beta1 and beta2 subunits. We found significant gating changes leading to a gain-of-function, such as an increased persistent Na(+) current, accelerated recovery from fast inactivation or altered voltage-dependence of steady-state activation. Those were restricted to the neonatal splice variant for one mutation, but more pronounced for the adult form for the other, suggesting that a differential developmental splicing does not provide a general explanation for seizure remission. We therefore analysed the developmental expression of Na(V)1.2 and of another voltage-gated Na(+) channel, Na(V)1.6, using immunohistochemistry and real-time reverse transcription-polymerase chain reaction in mouse brain slices. We found that Na(V)1.2 channels are expressed early in development at axon initial segments of principal neurons in the hippocampus and cortex, but their expression is diminished and they are gradually replaced as the dominant channel type by Na(V)1.6 during maturation. This finding provides a plausible explanation for the transient expression of seizures that occur due to a gain-of-function of mutant Na(V)1.2 channels.


Subject(s)
Epilepsy, Benign Neonatal/genetics , Mutation , Nerve Tissue Proteins/genetics , Sodium Channels/genetics , Adult , Aging/metabolism , Animals , Axons/metabolism , Base Sequence , Cell Line , Electrophysiology , Epilepsy, Benign Neonatal/physiopathology , Hippocampus/metabolism , Humans , Immunohistochemistry , Infant , Infant, Newborn , Male , Mice , Mice, Inbred C57BL , NAV1.2 Voltage-Gated Sodium Channel , NAV1.6 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/metabolism , Pedigree , Protein Isoforms , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sodium Channels/metabolism
6.
Nat Commun ; 11(1): 714, 2020 02 05.
Article in English | MEDLINE | ID: mdl-32024835

ABSTRACT

Synthetic riboswitches mediating ligand-dependent RNA cleavage or splicing-modulation represent elegant tools to control gene expression in various applications, including next-generation gene therapy. However, due to the limited understanding of context-dependent structure-function relationships, the identification of functional riboswitches requires large-scale-screening of aptamer-effector-domain designs, which is hampered by the lack of suitable cellular high-throughput methods. Here we describe a fast and broadly applicable method to functionally screen complex riboswitch libraries (~1.8 × 104 constructs) by cDNA-amplicon-sequencing in transiently transfected and stimulated human cells. The self-barcoding nature of each construct enables quantification of differential mRNA levels without additional pre-selection or cDNA-manipulation steps. We apply this method to engineer tetracycline- and guanine-responsive ON- and OFF-switches based on hammerhead, hepatitis-delta-virus and Twister ribozymes as well as U1-snRNP polyadenylation-dependent RNA devices. In summary, our method enables fast and efficient high-throughput riboswitch identification, thereby overcoming a major hurdle in the development cascade for therapeutically applicable gene switches.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Riboswitch/genetics , Computational Biology/methods , DNA Barcoding, Taxonomic , DNA, Complementary , Gene Expression Regulation/drug effects , Guanine/pharmacology , HEK293 Cells , Hepatitis Delta Virus/genetics , Humans , RNA, Catalytic/genetics , Ribonucleoprotein, U1 Small Nuclear/genetics , Riboswitch/drug effects , Synthetic Biology/methods , Tetracycline/pharmacology
7.
ACS Synth Biol ; 9(6): 1292-1305, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32427483

ABSTRACT

Adeno-associated viral (AAV) vector-mediated gene therapy holds great potential for future medical applications. However, to facilitate safer and broader applicability and to enable patient-centric care, therapeutic protein expression should be controllable, ideally by an orally administered drug. The use of protein-based systems is considered rather undesirable, due to potential immunogenicity and the limited coding space of AAV. Ligand-dependent riboswitches, in contrast, are small and characterized by an attractive mode-of-action based on mRNA-self-cleavage, independent of coexpressed foreign protein. While a promising approach, switches available to date have only shown moderate potency in animals. In particular, ON-switches that induce transgene expression upon ligand administration so far have achieved rather disappointing results. Here we present the utilization of the previously described tetracycline-dependent ribozyme K19 for controlling AAV-mediated transgene expression in mice. Using this tool switch, we provide first proof for the feasibility of clinically desired key features, including multiorgan functionality, potent regulation (up to 15-fold induction), reversibility, and the possibility to fine-tune and repeatedly induce expression. The systematic assessment of ligand and reporter protein plasma levels further enabled the characterization of pharmacokinetic-pharmacodynamic relationships. Thus, our results strongly support future efforts to develop engineered riboswitches for applications in clinical gene therapy.


Subject(s)
Anti-Bacterial Agents/pharmacology , Dependovirus/genetics , Gene Expression/drug effects , Genetic Vectors/metabolism , RNA, Catalytic/metabolism , 3' Untranslated Regions , Animals , Aptamers, Nucleotide/genetics , Aptamers, Nucleotide/metabolism , Cell Line , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Liver/metabolism , Lung/metabolism , Mice , RNA, Catalytic/genetics , Tetracycline/pharmacology
8.
Plant Physiol ; 134(1): 43-58, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14730064

ABSTRACT

Calcium signals mediate a multitude of plant responses to external stimuli and regulate a wide range of physiological processes. Calcium-binding proteins, like calcineurin B-like (CBL) proteins, represent important relays in plant calcium signaling. These proteins form a complex network with their target kinases being the CBL-interacting protein kinases (CIPKs). Here, we present a comparative genomics analysis of the full complement of CBLs and CIPKs in Arabidopsis and rice (Oryza sativa). We confirm the expression and transcript composition of the 10 CBLs and 25 CIPKs encoded in the Arabidopsis genome. Our identification of 10 CBLs and 30 CIPKs from rice indicates a similar complexity of this signaling network in both species. An analysis of the genomic evolution suggests that the extant number of gene family members largely results from segmental duplications. A phylogenetic comparison of protein sequences and intron positions indicates an early diversification of separate branches within both gene families. These branches may represent proteins with different functions. Protein interaction analyses and expression studies of closely related family members suggest that even recently duplicated representatives may fulfill different functions. This work provides a basis for a defined further functional dissection of this important plant-specific signaling system.


Subject(s)
Arabidopsis/genetics , Arabidopsis/metabolism , Calcium Signaling , Oryza/genetics , Oryza/metabolism , Protein Kinases/genetics , Protein Kinases/metabolism , Amino Acid Sequence , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Chromosome Mapping , Chromosomes, Plant/genetics , Evolution, Molecular , Exons , Genes, Plant , Genomics , Introns , Molecular Sequence Data , Multigene Family , Phylogeny , Sequence Homology, Amino Acid , Signal Transduction , Species Specificity
9.
Plant J ; 40(3): 428-38, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15469500

ABSTRACT

Dynamic networks of protein-protein interactions regulate numerous cellular processes and determine the ability to respond appropriately to environmental stimuli. However, the investigation of protein complex formation in living plant cells by methods such as fluorescence resonance energy transfer has remained experimentally difficult, time consuming and requires sophisticated technical equipment. Here, we report the implementation of a bimolecular fluorescence complementation (BiFC) technique for visualization of protein-protein interactions in plant cells. This approach relies on the formation of a fluorescent complex by two non-fluorescent fragments of the yellow fluorescent protein brought together by association of interacting proteins fused to these fragments (Hu et al., 2002). To enable BiFC analyses in plant cells, we generated different complementary sets of expression vectors, which enable protein interaction studies in transiently or stably transformed cells. These vectors were used to investigate and visualize homodimerization of the basic leucine zipper (bZIP) transcription factor bZIP63 and the zinc finger protein lesion simulating disease 1 (LSD1) from Arabidopsis as well as the dimer formation of the tobacco 14-3-3 protein T14-3c. The interaction analyses of these model proteins established the feasibility of BiFC analyses for efficient visualization of structurally distinct proteins in different cellular compartments. Our investigations revealed a remarkable signal fluorescence intensity of interacting protein complexes as well as a high reproducibility and technical simplicity of the method in different plant systems. Consequently, the BiFC approach should significantly facilitate the visualization of the subcellular sites of protein interactions under conditions that closely reflect the normal physiological environment.


Subject(s)
Arabidopsis/metabolism , Nicotiana/metabolism , Plant Proteins/metabolism , Spectrometry, Fluorescence/methods , Arabidopsis Proteins/metabolism , Bacterial Proteins , Basic-Leucine Zipper Transcription Factors , Cytoplasm/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Plant , Luminescent Proteins , Protein Binding , Protein Multimerization , Sensitivity and Specificity , Transcription Factors/metabolism
10.
Plant J ; 36(4): 457-70, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14617077

ABSTRACT

Calcium ions represent both an integrative signal and an important convergence point of many disparate signaling pathways. Calcium-binding proteins, like calcineurin B-like (CBL) proteins, have been implicated as important relays in calcium signaling. Here, we report the in vivo study of CBL1 function in Arabidopsis. Analyses of loss-of-function as well as CBL1-overexpressing lines indicate a crucial function of this calcium sensor protein in abiotic stress responses. Mutation of CBL1 impairs plant responses to drought and salt stresses and affects gene expression of cold-regulated genes, but does not affect abscisic acid (ABA) responsiveness. Conversely, overexpression of CBL1 reduces transpirational water loss and induces the expression of early stress-responsive transcription factors and stress adaptation genes in non-stressed plants. Together, our data indicate that the calcium sensor protein CBL1 may constitute an integrative node in plant responses to abiotic stimuli and contributes to the regulation of early stress-related transcription factors of the C-Repeat-Binding Factor/dehydration-responsive element (CBF/DREB) type.


Subject(s)
Arabidopsis Proteins/genetics , Arabidopsis/genetics , Calcium Signaling/genetics , Calcium-Binding Proteins/genetics , Calcium/metabolism , Abscisic Acid/pharmacology , Arabidopsis/metabolism , Arabidopsis Proteins/metabolism , Calcium Signaling/drug effects , Calcium-Binding Proteins/metabolism , Cold Temperature , DNA, Bacterial/chemistry , DNA, Bacterial/genetics , Disasters , Gene Expression Regulation, Plant , Mutation , Plant Transpiration/drug effects , Plants, Genetically Modified , Sodium Chloride/pharmacology , Stress, Mechanical , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL