Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 442
Filter
Add more filters

Country/Region as subject
Publication year range
1.
RNA ; 30(6): 624-643, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38413166

ABSTRACT

Antisense oligomer (ASO)-based antibiotics that target mRNAs of essential bacterial genes have great potential for counteracting antimicrobial resistance and for precision microbiome editing. To date, the development of such antisense antibiotics has primarily focused on using phosphorodiamidate morpholino (PMO) and peptide nucleic acid (PNA) backbones, largely ignoring the growing number of chemical modalities that have spurred the success of ASO-based human therapy. Here, we directly compare the activities of seven chemically distinct 10mer ASOs, all designed to target the essential gene acpP upon delivery with a KFF-peptide carrier into Salmonella. Our systematic analysis of PNA, PMO, phosphorothioate (PTO)-modified DNA, 2'-methylated RNA (RNA-OMe), 2'-methoxyethylated RNA (RNA-MOE), 2'-fluorinated RNA (RNA-F), and 2'-4'-locked RNA (LNA) is based on a variety of in vitro and in vivo methods to evaluate ASO uptake, target pairing and inhibition of bacterial growth. Our data show that only PNA and PMO are efficiently delivered by the KFF peptide into Salmonella to inhibit bacterial growth. Nevertheless, the strong target binding affinity and in vitro translational repression activity of LNA and RNA-MOE make them promising modalities for antisense antibiotics that will require the identification of an effective carrier.


Subject(s)
Anti-Bacterial Agents , Oligonucleotides, Antisense , Peptide Nucleic Acids , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Peptide Nucleic Acids/pharmacology , Peptide Nucleic Acids/chemistry , Oligonucleotides, Antisense/pharmacology , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/genetics , Morpholinos/chemistry , Morpholinos/pharmacology , Morpholinos/genetics , Peptides/pharmacology , Peptides/chemistry , Peptides/genetics , Humans
2.
Dev Biol ; 516: 59-70, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39098630

ABSTRACT

Dicer substrate interfering RNAs (DsiRNAs) destroy targeted transcripts using the RNA-Induced Silencing Complex (RISC) through a process called RNA interference (RNAi). This process is ubiquitous among eukaryotes. Here we report the utility of DsiRNA in embryos of the sea urchin Lytechinus variegatus (Lv). Specific knockdowns phenocopy known morpholino and inhibitor knockdowns, and DsiRNA offers a useful alternative to morpholinos. Methods are described for the design of specific DsiRNAs that lead to destruction of targeted mRNA. DsiRNAs directed against pks1, an enzyme necessary for pigment production, show how successful DsiRNA perturbations are monitored by RNA in situ analysis and by qPCR to determine relative destruction of targeted mRNA. DsiRNA-based knockdowns phenocopy morpholino- and drug-based inhibition of nodal and lefty. Other knockdowns demonstrate that the RISC operates early in development as well as on genes that are first transcribed hours after gastrulation is completed. Thus, DsiRNAs effectively mediate destruction of targeted mRNA in the sea urchin embryo. The approach offers significant advantages over other widely used methods in the urchin in terms of cost, and ease of procurement, and offers sizeable experimental advantages in terms of ease of handling, injection, and knockdown validation.


Subject(s)
Gene Knockdown Techniques , Nodal Protein , RNA Interference , Signal Transduction , Animals , Nodal Protein/metabolism , Nodal Protein/genetics , Signal Transduction/genetics , Gene Knockdown Techniques/methods , Sea Urchins/genetics , Sea Urchins/embryology , Gene Expression Regulation, Developmental , Morpholinos/genetics , Morpholinos/pharmacology , Embryo, Nonmammalian/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Lytechinus/genetics , Lytechinus/embryology , RNA, Small Interfering/metabolism , RNA, Small Interfering/genetics , Ribonuclease III/metabolism , Ribonuclease III/genetics
3.
Nucleic Acids Res ; 51(6): 2529-2573, 2023 04 11.
Article in English | MEDLINE | ID: mdl-36881759

ABSTRACT

Eighteen nucleic acid therapeutics have been approved for treatment of various diseases in the last 25 years. Their modes of action include antisense oligonucleotides (ASOs), splice-switching oligonucleotides (SSOs), RNA interference (RNAi) and an RNA aptamer against a protein. Among the diseases targeted by this new class of drugs are homozygous familial hypercholesterolemia, spinal muscular atrophy, Duchenne muscular dystrophy, hereditary transthyretin-mediated amyloidosis, familial chylomicronemia syndrome, acute hepatic porphyria, and primary hyperoxaluria. Chemical modification of DNA and RNA was central to making drugs out of oligonucleotides. Oligonucleotide therapeutics brought to market thus far contain just a handful of first- and second-generation modifications, among them 2'-fluoro-RNA, 2'-O-methyl RNA and the phosphorothioates that were introduced over 50 years ago. Two other privileged chemistries are 2'-O-(2-methoxyethyl)-RNA (MOE) and the phosphorodiamidate morpholinos (PMO). Given their importance in imparting oligonucleotides with high target affinity, metabolic stability and favorable pharmacokinetic and -dynamic properties, this article provides a review of these chemistries and their use in nucleic acid therapeutics. Breakthroughs in lipid formulation and GalNAc conjugation of modified oligonucleotides have paved the way to efficient delivery and robust, long-lasting silencing of genes. This review provides an account of the state-of-the-art of targeted oligo delivery to hepatocytes.


Subject(s)
Oligonucleotides, Antisense , Humans , Morpholinos/pharmacology , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/genetics , Oligonucleotides, Antisense/chemistry , Oligonucleotides, Antisense/metabolism , Oligonucleotides, Antisense/therapeutic use , RNA/chemistry , RNA Interference
4.
Proc Natl Acad Sci U S A ; 119(36): e2207956119, 2022 09 06.
Article in English | MEDLINE | ID: mdl-36037350

ABSTRACT

Recent advances in drug development have seen numerous successful clinical translations using synthetic antisense oligonucleotides (ASOs). However, major obstacles, such as challenging large-scale production, toxicity, localization of oligonucleotides in specific cellular compartments or tissues, and the high cost of treatment, need to be addressed. Thiomorpholino oligonucleotides (TMOs) are a recently developed novel nucleic acid analog that may potentially address these issues. TMOs are composed of a morpholino nucleoside joined by thiophosphoramidate internucleotide linkages. Unlike phosphorodiamidate morpholino oligomers (PMOs) that are currently used in various splice-switching ASO drugs, TMOs can be synthesized using solid-phase oligonucleotide synthesis methodologies. In this study, we synthesized various TMOs and evaluated their efficacy to induce exon skipping in a Duchenne muscular dystrophy (DMD) in vitro model using H2K mdx mouse myotubes. Our experiments demonstrated that TMOs can efficiently internalize and induce excellent exon 23 skipping potency compared with a conventional PMO control and other widely used nucleotide analogs, such as 2'-O-methyl and 2'-O-methoxyethyl ASOs. Notably, TMOs performed well at low concentrations (5-20 nM). Therefore, the dosages can be minimized, which may improve the drug safety profile. Based on the present study, we propose that TMOs represent a new, promising class of nucleic acid analogs for future oligonucleotide therapeutic development.


Subject(s)
Genetic Therapy , Morpholinos , Muscular Dystrophy, Duchenne , RNA Splicing , Animals , Disease Models, Animal , Genetic Therapy/methods , In Vitro Techniques , Mice , Mice, Inbred mdx , Morpholinos/genetics , Morpholinos/pharmacology , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/genetics , Oligonucleotides/genetics , Oligonucleotides/pharmacology , Oligonucleotides/therapeutic use , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , Oligonucleotides, Antisense/therapeutic use , RNA, Messenger
5.
Development ; 148(2)2021 01 25.
Article in English | MEDLINE | ID: mdl-33462117

ABSTRACT

The regulated expansion of chondrocytes within growth plates and joints ensures proper skeletal development through adulthood. Mutations in the transcription factor NKX3.2 underlie spondylo-megaepiphyseal-metaphyseal dysplasia (SMMD), which is characterized by skeletal defects including scoliosis, large epiphyses, wide growth plates and supernumerary distal limb joints. Whereas nkx3.2 knockdown zebrafish and mouse Nkx3.2 mutants display embryonic lethal jaw joint fusions and skeletal reductions, respectively, they lack the skeletal overgrowth seen in SMMD patients. Here, we report adult viable nkx3.2 mutant zebrafish displaying cartilage overgrowth in place of a missing jaw joint, as well as severe dysmorphologies of the facial skeleton, skullcap and spine. In contrast, cartilage overgrowth and scoliosis are absent in rare viable nkx3.2 knockdown animals that lack jaw joints, supporting post-embryonic roles for Nkx3.2. Single-cell RNA-sequencing and in vivo validation reveal increased proliferation and upregulation of stress-induced pathways, including prostaglandin synthases, in mutant chondrocytes. By generating a zebrafish model for the skeletal overgrowth defects of SMMD, we reveal post-embryonic roles for Nkx3.2 in dampening proliferation and buffering the stress response in joint-associated chondrocytes.


Subject(s)
Bone and Bones/embryology , Bone and Bones/metabolism , Homeodomain Proteins/metabolism , Osteochondrodysplasias/embryology , Transcription Factors/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Animals , Cartilage/embryology , Cartilage/pathology , Chondrocytes/metabolism , Disease Models, Animal , Embryo, Nonmammalian/abnormalities , Embryo, Nonmammalian/pathology , Gene Expression Regulation, Developmental , Jaw/embryology , Jaw/pathology , Joints/abnormalities , Joints/embryology , Joints/pathology , Mitosis/genetics , Morpholinos/pharmacology , Mutation/genetics , RNA-Seq , Single-Cell Analysis , Skull/abnormalities , Skull/embryology , Skull/pathology , Spine/abnormalities , Spine/embryology , Spine/pathology , Stress, Physiological/genetics , Up-Regulation/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
6.
Development ; 148(2)2021 01 20.
Article in English | MEDLINE | ID: mdl-33298464

ABSTRACT

During sea urchin development, secretion of Nodal and BMP2/4 ligands and their antagonists Lefty and Chordin from a ventral organiser region specifies the ventral and dorsal territories. This process relies on a complex interplay between the Nodal and BMP pathways through numerous regulatory circuits. To decipher the interplay between these pathways, we used a combination of treatments with recombinant Nodal and BMP2/4 proteins and a computational modelling approach. We assembled a logical model focusing on cell responses to signalling inputs along the dorsal-ventral axis, which was extended to cover ligand diffusion and enable multicellular simulations. Our model simulations accurately recapitulate gene expression in wild-type embryos, accounting for the specification of ventral ectoderm, ciliary band and dorsal ectoderm. Our model simulations further recapitulate various morphant phenotypes, reveal a dominance of the BMP pathway over the Nodal pathway and stress the crucial impact of the rate of Smad activation in dorsal-ventral patterning. These results emphasise the key role of the mutual antagonism between the Nodal and BMP2/4 pathways in driving early dorsal-ventral patterning of the sea urchin embryo.


Subject(s)
Body Patterning , Embryo, Nonmammalian/metabolism , Paracentrotus/embryology , Signal Transduction , Transforming Growth Factor beta/metabolism , Animals , Blastula/metabolism , Body Patterning/drug effects , Body Patterning/genetics , Bone Morphogenetic Proteins/metabolism , Cell Lineage/drug effects , Cell Lineage/genetics , Computer Simulation , Embryo, Nonmammalian/drug effects , Gene Expression Regulation, Developmental/drug effects , Glycoproteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Models, Biological , Morpholinos/pharmacology , Nodal Protein/metabolism , Paracentrotus/drug effects , Paracentrotus/genetics , Phenotype , Probability , Signal Transduction/drug effects , Signal Transduction/genetics , Stochastic Processes
7.
Nucleic Acids Res ; 50(20): 11401-11414, 2022 11 11.
Article in English | MEDLINE | ID: mdl-35944903

ABSTRACT

Current therapies for Duchenne muscular dystrophy (DMD) use phosphorodiamidate morpholino oligomers (PMO) to induce exon skipping in the dystrophin pre-mRNA, enabling the translation of a shortened but functional dystrophin protein. This strategy has been hampered by insufficient delivery of PMO to cardiac and skeletal muscle. To overcome these limitations, we developed the FORCETM platform consisting of an antigen-binding fragment, which binds the transferrin receptor 1, conjugated to an oligonucleotide. We demonstrate that a single dose of the mouse-specific FORCE-M23D conjugate enhances muscle delivery of exon skipping PMO (M23D) in mdx mice, achieving dose-dependent and robust exon skipping and durable dystrophin restoration. FORCE-M23D-induced dystrophin expression reached peaks of 51%, 72%, 62%, 90% and 77%, of wild-type levels in quadriceps, tibialis anterior, gastrocnemius, diaphragm, and heart, respectively, with a single 30 mg/kg PMO-equivalent dose. The shortened dystrophin localized to the sarcolemma, indicating expression of a functional protein. Conversely, a single 30 mg/kg dose of unconjugated M23D displayed poor muscle delivery resulting in marginal levels of exon skipping and dystrophin expression. Importantly, FORCE-M23D treatment resulted in improved functional outcomes compared with administration of unconjugated M23D. Our results suggest that FORCE conjugates are a potentially effective approach for the treatment of DMD.


The biggest problem confronting oligonucleotide therapeutics is a lack of compounds capable of targeting compounds to diseased tissues. This paper reports a major advance targeting the transferrin receptor to increase the delivery of morpholine oligomers to muscle cells in vivo. This work suggests the possibility for improved treatments of muscular dystrophy and other diseases.


Subject(s)
Dystrophin , Exons , Morpholinos , Muscular Dystrophy, Duchenne , Oligonucleotides, Antisense , Animals , Mice , Dystrophin/genetics , Exons/genetics , Mice, Inbred mdx , Morpholinos/pharmacology , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/therapy , Oligonucleotides, Antisense/pharmacology , Receptors, Transferrin/genetics
8.
Angew Chem Int Ed Engl ; 63(17): e202318773, 2024 04 22.
Article in English | MEDLINE | ID: mdl-38411401

ABSTRACT

Conditionally controlled antisense oligonucleotides provide precise interrogation of gene function at different developmental stages in animal models. Only one example of small molecule-induced activation of antisense function exist. This has been restricted to cyclic caged morpholinos that, based on sequence, can have significant background activity in the absence of the trigger. Here, we provide a new approach using azido-caged nucleobases that are site-specifically introduced into antisense morpholinos. The caging group design is a simple azidomethylene (Azm) group that, despite its very small size, efficiently blocks Watson-Crick base pairing in a programmable fashion. Furthermore, it undergoes facile decaging via Staudinger reduction when exposed to a small molecule phosphine, generating the native antisense oligonucleotide under conditions compatible with biological environments. We demonstrated small molecule-induced gene knockdown in mammalian cells, zebrafish embryos, and frog embryos. We validated the general applicability of this approach by targeting three different genes.


Subject(s)
Oligonucleotides , Zebrafish , Animals , Morpholinos/genetics , Morpholinos/pharmacology , Oligonucleotides, Antisense , Phenotype , Mammals
9.
Dev Biol ; 484: 63-74, 2022 04.
Article in English | MEDLINE | ID: mdl-35183512

ABSTRACT

Sea urchin larval skeletons are produced by skeletogenic primary mesenchyme cells (PMCs), which migrate to form two ventrolateral clusters (VLCs) at the sites where biomineralization is initiated. Both PMC migration and biomineralization are controlled by VEGF signals emitted from lateral ectodermal cells. In mammals, VEGF signaling can be activated by hypoxia-inducible factor alpha (HIFα), an oxygen-sensitive transcription factor. Our previous study showed that the sea urchin maternal HIFα is involved in regulating gene expression along the dorsoventral axis. In this study, we discovered that zygotic hifα is expressed in PMCs, and at the late gastrula stage, hifα transcripts display a graded pattern, with stronger signal in the ventral PMCs than in the dorsal PMCs. We further showed that PMCs are hypoxic, which is a condition typically required for HIFα function. In embryos injected with a splice-blocking morpholino against hifα, elongation of the skeleton was impaired, and expression of vegfr-10-Ig (encodes VEGF receptor; VEGFR) was significantly reduced. This morpholino-caused defect could be partially rescued by injection of vegfr-10-Ig mRNA. Expression patterns of transcription factor and biomineralization genes, such as alx1, tbr, msp130, and the sm30 family, were affected when HIFα was knocked down or when VEGF signaling was inhibited. These results suggest that zygotic HIFα acts upstream or in parallel with VEGF signaling to regulate skeletogenic gene expression and participate in spicule elongation. Our study therefore links HIFα with the known role of VEGF signaling in sea urchin biomineralization.


Subject(s)
Embryo, Nonmammalian , Vascular Endothelial Growth Factor A , Animals , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental , Hypoxia/metabolism , Mammals/genetics , Morpholinos/genetics , Morpholinos/metabolism , Morpholinos/pharmacology , Sea Urchins/genetics , Sea Urchins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Vascular Endothelial Growth Factor A/metabolism
10.
Exp Eye Res ; 226: 109333, 2023 01.
Article in English | MEDLINE | ID: mdl-36436570

ABSTRACT

Work in the catshark Scyliorhinus canicula has shown that the evolutionary origin of postnatal neurogenesis in vertebrates is earlier than previously thought. Thus, the catshark can serve as a model of interest to understand postnatal neurogenic processes and their evolution in vertebrates. One of the best characterized neurogenic niches of the catshark CNS is found in the peripheral region of the retina. Unfortunately, the lack of genetic tools in sharks limits the possibilities to deepen in the study of genes involved in the neurogenic process. Here, we report a method for gene knockdown in the juvenile catshark retina based on the use of Vivo-Morpholinos. To establish the method, we designed Vivo-Morpholinos against the proliferation marker PCNA. We first evaluated the possible toxicity of 3 different intraocular administration regimes. After this optimization step, we show that a single intraocular injection of the PCNA Vivo-Morpholino decreases the expression of PCNA in the peripheral retina, which leads to reduced mitotic activity in this region. This method will help in deciphering the role of other genes potentially involved in postnatal neurogenesis in this animal model.


Subject(s)
Sharks , Animals , Sharks/genetics , Sharks/metabolism , Morpholinos/genetics , Morpholinos/pharmacology , Morpholinos/metabolism , Gene Knockdown Techniques , Proliferating Cell Nuclear Antigen/genetics , Retina/metabolism
11.
Nat Rev Genet ; 18(1): 24-40, 2017 01.
Article in English | MEDLINE | ID: mdl-27795562

ABSTRACT

Our understanding of the genetic mechanisms that underlie biological processes has relied extensively on loss-of-function (LOF) analyses. LOF methods target DNA, RNA or protein to reduce or to ablate gene function. By analysing the phenotypes that are caused by these perturbations the wild-type function of genes can be elucidated. Although all LOF methods reduce gene activity, the choice of approach (for example, mutagenesis, CRISPR-based gene editing, RNA interference, morpholinos or pharmacological inhibition) can have a major effect on phenotypic outcomes. Interpretation of the LOF phenotype must take into account the biological process that is targeted by each method. The practicality and efficiency of LOF methods also vary considerably between model systems. We describe parameters for choosing the optimal combination of method and system, and for interpreting phenotypes within the constraints of each method.


Subject(s)
CRISPR-Cas Systems , Gene Silencing , Models, Animal , Morpholinos/pharmacology , Mutagenesis , Mutation/genetics , RNA Interference , Animals , Genotype , Humans , Phenotype , Species Specificity
12.
Dev Biol ; 475: 21-29, 2021 07.
Article in English | MEDLINE | ID: mdl-33684434

ABSTRACT

Echinoderms are important experimental models for analyzing embryonic development, but a lack of spatial and temporal control over gene perturbations has hindered developmental studies using these animals. Morpholino antisense oligonucleotides (MOs) have been used successfully by the echinoderm research community for almost two decades, and MOs remain the most widely used tool for acute gene knockdowns in these organisms. Echinoderm embryos develop externally and are optically transparent, making them ideally-suited to many light-based approaches for analyzing and manipulating development. Studies using zebrafish embryos have demonstrated the effectiveness of photoactivatable (caged) MOs for conditional gene knockdowns. Here we show that caged MOs, synthesized using nucleobase-caged monomers, provide light-regulated control over gene expression in sea urchin embryos. Our work provides the first robust approach for conditional gene silencing in this prominent model system.


Subject(s)
Gene Knockdown Techniques/methods , Morpholinos/pharmacology , Sea Urchins/genetics , Animals , Embryo, Nonmammalian/metabolism , Embryonic Development/genetics , Gene Expression/drug effects , Gene Expression/genetics , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Gene Silencing/physiology , Morpholinos/chemistry , Oligonucleotides, Antisense/genetics
13.
J Am Chem Soc ; 144(37): 16819-16826, 2022 09 21.
Article in English | MEDLINE | ID: mdl-36073798

ABSTRACT

MicroRNAs play crucial and dynamic roles in vertebrate development and diseases. Some, like miR-430, are highly expressed during early embryo development and regulate hundreds of transcripts, which can make it difficult to study their role in the timing and location of specific developmental processes using conventional morpholino oligonucleotide (MO) knockdown or genetic deletion approaches. We demonstrate that light-activated circular morpholino oligonucleotides (cMOs) can be applied to the conditional control of microRNA function. We targeted miR-430 in zebrafish embryos to study its role in the development of the embryo body and the heart. Using 405 nm irradiation, precise spatial and temporal control over miR-430 function was demonstrated, offering insight into the cell populations and developmental timepoints involved in each process.


Subject(s)
MicroRNAs , Zebrafish , Animals , Embryo, Nonmammalian , MicroRNAs/genetics , Morpholinos/pharmacology , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacology , Zebrafish Proteins/genetics
14.
Br J Cancer ; 127(1): 43-55, 2022 07.
Article in English | MEDLINE | ID: mdl-35277659

ABSTRACT

BACKGROUND: Osteosarcoma (OS) is the most common primary bone malignancy. Chemotherapy plays an essential role in OS treatment, potentially doubling 5-year event-free survival if tumour necrosis can be stimulated. The canonical Wnt inhibitor Dickkopf-1 (Dkk-1) enhances OS survival in part through upregulation of aldehyde-dehydrogenase-1A1 which neutralises reactive oxygen species originating from nutritional stress and chemotherapeutic challenge. METHODS: A vivo morpholino (DkkMo) was employed to block the expression of Dkk-1 in OS cells. Cell mitosis, gene expression and bone destruction were measured in vitro and in vivo in the presence and absence of doxorubicin (DRB). RESULTS: DkkMo reduced the expression of Dkk-1 and Aldh1a1, reduced expansion of OS tumours, preserved bone volume and architecture and stimulated tumour necrosis. This was observed in the presence or absence of DRB. CONCLUSION: These results indicate that administration of DkkMo with or without chemotherapeutics can substantially improve OS outcome with respect to tumour expansion and osteolytic corruption of bone in experimental OS model.


Subject(s)
Bone Neoplasms , Osteosarcoma , Bone Neoplasms/drug therapy , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Cell Line, Tumor , Humans , Intercellular Signaling Peptides and Proteins/genetics , Morpholinos/genetics , Morpholinos/pharmacology , Necrosis , Osteosarcoma/drug therapy , Osteosarcoma/genetics , Osteosarcoma/metabolism
15.
Muscle Nerve ; 66(3): 262-269, 2022 09.
Article in English | MEDLINE | ID: mdl-35715998

ABSTRACT

INTRODUCTION/AIMS: Pulmonary decline is a major issue in patients with Duchenne muscular dystrophy (DMD). Eteplirsen is a United States-approved treatment for patients with DMD and exon 51 skip-amenable mutations. Previous analyses have shown that eteplirsen is associated with a statistically significant attenuation of pulmonary decline. In this study we evaluate the effect of eteplirsen treatment from newly available data sources on pulmonary function over time in patients with DMD. METHODS: We used a post hoc pooled analysis to compare the percentage of predicted forced vital capacity (FVC%p) and projected time with pulmonary function milestones in patients with DMD and exon 51 skip-amenable mutations receiving eteplirsen (Studies 204 and 301) or standard of care (SoC; Cooperative International Neuromuscular Research Group Duchenne Natural History Study). A mixed model for repeated-measures framework was applied to evaluate the impact of eteplirsen. RESULTS: An average annual rate of FVC%p decline for eteplirsen-treated patients was estimated to be 3.47%, a statistically significant attenuation from the 5.95% rate of decline estimated in SoC patients (P = .0001). Using linear extrapolations of the model-estimated decline in FVC%p, the attenuation in FVC%p decline for eteplirsen-treated patients corresponded to a delay of 5.72 years in time to needing continuous ventilation, 3.31 years in time to needing nighttime ventilation, and 2.11 years in time to needing a cough assist device compared with SoC patients. DISCUSSION: The attenuation of FVC%p decline suggests that eteplirsen-treated patients had statistically significant and clinically meaningful attenuations in pulmonary decline compared with SoC patients.


Subject(s)
Muscular Dystrophy, Duchenne , Humans , Lung , Morpholinos/pharmacology , Vital Capacity
16.
Bioorg Med Chem Lett ; 59: 128570, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35063631

ABSTRACT

Several diaryl triazene derivatives were synthesized and tested for their ability to inhibit cytochrome P450 1A1 and 1B1 as a potential means to prevent and treat cancer. These compounds are more planar than their conformational flexible aryl morpholino triazene counterparts that were previously shown to inhibit the above enzymes. As a result, the diaryl triazenes are more likely to exhibit increased binding to the enzyme active sites and inhibit these enzymes more strongly than the aryl morpholino triazenes. The data indicates that the diaryl triazenes inhibit cytochrome P450 1A1 and 1B1 one to two orders of magnitude more strongly than the aryl morpholino triazenes. Furthermore, compounds 8-10 strongly inhibited cytochrome P450 1B1 with IC50 values of 51 nM, 740 nM, and 590 nM respectively. Thus, diaryl triazenes should be further investigated as a potential chemopreventive agent.


Subject(s)
Cytochrome P-450 CYP1A1/antagonists & inhibitors , Cytochrome P-450 CYP1B1/antagonists & inhibitors , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Morpholinos/pharmacology , Triazenes/pharmacology , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP1B1/metabolism , Cytochrome P-450 Enzyme Inhibitors/chemical synthesis , Cytochrome P-450 Enzyme Inhibitors/chemistry , Dose-Response Relationship, Drug , Humans , Molecular Structure , Morpholinos/chemical synthesis , Morpholinos/chemistry , Structure-Activity Relationship , Triazenes/chemical synthesis , Triazenes/chemistry
17.
Bioorg Med Chem ; 72: 116995, 2022 10 15.
Article in English | MEDLINE | ID: mdl-36095945

ABSTRACT

Aiming to develop novel tropomyosin receptor kinase A (TrkA) inhibitors, a scaffold hopping strategy was utilized by transforming the fused indazole of Entrectinib to phenyl triazole/thiazole skeleton to obtain compounds 7a-7 h and 13a-13 h. In the light of MTT assay, phenyl triazole derivatives 7a-7 h exhibited moderate anti-proliferative activities against KM-12 cells with the IC50 values of 1.78-17.51 µM, while phenyl thiazole derivatives 13a-13 h showed the weaker efficacy. Further structure-guided optimizations by combining the phenyl triazole skeleton with 3,5­difluorophenyl and 3-carbamoyl-4-piperazinylaniline moiety led to compounds 19a-19d and 20. Eventually, 19c bearing (2-(4-methylpiperazin-1-yl)phenyl)(morpholino)methanone moiety exhibited excellent anti-proliferative activity on TrkA-positive KM-12 cells with IC50 value of 0.17 µM. Meanwhile, compound 19c showed the inhibitory potency on TrkA with IC50 value of 1.6 nM, and displayed higher selectivity on TrkA over TrkB (IC50 = 12.3 nM) and TrkC (IC50 = 18.4 nM). The dedicated wound healing and colony formation assay indicated that the optimal compound 19c could suppress migration and significantly inhibit KM-12 cell colony formation in a dose-dependent manner. In addition, 19c could weakly induce apoptosis of KM-12 cell in immunofluorescent staining analysis. Taken together, the above results suggest 19c as a novel TrkA inhibitor worthy of further profiling.


Subject(s)
Antineoplastic Agents , Thiazoles , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Proliferation , Drug Design , Drug Screening Assays, Antitumor , Indazoles/pharmacology , Molecular Structure , Morpholinos/pharmacology , Protein Kinase Inhibitors/pharmacology , Structure-Activity Relationship , Thiazoles/pharmacology , Triazoles/pharmacology , Tropomyosin/pharmacology
18.
Nucleic Acids Res ; 48(6): 2853-2865, 2020 04 06.
Article in English | MEDLINE | ID: mdl-32103257

ABSTRACT

Spinal muscular atrophy (SMA) is a motor neuron disease. Nusinersen, a splice-switching antisense oligonucleotide (ASO), was the first approved drug to treat SMA. Based on prior preclinical studies, both 2'-O-methoxyethyl (MOE) with a phosphorothioate backbone and morpholino with a phosphorodiamidate backbone-with the same or extended target sequence as nusinersen-displayed efficient rescue of SMA mouse models. Here, we compared the therapeutic efficacy of these two modification chemistries in rescue of a severe mouse model using ASO10-29-a 2-nt longer version of nusinersen-via subcutaneous injection. Although both chemistries efficiently corrected SMN2 splicing in various tissues, restored motor function and improved the integrity of neuromuscular junctions, MOE-modified ASO10-29 (MOE10-29) was more efficacious than morpholino-modified ASO10-29 (PMO10-29) at the same molar dose, as seen by longer survival, greater body-weight gain and better preservation of motor neurons. Time-course analysis revealed that MOE10-29 had more persistent effects than PMO10-29. On the other hand, PMO10-29 appears to more readily cross an immature blood-brain barrier following systemic administration, showing more robust initial effects on SMN2 exon 7 inclusion, but less persistence in the central nervous system. We conclude that both modifications can be effective as splice-switching ASOs in the context of SMA and potentially other diseases, and discuss the advantages and disadvantages of each.


Subject(s)
Amides/chemistry , Morpholinos/therapeutic use , Muscular Atrophy, Spinal/drug therapy , Oligonucleotides, Antisense/therapeutic use , Phosphoric Acids/chemistry , Animals , Disease Models, Animal , Exons/genetics , Humans , Mice, Transgenic , Morpholinos/pharmacology , Motor Activity/drug effects , Motor Neurons/drug effects , Motor Neurons/pathology , Muscles/pathology , Muscular Atrophy, Spinal/pathology , Muscular Atrophy, Spinal/physiopathology , Neuromuscular Junction/pathology , Neuromuscular Junction/physiopathology , Oligonucleotides, Antisense/pharmacology , Phenotype , RNA Splicing/drug effects , RNA Splicing/genetics , Spinal Cord/pathology , Survival of Motor Neuron 2 Protein/genetics , Treatment Outcome
19.
Dev Biol ; 458(1): 98-105, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31682806

ABSTRACT

Attempts to constitutively knockout HTT in rodents resulted in embryonic lethality, curtailing efforts to study HTT function later in development. Here we show that HTT is dispensable for early zebrafish development, contrasting published zebrafish morpholino experiment results. Homozygous HTT knockouts were embryonically viable and appeared developmentally normal through juvenile stages. Comparison of adult fish revealed significant reduction in body size and fitness in knockouts compared to hemizygotes and wildtype fish, indicating an important role for wildtype HTT in postnatal development. Our zebrafish model provides an opportunity to understand the function of wildtype HTT later in development.


Subject(s)
Models, Animal , Nerve Tissue Proteins/physiology , Zebrafish Proteins/physiology , Zebrafish/genetics , Amino Acid Sequence , Animals , Body Size , CRISPR-Cas Systems , Conserved Sequence , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/embryology , Gene Editing , Gene Knockout Techniques , Genetic Association Studies , Genetic Fitness , Humans , Huntingtin Protein/chemistry , Morpholinos/pharmacology , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Neurulation/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Zebrafish/embryology , Zebrafish/growth & development , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics
20.
Neurobiol Dis ; 159: 105488, 2021 11.
Article in English | MEDLINE | ID: mdl-34425216

ABSTRACT

Spinal muscular atrophy (SMA) is an autosomal recessive disease characterized by survival motor neuron (SMN) protein deficiency which results in motor neuron loss and muscle atrophy. SMA is caused by a mutation or deletion of the survival motor neuron 1 (SMN1) gene and retention of the nearly identical SMN2 gene. SMN2 contains a C to T change in exon 7 that results in exon 7 exclusion from 90% of transcripts. SMN protein lacking exon 7 is unstable and rapidly degraded. The remaining full-length transcripts from SMN2 are insufficient for normal motor neuron function leading to the development of SMA. Three different therapeutic approaches that increase full-length SMN (FL-SMN) protein production are approved for treatment of SMA patients. Studies in both animal models and humans have demonstrated increasing SMN levels prior to onset of symptoms provides the greatest therapeutic benefit. Treatment of SMA, after some motor neuron loss has occurred, is also effective but to a lesser degree. The SMN∆7 mouse model is a well characterized model of severe or type 1 SMA, dying at 14 days of age. Here we treated three groups of ∆7SMA mice starting before, roughly during, and after symptom onset to determine if combining two mechanistically distinct SMN inducing therapies could improve the therapeutic outcome both before and after motor neuron loss. We found, compared with individual therapies, that morpholino antisense oligonucleotide (ASO) directed against ISS-N1 combined with the small molecule compound RG7800 significantly increased FL-SMN transcript and protein production resulting in improved survival and weight of ∆7SMA mice. Moreover, when give late symptomatically, motor unit function was completely rescued with no loss in function at 100 days of age in the dual treatment group. We have therefore shown that this dual therapeutic approach successfully increases SMN protein and rescues motor function in symptomatic ∆7SMA mice.


Subject(s)
Action Potentials/drug effects , Asymptomatic Diseases , Muscle, Skeletal/drug effects , Neuromuscular Junction/drug effects , Oligonucleotides, Antisense/pharmacology , Pyrazines/pharmacology , Pyrimidines/pharmacology , Spinal Muscular Atrophies of Childhood/physiopathology , Action Potentials/physiology , Animals , Disease Models, Animal , Mice , Mice, Knockout , Morpholinos/pharmacology , Muscle, Skeletal/innervation , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Neuromuscular Junction/pathology , Neuromuscular Junction/physiopathology , Spinal Muscular Atrophies of Childhood/genetics , Survival of Motor Neuron 1 Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL