Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 14.581
Filtrer
2.
Transl Neurodegener ; 13(1): 50, 2024 Oct 08.
Article de Anglais | MEDLINE | ID: mdl-39380076

RÉSUMÉ

BACKGROUND: Therapeutic approaches aimed at lowering toxic mutant huntingtin (mHTT) levels in the brain can reverse disease phenotypes in animal models of Huntington's disease (HD) and are currently being evaluated in clinical trials. Sensitive and dynamic response biomarkers are needed to assess the efficacy of such candidate therapies. Neurofilament light chain (NfL) is a biomarker of neurodegeneration that increases in cerebrospinal fluid (CSF) and blood with progression of HD. However, it remains unknown whether NfL in biofluids could serve as a response biomarker for assessing the efficacy of disease-modifying therapies for HD. METHODS: Longitudinal plasma and cross-sectional CSF samples were collected from the YAC128 transgenic mouse model of HD and wild-type (WT) littermate control mice throughout the natural history of disease. Additionally, biofluids were collected from YAC128 mice following intracerebroventricular administration of an antisense oligonucleotide (ASO) targeting the mutant HTT transgene (HTT ASO), at ages both before and after the onset of disease phenotypes. NfL concentrations in plasma and CSF were quantified using ultrasensitive single-molecule array technology. RESULTS: Plasma and CSF NfL concentrations were significantly elevated in YAC128 compared to WT littermate control mice from 9 months of age. Treatment of YAC128 mice with either 15 or 50 µg HTT ASO resulted in a dose-dependent, allele-selective reduction of mHTT throughout the brain at a 3-month interval, which was sustained with high-dose HTT ASO treatment for up to 6 months. Lowering of brain mHTT prior to the onset of regional brain atrophy and HD-like motor deficits in this model had minimal effect on plasma NfL at either dose, but led to a dose-dependent reduction of CSF NfL. In contrast, initiating mHTT lowering in the brain after the onset of neuropathological and behavioural phenotypes in YAC128 mice resulted in a dose-dependent stabilization of NfL increases in both plasma and CSF. CONCLUSIONS: Our data provide evidence that the response of NfL in biofluids is influenced by the magnitude of mHTT lowering in the brain and the timing of intervention, suggesting that NfL may serve as a promising exploratory response biomarker for HD.


Sujet(s)
Encéphale , Modèles animaux de maladie humaine , Protéine huntingtine , Maladie de Huntington , Souris transgéniques , Protéines neurofilamenteuses , Animaux , Maladie de Huntington/génétique , Maladie de Huntington/sang , Maladie de Huntington/liquide cérébrospinal , Protéine huntingtine/génétique , Protéines neurofilamenteuses/sang , Protéines neurofilamenteuses/liquide cérébrospinal , Souris , Encéphale/métabolisme , Encéphale/effets des médicaments et des substances chimiques , Encéphale/anatomopathologie , Marqueurs biologiques/sang , Marqueurs biologiques/liquide cérébrospinal , Oligonucléotides antisens/usage thérapeutique , Oligonucléotides antisens/pharmacologie , Mâle
3.
Acta Neurol Taiwan ; 33(3): 81-88, 2024 Sep 30.
Article de Anglais | MEDLINE | ID: mdl-39363429

RÉSUMÉ

Spinal muscular atrophy (SMA) is an autosomal recessive motor neuron disease characterized by progressive weakness and atrophy of skeletal muscles. With homozygous survival motor neuron 1 (SMN1) gene mutation, all SMA patients have at least one copy of the SMN2 gene, which provides an opportunity for drug targeting to enhance SMN expression. Current three disease modifying drugs, including nusinersen, onasemnogene abeparvovec, and risdiplam, have demonstrated impressive effectiveness in SMA treatment. Nusinersen is an antisense oligonucleotide targeting SMN2 pre-messenger RNA (mRNA) to modify alternative splicing and is effective in SMA children and adults, administrating via intermittent intrathecal injection. Onasemnogene abeparvovec is an adeno-associated viral vector carrying human SMN1 gene, featuring intravenous injection once in a lifetime for SMA patients less than 2 years of the age. Risdiplam is a small molecule also targeting SMN2 pre-mRNA and is effective in SMA children and adults with administration via oral intake once per day. Patients with SMA should receive these disease modifying therapies as soon as possible to not only stabilize disease progression, but potentially obtain neurological improvement. The development in these therapies has benefited patients with SMA and will potentially provide insight in future drug discovery for other neurodegenerative diseases. Keywords: Adeno-associated viral vector, antisense oligonucleotide, disease modifying therapy, gene therapy, motor neuron disease, spinal muscular atrophy.


Sujet(s)
Amyotrophie spinale , Oligonucléotides , Humains , Amyotrophie spinale/thérapie , Amyotrophie spinale/traitement médicamenteux , Amyotrophie spinale/génétique , Oligonucléotides/usage thérapeutique , Oligonucléotides/administration et posologie , Pyrimidines/usage thérapeutique , Sulfonamides/usage thérapeutique , Protéine-2 de survie du motoneurone/génétique , Thérapie génétique/méthodes , Protéine-1 de survie du motoneurone/génétique , Oligonucléotides antisens/usage thérapeutique , Produits biologiques/usage thérapeutique , Composés azoïques , Protéines de fusion recombinantes
4.
AAPS PharmSciTech ; 25(7): 239, 2024 Oct 10.
Article de Anglais | MEDLINE | ID: mdl-39390148

RÉSUMÉ

Liquid chromatography-mass spectrometry (LC-MS) is an effective tool for high-throughput quantification of oligonucleotides that is crucial for understanding their biological roles and developing diagnostic tests. This paper presents a high-throughput LC-MS/MS method that may be versatilely applied for a wide range of oligonucleotides, making it a valuable tool for rapid screening and discovery. The method is demonstrated using an in-house synthesized MALAT-1 Antisense oligonucleotide (ASO) as a test case. Biological samples were purified using a reversed liquid-liquid extraction process automated by a liquid handling workstation and analyzed with ion-pairing LC-MS/MS. The assay was evaluated for sensitivity (LLOQ = 2 nM), specificity, precision, accuracy, recovery, matrix effect, and stability in rat cerebrospinal fluid (CSF) and plasma. Besides some existing considerations such as column selection, ion-pairing reagent, and sample purification, our work focused on the following four subtopics: 1) selecting the appropriate Multiple Reaction Monitoring (MRM) transition to maximize sensitivity for trace-level ASO in biological samples; 2) utilizing a generic risk-free internal standard (tenofovir) to avoid crosstalk interference from the oligo internal standard commonly utilized in the LC-MS assay; 3) automating the sample preparation process to increase precision and throughput; and 4) comparing liquid-liquid extraction (LLE) and solid-phase extraction (SPE) as sample purification methods in oligo method development. The study quantified the concentration of MALAT-1 ASO in rat CSF and plasma after intrathecal injection and used the difference between the two matrices to evaluate the injection technique. The results provide a solid foundation for further internal oligonucleotide discovery and development.


Sujet(s)
Découverte de médicament , Spectrométrie de masse en tandem , Animaux , Rats , Chromatographie en phase liquide/méthodes , Spectrométrie de masse en tandem/méthodes , Découverte de médicament/méthodes , Oligonucléotides antisens/administration et posologie , Extraction liquide-liquide/méthodes , Oligonucléotides , Tests de criblage à haut débit/méthodes , ARN long non codant , Liquid Chromatography-Mass Spectrometry
5.
Lipids Health Dis ; 23(1): 329, 2024 Oct 03.
Article de Anglais | MEDLINE | ID: mdl-39363329

RÉSUMÉ

BACKGROUND: Olezarsen is a GalNAc3-conjugated, hepatic-targeted antisense oligonucleotide that lowers apolipoprotein C-III (apoC-III) and triglyceride levels. The efficacy and safety of olezarsen has not previously been studied in ethnically diverse American populations. The aim of this study is to assess the effect of olezarsen in healthy Japanese Americans. METHODS: A randomized, placebo-controlled, double-blind phase 1 study was performed in 28 healthy Japanese American participants treated with olezarsen in single-ascending doses (SAD; 30, 60, 90 mg) or multiple doses (MD; 60 mg every 4 weeks for 4 doses). The primary, secondary, and exploratory objectives were safety and tolerability, pharmacokinetics, and effects of olezarsen on fasting serum triglycerides and apoC-III, respectively. RESULTS: There were 20 participants (16 active:4 placebo) in the SAD part of the study, and 8 participants (6 active:2 placebo) in the MD part of the study. For the primary endpoint, no serious adverse events or clinically relevant laboratory abnormalities were reported. The majority of olezarsen plasma exposure occurred within 24 h post-dose. In the SAD cohorts at Day 15 the percentage reduction in apoC-III/TG was - 39.4%/ - 17.8%, - 60.8%/ - 52.7%, and - 68.1%/ - 39.2% in the 30, 60 and 90 mg doses, respectively, vs 2.3%/44.5% increases in placebo. In the MD cohort, at Day 92 the percentage reduction in apoC-III/TG was - 81.6/ - 73.8% vs - 17.2/ - 40.8% reduction in placebo. Favorable changes were also present in VLDL-C, apoB and HDL-C. CONCLUSIONS: Single- and multiple-dose administration of olezarsen was safe, was well tolerated, and significantly reduced apoC-III and triglyceride levels in healthy Japanese Americans.


Sujet(s)
Apolipoprotéine C-III , Triglycéride , Adulte , Femelle , Humains , Mâle , Adulte d'âge moyen , Jeune adulte , Apolipoprotéine C-III/sang , 23895 , Méthode en double aveugle , Oligonucléotides/administration et posologie , Oligonucléotides antisens/administration et posologie , Triglycéride/sang , Japon/ethnologie , États-Unis
6.
Medicina (B Aires) ; 84 Suppl 3: 15-20, 2024 Sep.
Article de Espagnol | MEDLINE | ID: mdl-39331770

RÉSUMÉ

Angelman syndrome is a severe neurodevelopmental disorder secondary to disruption of the UBE3A gene in the maternal allele of chromosome 15. Its manifestations are mainly neurological, but a multidisciplinary management is required for its treatment. There are consensus guidelines available for best clinical management. Current clinical trials with antisense oligonucleotides promise, for the first time, to treat the cause by activating the UBE3A gene in the paternal allele, showing encouraging preliminary clinical effects. Inoculation of UBE3A gene through a viral vector has been tested in animal models and is underway for future clinical trials.


El síndrome de Angelman es un grave desorden del neurodesarrollo secundario a disrupción del gen UBE3A en el alelo materno del cromosoma 15. Sus manifestaciones son principalmente neurológicas, pero se requiere de un manejo multidisciplinario para su tratamiento. Existen guías por consenso para el manejo clínico adecuado. Actuales ensayos clínicos con oligonucleótidos antisentido prometen, por primera vez, tratar la causa por medio de activación del gen UBE3A en el alelo paterno, demostrando efectos clínicos preliminares alentadores. La inoculación del gen UBE3A a través de un vector viral ha sido probada en modelos animales y está en vías para futuros ensayos clínicos.


Sujet(s)
Syndrome d'Angelman , Thérapie génétique , Humains , Syndrome d'Angelman/thérapie , Syndrome d'Angelman/génétique , Oligonucléotides antisens/usage thérapeutique , Ubiquitin-protein ligases/génétique , Animaux
7.
Sci Transl Med ; 16(765): eadk0845, 2024 09 18.
Article de Anglais | MEDLINE | ID: mdl-39292801

RÉSUMÉ

Autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED) is a life-threatening monogenic autoimmune disorder primarily caused by biallelic deleterious variants in the autoimmune regulator (AIRE) gene. We prospectively evaluated 104 patients with clinically diagnosed APECED syndrome and identified 17 patients (16%) from 14 kindreds lacking biallelic AIRE variants in exons or flanking intronic regions; 15 had Puerto Rican ancestry. Through whole-genome sequencing, we identified a deep intronic AIRE variant (c.1504-818 G>A) cosegregating with the disease in all 17 patients. We developed a culture system of AIRE-expressing primary patient monocyte-derived dendritic cells and demonstrated that c.1504-818 G>A creates a cryptic splice site and activates inclusion of a 109-base pair frame-shifting pseudoexon. We also found low-level AIRE expression in patient-derived lymphoblastoid cell lines (LCLs) and confirmed pseudoexon inclusion in independent extrathymic AIRE-expressing cell lines. Through protein modeling and transcriptomic analyses of AIRE-transfected human embryonic kidney 293 and thymic epithelial cell 4D6 cells, we showed that this variant alters the carboxyl terminus of the AIRE protein, abrogating its function. Last, we developed an antisense oligonucleotide (ASO) that reversed pseudoexon inclusion and restored the normal AIRE transcript sequence in LCLs. Thus, our findings revealed c.1504-818 G>A as a founder APECED-causing AIRE variant in the Puerto Rican population and uncovered pseudoexon inclusion as an ASO-reversible genetic mechanism underlying APECED.


Sujet(s)
AIRE Protein , Exons , Introns , Oligonucléotides antisens , Polyendocrinopathies auto-immunes , Facteurs de transcription , Adolescent , Adulte , Enfant , Femelle , Humains , Mâle , Séquence nucléotidique , Lignée cellulaire , Exons/génétique , Introns/génétique , Mutation/génétique , Pedigree , Polyendocrinopathies auto-immunes/génétique , Épissage des ARN/génétique , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme
8.
Genes (Basel) ; 15(9)2024 Sep 08.
Article de Anglais | MEDLINE | ID: mdl-39336772

RÉSUMÉ

RNA splicing is an essential post-transcriptional mechanism that facilitates the excision of introns and the connection of exons to produce mature mRNA, which is essential for gene expression and proteomic diversity. In the liver, precise splicing regulation is critical for maintaining metabolic balance, detoxification, and protein synthesis. This review explores the mechanisms of RNA splicing and the role of splicing factors, particularly in the context of Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD). This review also highlights how RNA splicing dysregulation can lead to aberrant splicing and impact the progression of liver diseases such as MASLD, with a particular focus on Metabolic Dysfunction-Associated Steatohepatitis (MASH), which represents the advanced stage of MASLD. Recent advances in the clinical application of antisense oligonucleotides (ASOs) to correct splicing errors offer promising therapeutic strategies for restoring normal liver function. Additionally, the dysregulation of splicing observed in liver diseases may serve as a potential diagnostic marker, offering new opportunities for early identification of individuals more susceptible to disease progression. This review provides insights into the molecular mechanisms that govern splicing regulation in the liver, with a particular emphasis on MASLD, and discusses potential therapeutic approaches targeting RNA splicing to treat MASLD and related metabolic disorders.


Sujet(s)
Foie , Épissage des ARN , Humains , Épissage des ARN/génétique , Foie/métabolisme , Foie/anatomopathologie , Stéatose hépatique/génétique , Stéatose hépatique/métabolisme , Stéatose hépatique/anatomopathologie , Animaux , Oligonucléotides antisens/génétique , Oligonucléotides antisens/usage thérapeutique
9.
Front Immunol ; 15: 1441733, 2024.
Article de Anglais | MEDLINE | ID: mdl-39267760

RÉSUMÉ

This review will briefly introduce microRNAs (miRNAs) and dissect their contribution to multiple sclerosis (MS) and its clinical outcomes. For this purpose, we provide a concise overview of the present knowledge of MS pathophysiology, biomarkers and treatment options, delving into the role of selectively expressed miRNAs in clinical forms of this disease, as measured in several biofluids such as serum, plasma or cerebrospinal fluid (CSF). Additionally, up-to-date information on current strategies applied to miRNA-based therapeutics will be provided, including miRNA restoration therapy (lentivirus expressing a specific type of miRNA and miRNA mimic) and miRNA inhibition therapy such as antisense oligonucleotides, small molecules inhibitors, locked nucleic acids (LNAs), anti-miRNAs, and antagomirs. Finally, it will highlight future directions and potential limitations associated with their application in MS therapy, emphasizing the need for improved delivery methods and validation of therapeutic efficacy.


Sujet(s)
microARN , Sclérose en plaques , Antagomirs/usage thérapeutique , Marqueurs biologiques/sang , Exosomes , Thérapie génétique , microARN/sang , microARN/liquide cérébrospinal , microARN/usage thérapeutique , Sclérose en plaques/génétique , Sclérose en plaques/physiopathologie , Sclérose en plaques/thérapie , Oligonucléotides antisens/usage thérapeutique , Humains , Animaux
10.
Nat Commun ; 15(1): 8009, 2024 Sep 13.
Article de Anglais | MEDLINE | ID: mdl-39271668

RÉSUMÉ

Chemical modification of DNA is a common strategy to improve the properties of oligonucleotides, particularly for therapeutics and nanotechnology. Existing synthetic methods essentially rely on phosphoramidite chemistry or the polymerization of nucleoside triphosphates but are limited in terms of size, scalability, and sustainability. Herein, we report a robust alternative method for the de novo synthesis of modified oligonucleotides using template-dependent DNA ligation of shortmer fragments. Our approach is based on the fast and scaled accessibility of chemically modified shortmer monophosphates as substrates for the T3 DNA ligase. This method has shown high tolerance to chemical modifications, flexibility, and overall efficiency, thereby granting access to a broad range of modified oligonucleotides of different lengths (20 → 120 nucleotides). We have applied this method to the synthesis of clinically relevant antisense drugs and ultramers containing diverse modifications. Furthermore, the designed chemoenzymatic approach has great potential for diverse applications in therapeutics and biotechnology.


Sujet(s)
DNA ligases , ADN , Oligonucléotides , Oligonucléotides/composition chimique , Oligonucléotides/synthèse chimique , DNA ligases/métabolisme , ADN/composition chimique , Matrices (génétique) , Oligonucléotides antisens/composition chimique , Oligonucléotides antisens/synthèse chimique
11.
Front Immunol ; 15: 1426657, 2024.
Article de Anglais | MEDLINE | ID: mdl-39234236

RÉSUMÉ

Our goal is to improve the outcomes of cancer immunotherapy by targeting FOXP3+ T-regulatory (Treg) cells with a next generation of antisense oligonucleotides (ASO), termed FOXP3 AUMsilence ASO. We performed in vitro experiments with human healthy donor PBMC and clinical samples from patients with lung cancer, mesothelioma and melanoma, and tested our approach in vivo using ASO FOXP3 in syngeneic murine cancer models and in humanized mice. ASO FOXP3 had no effects on cell viability or cell division, did not affect expression of other FOXP members, but decreased expression of FOXP3 mRNA in PBMC by 54.9% and in cancer samples by 64.7%, with corresponding 41.0% (PBMC) and 60.0% (cancer) decreases of Treg numbers (all p<0.0001). Hence, intratumoral Treg were more sensitive to the effects of ASO FOXP3 than peripheral blood Tregs. Isolated human Treg, incubated with ASO FOXP3 for 3.5 hours, had significantly impaired suppressive function (66.4%) versus Scramble control. In murine studies, we observed a significant inhibition of tumor growth, while 13.6% (MC38) to 22% (TC1) of tumors were completely resorbed, in conjunction with ~50% decrease of Foxp3 mRNA by qPCR and decreased numbers of intratumoral Tregs. In addition, there were no changes in FOXP3 mRNA expression or in the numbers of Tregs in draining lymph nodes and in spleens of tumor bearing mice, confirming that intratumoral Treg had enhanced sensitivity to ASO FOXP3 in vivo compared to other Treg populations. ASO FOXP3 Treg targeting in vivo and in vitro was accompanied by significant downregulation of multiple exhaustion markers, and by increased expression of perforin and granzyme-B by intratumoral T cells. To conclude, we report that targeting the key Treg transcription factor FOXP3, with ASO FOXP3, has a powerful anti-tumoral effect and enhances T cell response in vitro and in vivo.


Sujet(s)
Facteurs de transcription Forkhead , Oligonucléotides antisens , Lymphocytes T régulateurs , Animaux , Facteurs de transcription Forkhead/métabolisme , Facteurs de transcription Forkhead/génétique , Lymphocytes T régulateurs/immunologie , Lymphocytes T régulateurs/métabolisme , Humains , Souris , Femelle , Tumeurs/immunologie , Tumeurs/thérapie , Lignée cellulaire tumorale , Souris de lignée C57BL , Immunothérapie/méthodes
13.
Int Immunopharmacol ; 142(Pt B): 113157, 2024 Dec 05.
Article de Anglais | MEDLINE | ID: mdl-39288629

RÉSUMÉ

Nucleic acid is an essential biopolymer in all living cells, performing the functions of storing and transmitting genetic information and synthesizing protein. In recent decades, with the progress of science and biotechnology and the continuous exploration of the functions performed by nucleic acid, more and more studies have confirmed that nucleic acid therapy for living organisms has great medical therapeutic potential. Nucleic acid drugs began to become independent therapeutic agents. As a new therapeutic method, nucleic acid therapy plays an important role in the treatment of genetic diseases, viral infections and cancers. There are currently 19 nucleic acid drugs approved by the Food and Drug Administration (FDA). In the following review, we start from principles and advantages of nucleic acid therapy, and briefly describe development history of nucleic acid drugs. And then we give examples of various RNA therapeutic drugs, including antisense oligonucleotides (ASO), mRNA vaccines, small interfering RNA (siRNA) and microRNA (miRNA), aptamers, and small activating RNA (saRNA). In addition, we also focused on the current status of nucleic acid drugs used in cancer therapy and the breakthrough in recent years. Clinical trials of nucleic acid drugs for cancer treatment are under way, conventional radiotherapy and chemotherapy combined with the immunotherapies such as checkpoint inhibitors and nucleic acid drugs may be the main prospects for successful cancer treatment.


Sujet(s)
Tumeurs , Acides nucléiques , Humains , Tumeurs/thérapie , Tumeurs/traitement médicamenteux , Animaux , Acides nucléiques/usage thérapeutique , Thérapie génétique/méthodes , Oligonucléotides antisens/usage thérapeutique , Petit ARN interférent/usage thérapeutique , Immunothérapie/méthodes
14.
Acta Neuropathol ; 148(1): 45, 2024 Sep 21.
Article de Anglais | MEDLINE | ID: mdl-39305312

RÉSUMÉ

Amyotrophic lateral sclerosis (ALS) is an adult-onset motor neuron disease with a mean survival time of three years. The 97% of the cases have TDP-43 nuclear depletion and cytoplasmic aggregation in motor neurons. TDP-43 prevents non-conserved cryptic exon splicing in certain genes, maintaining transcript stability, including ATG4B, which is crucial for autophagosome maturation and Microtubule-associated proteins 1A/1B light chain 3B (LC3B) homeostasis. In ALS mice (G93A), Atg4b depletion worsens survival rates and autophagy function. For the first time, we observed an elevation of LC3ylation in the CNS of both ALS patients and atg4b-/- mouse spinal cords. Furthermore, LC3ylation modulates the distribution of ATG3 across membrane compartments. Antisense oligonucleotides (ASOs) targeting cryptic exon restore ATG4B mRNA in TARDBP knockdown cells. We further developed multi-target ASOs targeting TDP-43 binding sequences for a broader effect. Importantly, our ASO based in peptide-PMO conjugates show brain distribution post-IV administration, offering a non-invasive ASO-based treatment avenue for neurodegenerative diseases.


Sujet(s)
Sclérose latérale amyotrophique , Protéines associées à l'autophagie , Cysteine endopeptidases , Protéines de liaison à l'ADN , Protéines associées aux microtubules , Animaux , Protéines associées à l'autophagie/métabolisme , Protéines associées à l'autophagie/génétique , Humains , Protéines de liaison à l'ADN/métabolisme , Protéines de liaison à l'ADN/génétique , Souris , Protéines associées aux microtubules/métabolisme , Protéines associées aux microtubules/génétique , Sclérose latérale amyotrophique/métabolisme , Sclérose latérale amyotrophique/génétique , Sclérose latérale amyotrophique/anatomopathologie , Cysteine endopeptidases/métabolisme , Cysteine endopeptidases/génétique , Mâle , Moelle spinale/métabolisme , Moelle spinale/anatomopathologie , Autophagie/physiologie , Souris knockout , Épissage des ARN/génétique , Femelle , Souris transgéniques , Motoneurones/métabolisme , Motoneurones/anatomopathologie , Oligonucléotides antisens/pharmacologie
15.
J Vis Exp ; (210)2024 Aug 23.
Article de Anglais | MEDLINE | ID: mdl-39248512

RÉSUMÉ

Metastatic breast cancer is a devastating disease with very limited therapeutic options, calling for new therapeutic strategies. Oncogenic miRNAs have been shown to be associated with the metastatic potential of breast cancer and are implicated in tumor cell migration, invasion, and viability. However, it can be difficult to deliver an inhibitory RNA molecule to the tissue of interest. To overcome this challenge and deliver active antisense oligonucleotides to tumors, we utilized magnetic iron oxide nanoparticles as a delivery platform. These nanoparticles target tissues with increased vascular permeability, such as sites of inflammation or cancer. Delivery of these nanoparticles can be monitored in vivo by magnetic resonance imaging (MRI) due to their magnetic properties. Translation of this therapeutic approach into the clinic will be more accessible because of its compatibility with this relevant imaging modality. They can also be labeled with other imaging reporters such as a Cy5.5 near-infrared optical dye for correlative optical imaging and fluorescence microscopy. Here, we demonstrate that nanoparticles labeled with Cy5.5 and conjugated to therapeutic oligomers targeting oncogenic miRNA-10b (termed MN-anti-miR10b, or "nanodrug") administered intravenously accumulate in metastatic sites, opening a possibility for therapeutic intervention of metastatic breast cancer.


Sujet(s)
Carbocyanines , microARN , Animaux , Femelle , Souris , microARN/génétique , microARN/administration et posologie , Carbocyanines/composition chimique , Tumeurs expérimentales de la mamelle/anatomopathologie , Tumeurs expérimentales de la mamelle/métabolisme , Tumeurs expérimentales de la mamelle/imagerie diagnostique , Nanoparticules magnétiques d'oxyde de fer/composition chimique , Imagerie par résonance magnétique/méthodes , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Tumeurs du sein/imagerie diagnostique , Oligonucléotides antisens/administration et posologie , Oligonucléotides antisens/composition chimique
16.
Sci Transl Med ; 16(760): eadi2245, 2024 Aug 14.
Article de Anglais | MEDLINE | ID: mdl-39141703

RÉSUMÉ

Antisense oligonucleotides (ASOs) are promising therapeutics for treating various neurological disorders. However, ASOs are unable to readily cross the mammalian blood-brain barrier (BBB) and therefore need to be delivered intrathecally to the central nervous system (CNS). Here, we engineered a human transferrin receptor 1 (TfR1) binding molecule, the oligonucleotide transport vehicle (OTV), to transport a tool ASO across the BBB in human TfR knockin (TfRmu/hu KI) mice and nonhuman primates. Intravenous injection and systemic delivery of OTV to TfRmu/hu KI mice resulted in sustained knockdown of the ASO target RNA, Malat1, across multiple mouse CNS regions and cell types, including endothelial cells, neurons, astrocytes, microglia, and oligodendrocytes. In addition, systemic delivery of OTV enabled Malat1 RNA knockdown in mouse quadriceps and cardiac muscles, which are difficult to target with oligonucleotides alone. Systemically delivered OTV enabled a more uniform ASO biodistribution profile in the CNS of TfRmu/hu KI mice and greater knockdown of Malat1 RNA compared with a bivalent, high-affinity TfR antibody. In cynomolgus macaques, an OTV directed against MALAT1 displayed robust ASO delivery to the primate CNS and enabled more uniform biodistribution and RNA target knockdown compared with intrathecal dosing of the same unconjugated ASO. Our data support systemically delivered OTV as a potential platform for delivering therapeutic ASOs across the BBB.


Sujet(s)
Barrière hémato-encéphalique , Oligonucléotides antisens , ARN long non codant , Récepteurs à la transferrine , Animaux , Humains , Souris , Transport biologique , Barrière hémato-encéphalique/métabolisme , Techniques de knock-down de gènes , Macaca fascicularis , Oligonucléotides antisens/pharmacocinétique , Oligonucléotides antisens/administration et posologie , Récepteurs à la transferrine/métabolisme , ARN long non codant/métabolisme , ARN long non codant/génétique , Distribution tissulaire
17.
ACS Appl Mater Interfaces ; 16(35): 45871-45887, 2024 Sep 04.
Article de Anglais | MEDLINE | ID: mdl-39163516

RÉSUMÉ

The long noncoding RNAs (lncRNA) are primarily associated with several essential gene regulations but are also connected to cancer metabolism and progression. HOTAIR and MALAT1 are two such lncRNAs that are detected in malignancies of various origins and are responsible for the poor prognosis of cancer patients. Due to these factors, the lncRNAs have emerged as prime targets for the development of anticancer therapeutics. However, nonviral delivery of lncRNA-targeted antisense oligonucleotides (ASOs) still remains a critical challenge while maintaining their structural and functional integrity. Herein, we have designed and synthesized a new series of ionizable lipids with variations in their head groups to prepare lipid nanoparticle (LNP) formulation along with cholesterol-based twin cationic lipid and amphiphilic zwitterionic lipid. The context responsiveness of these formulations in delivering the ASOs has been thoroughly investigated by various bioanalytical techniques, and an optimum formulation has been identified. The LNPs are utilized to deliver the ASOs targeting HOTAIR lncRNA in human cancer cell lines and MALAT1 lncRNA in mouse models. This study thus standardizes an advanced nanomaterial system for nonviral gene delivery that has been validated by a considerable reduction in the target lncRNA level under in vitro and a significant reduction in tumor volume under in vivo settings.


Sujet(s)
Tumeurs du sein , Lipides , Nanoparticules , Oligonucléotides antisens , ARN long non codant , ARN long non codant/génétique , ARN long non codant/métabolisme , Humains , Nanoparticules/composition chimique , Oligonucléotides antisens/composition chimique , Oligonucléotides antisens/pharmacologie , Animaux , Souris , Femelle , Lipides/composition chimique , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/anatomopathologie , Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Lignée cellulaire tumorale , Souris nude
19.
Biomolecules ; 14(8)2024 Aug 06.
Article de Anglais | MEDLINE | ID: mdl-39199337

RÉSUMÉ

Alpha-synuclein (αSyn) aggregates are the primary component of Lewy bodies, which are pathological hallmarks of Parkinson's disease (PD). The toxicity of αSyn seems to increase with its elevated expression during injury, suggesting that therapeutic approaches focused on reducing αSyn burden in neurons could be beneficial. Additionally, studies have shown higher levels of SNCA mRNA in the midbrain tissues and substantia nigra dopaminergic neurons of sporadic PD post-mortem brains compared to controls. Therefore, the regulation of SNCA expression and inhibition of αSyn synthesis could play an important role in the pathogenesis of injury, resulting in an effective treatment approach for PD. In this context, we summarized the most recent and innovative strategies proposed that exploit the targeting of SNCA to regulate translation and efficiently knock down cytoplasmatic levels of αSyn. Significant progress has been made in developing antisense technologies for treating PD in recent years, with a focus on antisense oligonucleotides and short-interfering RNAs, which achieve high specificity towards the desired target. To provide a more exhaustive picture of this research field, we also reported less common but highly innovative strategies, including small molecules, designed to specifically bind 5'-untranslated regions and, targeting secondary nucleic acid structures present in the SNCA gene, whose formation can be modulated, acting as a transcription and translation control. To fully describe the efficiency of the reported strategies, the effect of αSyn reduction on cellular viability and dopamine homeostasis was also considered.


Sujet(s)
Maladie de Parkinson , alpha-Synucléine , Humains , alpha-Synucléine/génétique , alpha-Synucléine/métabolisme , Maladie de Parkinson/génétique , Maladie de Parkinson/métabolisme , Maladie de Parkinson/thérapie , Maladie de Parkinson/anatomopathologie , Animaux , Oligonucléotides antisens/pharmacologie , Oligonucléotides antisens/génétique , Oligonucléotides antisens/usage thérapeutique , Petit ARN interférent/génétique , Petit ARN interférent/métabolisme , Neurones dopaminergiques/métabolisme
20.
Int J Mol Sci ; 25(16)2024 Aug 13.
Article de Anglais | MEDLINE | ID: mdl-39201504

RÉSUMÉ

Vascular Ehlers-Danlos syndrome or Ehlers-Danlos syndrome type IV (vEDS) is a connective tissue disorder characterised by skin hyperextensibility, joint hypermobility and fatal vascular rupture caused by COL3A1 mutations that affect collagen III expression, homo-trimer assembly and secretion. Along with collagens I, II, V and XI, collagen III plays an important role in the extracellular matrix, particularly in the inner organs. To date, only symptomatic treatment for vEDS patients is available. Fibroblasts derived from vEDS patients carrying dominant negative and/or haploinsufficiency mutations in COL3A1 deposit reduced collagen III in the extracellular matrix. This study explored the potential of an antisense oligonucleotide (ASO)-mediated splice modulating strategy to bypass disease-causing COL3A1 mutations reported in the in-frame exons 10 and 15. Antisense oligonucleotides designed to redirect COL3A1 pre-mRNA processing and excise exons 10 or 15 were transfected into dermal fibroblasts derived from vEDS patients and a healthy control subject. Efficient exon 10 or 15 excision from the mature COL3A1 mRNA was achieved and intracellular collagen III expression was increased after treatment with ASOs; however, collagen III deposition into the extracellular matrix was reduced in patient cells. The region encoded by exon 10 includes a glycosylation site, and exon 15 encodes hydroxyproline and hydroxylysine-containing triplet repeats, predicted to be crucial for collagen III assembly. These results emphasize the importance of post-translational modification for collagen III homo-trimer assembly. In conclusion, while efficient skipping of target COL3A1 exons was achieved, the induced collagen III isoforms generated showed defects in extracellular matrix formation. While therapeutic ASO-mediated exon skipping is not indicated for the patients in this study, the observations are restricted to exons 10 and 15 and may not be applicable to other collagen III in-frame exons.


Sujet(s)
Collagène de type III , Syndrome d'Ehlers-Danlos , Exons , Fibroblastes , Mutation , Oligonucléotides antisens , Humains , Syndrome d'Ehlers-Danlos/génétique , Syndrome d'Ehlers-Danlos/thérapie , Collagène de type III/génétique , Collagène de type III/métabolisme , Exons/génétique , Fibroblastes/métabolisme , Oligonucléotides antisens/génétique , Oligonucléotides antisens/pharmacologie , Cellules cultivées , Épissage des ARN/génétique , Mâle , Matrice extracellulaire/métabolisme , Ehlers-Danlos Syndrome, Type IV
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE