Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 575(7781): 203-209, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31666698

RESUMEN

Accumulation of mutant proteins is a major cause of many diseases (collectively called proteopathies), and lowering the level of these proteins can be useful for treatment of these diseases. We hypothesized that compounds that interact with both the autophagosome protein microtubule-associated protein 1A/1B light chain 3 (LC3)1 and the disease-causing protein may target the latter for autophagic clearance. Mutant huntingtin protein (mHTT) contains an expanded polyglutamine (polyQ) tract and causes Huntington's disease, an incurable neurodegenerative disorder2. Here, using small-molecule-microarray-based screening, we identified four compounds that interact with both LC3 and mHTT, but not with the wild-type HTT protein. Some of these compounds targeted mHTT to autophagosomes, reduced mHTT levels in an allele-selective manner, and rescued disease-relevant phenotypes in cells and in vivo in fly and mouse models of Huntington's disease. We further show that these compounds interact with the expanded polyQ stretch and could lower the level of mutant ataxin-3 (ATXN3), another disease-causing protein with an expanded polyQ tract3. This study presents candidate compounds for lowering mHTT and potentially other disease-causing proteins with polyQ expansions, demonstrating the concept of lowering levels of disease-causing proteins using autophagosome-tethering compounds.


Asunto(s)
Alelos , Evaluación Preclínica de Medicamentos/métodos , Proteína Huntingtina/antagonistas & inhibidores , Proteína Huntingtina/genética , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/genética , Mutación/genética , Animales , Ataxina-3/genética , Autofagosomas/metabolismo , Autofagia , Modelos Animales de Enfermedad , Proteínas de Drosophila/antagonistas & inhibidores , Proteínas de Drosophila/química , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Femenino , Humanos , Proteína Huntingtina/química , Proteína Huntingtina/metabolismo , Masculino , Ratones , Proteínas Asociadas a Microtúbulos/genética , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutación/efectos de los fármacos , Neuronas/citología , Péptidos/genética , Fenotipo , Reproducibilidad de los Resultados
2.
Am J Hum Genet ; 105(6): 1112-1125, 2019 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-31708117

RESUMEN

Huntington disease (HD) is a fatal neurodegenerative disorder caused by a gain-of-function mutation in HTT. Suppression of mutant HTT has emerged as a leading therapeutic strategy for HD, with allele-selective approaches targeting HTT SNPs now in clinical trials. Haplotypes associated with the HD mutation (A1, A2, A3a) represent panels of allele-specific gene silencing targets for efficient treatment of individuals with HD of Northern European and indigenous South American ancestry. Here we extend comprehensive haplotype analysis of the HD mutation to key populations of Southern European, South Asian, Middle Eastern, and admixed African ancestry. In each of these populations, the HD mutation occurs predominantly on the A2 HTT haplotype. Analysis of HD haplotypes across all affected population groups enables rational selection of candidate target SNPs for development of allele-selective gene silencing therapeutics worldwide. Targeting SNPs on the A1 and A2 haplotypes in parallel is essential to achieve treatment of the most HD-affected subjects in populations where HD is most prevalent. Current allele-specific approaches will leave a majority of individuals with HD untreated in populations where the HD mutation occurs most frequently on the A2 haplotype. We further demonstrate preclinical development of potent and selective ASOs targeting SNPs on the A2 HTT haplotype, representing an allele-specific treatment strategy for these individuals. On the basis of comprehensive haplotype analysis, we show the maximum proportion of HD-affected subjects that may be treated with three or four allele targets in different populations worldwide, informing current allele-specific HTT silencing strategies.


Asunto(s)
Etnicidad/genética , Silenciador del Gen , Haplotipos , Proteína Huntingtina/antagonistas & inhibidores , Enfermedad de Huntington/terapia , Mutación , Oligonucleótidos Antisentido/uso terapéutico , Alelos , Humanos , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Polimorfismo de Nucleótido Simple , Pronóstico , Expansión de Repetición de Trinucleótido
3.
N Engl J Med ; 380(24): 2307-2316, 2019 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-31059641

RESUMEN

BACKGROUND: Huntington's disease is an autosomal-dominant neurodegenerative disease caused by CAG trinucleotide repeat expansion in HTT, resulting in a mutant huntingtin protein. IONIS-HTTRx (hereafter, HTTRx) is an antisense oligonucleotide designed to inhibit HTT messenger RNA and thereby reduce concentrations of mutant huntingtin. METHODS: We conducted a randomized, double-blind, multiple-ascending-dose, phase 1-2a trial involving adults with early Huntington's disease. Patients were randomly assigned in a 3:1 ratio to receive HTTRx or placebo as a bolus intrathecal administration every 4 weeks for four doses. Dose selection was guided by a preclinical model in mice and nonhuman primates that related dose level to reduction in the concentration of huntingtin. The primary end point was safety. The secondary end point was HTTRx pharmacokinetics in cerebrospinal fluid (CSF). Prespecified exploratory end points included the concentration of mutant huntingtin in CSF. RESULTS: Of the 46 patients who were enrolled in the trial, 34 were randomly assigned to receive HTTRx (at ascending dose levels of 10 to 120 mg) and 12 were randomly assigned to receive placebo. Each patient received all four doses and completed the trial. Adverse events, all of grade 1 or 2, were reported in 98% of the patients. No serious adverse events were seen in HTTRx-treated patients. There were no clinically relevant adverse changes in laboratory variables. Predose (trough) concentrations of HTTRx in CSF showed dose dependence up to doses of 60 mg. HTTRx treatment resulted in a dose-dependent reduction in the concentration of mutant huntingtin in CSF (mean percentage change from baseline, 10% in the placebo group and -20%, -25%, -28%, -42%, and -38% in the HTTRx 10-mg, 30-mg, 60-mg, 90-mg, and 120-mg dose groups, respectively). CONCLUSIONS: Intrathecal administration of HTTRx to patients with early Huntington's disease was not accompanied by serious adverse events. We observed dose-dependent reductions in concentrations of mutant huntingtin. (Funded by Ionis Pharmaceuticals and F. Hoffmann-La Roche; ClinicalTrials.gov number, NCT02519036.).


Asunto(s)
Proteína Huntingtina/antagonistas & inhibidores , Enfermedad de Huntington/tratamiento farmacológico , Nucleótidos/farmacología , Oligonucleótidos/uso terapéutico , Adulto , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Proteína Huntingtina/líquido cefalorraquídeo , Proteína Huntingtina/genética , Inyecciones Espinales , Masculino , Persona de Mediana Edad , Mutación , Nucleótidos/síntesis química , Oligonucleótidos/líquido cefalorraquídeo
4.
Nucleic Acids Res ; 48(1): 36-54, 2020 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-31745548

RESUMEN

Huntington disease (HD) is a fatal neurodegenerative disease caused by a pathogenic expansion of a CAG repeat in the huntingtin (HTT) gene. There are no disease-modifying therapies for HD. Artificial microRNAs targeting HTT transcripts for degradation have shown preclinical promise and will soon enter human clinical trials. Here, we examine the tolerability and efficacy of non-selective HTT lowering with an AAV5 encoded miRNA targeting human HTT (AAV5-miHTT) in the humanized Hu128/21 mouse model of HD. We show that intrastriatal administration of AAV5-miHTT results in potent and sustained HTT suppression for at least 7 months post-injection. Importantly, non-selective suppression of huntingtin was generally tolerated, however high dose AAV5-miHTT did induce astrogliosis. We observed an improvement of select behavioural and modest neuropathological HD-like phenotypes in Hu128/21 mice, suggesting a potential therapeutic benefit of miRNA-mediated non-selective HTT lowering. Finally, we also observed that potent reduction of wild type HTT (wtHTT) in Hu21 control mice was tolerated up to 7 months post-injection but may induce impairment of motor coordination and striatal atrophy. Taken together, our data suggests that in the context of HD, the therapeutic benefits of mHTT reduction may outweigh the potentially detrimental effects of wtHTT loss following non-selective HTT lowering.


Asunto(s)
Proteína Huntingtina/genética , Enfermedad de Huntington/terapia , MicroARNs/genética , Terapia Molecular Dirigida/métodos , Parvovirinae/genética , ARN Mensajero/genética , Animales , Animales Modificados Genéticamente , Astrocitos/metabolismo , Astrocitos/patología , Secuencia de Bases , Cuerpo Estriado/metabolismo , Cuerpo Estriado/patología , Dependovirus , Modelos Animales de Enfermedad , Dosificación de Gen , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Humanos , Proteína Huntingtina/antagonistas & inhibidores , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/patología , Ratones , MicroARNs/administración & dosificación , MicroARNs/metabolismo , Neuroglía/metabolismo , Neuroglía/patología , Neuronas/metabolismo , Neuronas/patología , Parvovirinae/metabolismo , Desempeño Psicomotor , Estabilidad del ARN , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/metabolismo , Repeticiones de Trinucleótidos
5.
Arch Biochem Biophys ; 712: 109033, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34534539

RESUMEN

The N-terminal 17-residue stretch of huntingtin (httN17) folds into an amphipathic α-helix. The httN17-harboring polyQ peptides form oligomers that are mediated via the assembly of the httN17 α-helices. The oligomerization results in higher local concentration of the polyglutamine (polyQ) region, thereby facilitating amyloid formation. The httN17 co-assembles with the httN17-harbouring polyQ peptides, thereby reducing the local polyQ concentration, and consequently inhibiting aggregation. This study presents the aggregation inhibition of the exon I region of pathogenic huntingtin by httN17 and its analogs. The C-terminal amidation of httN17 is found to be essential for activity. The httN17 peptides with free amino terminus and the acetylated amino terminus possess comparable activity. The httN17 analog, wherein the Leu7 and Ala10 are substituted with 2-aminoisobutyric acid residues, exhibits significantly higher activity than the native httN17.


Asunto(s)
Proteína Huntingtina/antagonistas & inhibidores , Proteína Huntingtina/química , Fragmentos de Péptidos/química , Multimerización de Proteína/efectos de los fármacos , Secuencia de Aminoácidos , Humanos , Fragmentos de Péptidos/síntesis química , Estructura Secundaria de Proteína
6.
EMBO Rep ; 19(3)2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29440125

RESUMEN

Trinucleotide repeat (TNR) expansions in the genome cause a number of degenerative diseases. A prominent TNR expansion involves the triplet CAG in the huntingtin (HTT) gene responsible for Huntington's disease (HD). Pathology is caused by protein and RNA generated from the TNR regions including small siRNA-sized repeat fragments. An inverse correlation between the length of the repeats in HTT and cancer incidence has been reported for HD patients. We now show that siRNAs based on the CAG TNR are toxic to cancer cells by targeting genes that contain long reverse complementary TNRs in their open reading frames. Of the 60 siRNAs based on the different TNRs, the six members in the CAG/CUG family of related TNRs are the most toxic to both human and mouse cancer cells. siCAG/CUG TNR-based siRNAs induce cell death in vitro in all tested cancer cell lines and slow down tumor growth in a preclinical mouse model of ovarian cancer with no signs of toxicity to the mice. We propose to explore TNR-based siRNAs as a novel form of anticancer reagents.


Asunto(s)
Proteína Huntingtina/genética , Neoplasias/genética , ARN Interferente Pequeño/farmacología , Repeticiones de Trinucleótidos/genética , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Modelos Animales de Enfermedad , Humanos , Proteína Huntingtina/antagonistas & inhibidores , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , Ratones , Neoplasias/patología , Neoplasias/terapia , Sistemas de Lectura Abierta , ARN Interferente Pequeño/genética , Expansión de Repetición de Trinucleótido/genética , Repeticiones de Trinucleótidos/efectos de los fármacos
7.
Neurobiol Dis ; 132: 104569, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31398458

RESUMEN

The types of treatments and interventions being developed for chronic neurodegenerative disorders have expanded considerably in recent years. In addition to the variety of targets being pursued, strategies have moved from symptom management to more directed disease-modifying approaches. Among them are antibody-based therapies, which are not only being evaluated for a range of tauopathies and synucleinopathies, but are also emerging as a potential application for monogenic disorders of the central nervous system (CNS), including Huntington's disease (HD). Despite the excitement around the early trial data of anti-sense oligonucleotides (ASO) treatment for such disorders, antibody therapies may hold the key to tackling another aspect of the disease that could be critical to its pathogenesis. While gene-based methodologies are designed to lower, predominantly within cellular elements, mutant huntingtin protein (mHtt) - the genetic product of HD - the pathological protein is abundant in free forms and in several compartments including the cerebrospinal fluid, the plasma and the extracellular matrix. With accumulating evidence for the spreading and seeding capacities of mHtt, it may indeed be essential to target the protein both intracellularly and extracellularly. Therefore, free forms of mHtt not only represents an ideal target for antibodies, but one that needs to be addressed if meaningful and maximal clinical benefits are to be expected. This review explores the potential use of antibody-based therapies to treat HD, including the rationale for this approach as well as the pre-clinical data supporting it. The potential challenges that will need to be considered if such route is to be pursued clinically are also discussed.


Asunto(s)
Anticuerpos/uso terapéutico , Enfermedad de Huntington/terapia , Inmunización/métodos , Inmunización/tendencias , Animales , Humanos , Proteína Huntingtina/antagonistas & inhibidores
8.
Neurobiol Dis ; 129: 29-37, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31042572

RESUMEN

Huntington's disease (HD) is a fatal autosomal dominant neurodegenerative disease caused by a CAG expansion, which translates into an elongated polyglutamine (polyQ) repeat near the amino-terminus of the huntingtin protein (HTT). This results in production of a toxic mutant huntingtin protein (mHTT) that leads to neuronal dysfunction and death. Currently, no disease-modifying treatments are available; however, numerous therapeutic strategies aimed at lowering HTT levels in the brain are under development. To date, studies have not closely examined the contribution of mHTT in neurons vs astrocytes to disease pathophysiology. To better understand the role of astrocytes in HD pathophysiology and the need for cell type specific targeting of HTT lowering therapeutic strategies, AAV capsids were employed that selectively transduce neurons, or both neurons and astrocytes. These vectors carrying miRNA sequences directed against HTT were injected into the YAC128 mouse model of HD to selectively lower HTT expression in neurons alone versus neurons and astrocytes. The results suggested that HTT lowering in neurons alone was not sufficient to rescue the motor phenotype in YAC128 mice. Furthermore, HTT lowering in both cell types was required to achieve maximal functional benefit. The study suggested that astrocyte dysfunction may play a critical role in HD pathogenesis, and thus astrocytes represent an important therapeutic target.


Asunto(s)
Astrocitos/metabolismo , Proteína Huntingtina/antagonistas & inhibidores , Enfermedad de Huntington/metabolismo , Animales , Astrocitos/patología , Encéfalo/metabolismo , Encéfalo/patología , Dependovirus , Modelos Animales de Enfermedad , Vectores Genéticos , Proteína Huntingtina/genética , Enfermedad de Huntington/patología , Ratones , Ratones Transgénicos , MicroARNs , Neuronas/metabolismo , Neuronas/patología , Fenotipo , Transducción Genética
9.
Chembiochem ; 20(16): 2133-2140, 2019 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-31166067

RESUMEN

Huntington's disease (HD) is classified as a protein-misfolding disease correlated with the mutant Huntingtin (mHtt) protein with abnormally expanded polyglutamine (polyQ) domains. Because no effective drugs have yet been reported, attempts to develop better therapy to delay the age of onset are in urgent demand. In this study, an amphiphilic peptide consisting of negatively charged hexaglutamic acid and a stretch of decaglutamine (E6 Q10 ) was chemically synthesized as an inhibitor against polyQ and mHtt toxicity. It is found that E6 Q10 selfassembles into spherical vesicles, as shown by means of TEM, cryoelectron microscopy, and dynamic light scattering. Assembled E6 Q10 prevented the polyQ-rich peptide (KKWQ20 AKK) from forming amyloid fibrils. To enable the cell-penetration ability of E6 Q10 , the E6 Q10 ⋅chitosan complex was generated. It is demonstrated that the complex penetrates cells, interferes with the mHtt oligomerization and aggregation process, and prevents mHtt cytotoxicity. By combining positively charged chitosan and amphiphilic peptides with a negatively charge moiety, a new strategy is provided to develop biocompatible and biodegradable inhibitors against mHtt toxicity.


Asunto(s)
Materiales Biocompatibles/farmacología , Quitosano/farmacología , Proteína Huntingtina/antagonistas & inhibidores , Enfermedad de Huntington/tratamiento farmacológico , Péptidos/farmacología , Tensoactivos/farmacología , Materiales Biocompatibles/síntesis química , Materiales Biocompatibles/química , Quitosano/química , Humanos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Estructura Molecular , Mutación , Tamaño de la Partícula , Péptidos/síntesis química , Péptidos/química , Agregado de Proteínas/efectos de los fármacos , Propiedades de Superficie , Tensoactivos/síntesis química , Tensoactivos/química
10.
Mol Ther ; 26(11): 2580-2591, 2018 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-30143435

RESUMEN

Effective transvascular delivery of therapeutic oligonucleotides to the brain presents a major hurdle to the development of gene silencing technologies for treatment of genetically defined neurological disorders. Distribution to the brain after systemic administrations is hampered by the low permeability of the blood-brain barrier (BBB) and the rapid clearance kinetics of these drugs from the blood. Here we show that transient osmotic disruption of the BBB enables transvascular delivery of hydrophobically modified small interfering RNA (hsiRNA) to the rat brain. Intracarotid administration of 25% mannitol and hsiRNA conjugated to phosphocholine-docosahexanoic acid (PC-DHA) resulted in broad ipsilateral distribution of PC-DHA-hsiRNAs in the brain. PC-DHA conjugation enables hsiRNA retention in the parenchyma proximal to the brain vasculature and enabled active internalization by neurons and astrocytes. Moreover, transvascular delivery of PC-DHA-hsiRNAs effected Htt mRNA silencing in the striatum (55%), hippocampus (51%), somatosensory cortex (52%), motor cortex (37%), and thalamus (33%) 1 week after administration. Aside from mild gliosis induced by osmotic disruption of the BBB, transvascular delivery of PC-DHA-hsiRNAs was not associated with neurotoxicity. Together, these findings provide proof-of-concept that temporary disruption of the BBB is an effective strategy for the delivery of therapeutic oligonucleotides to the brain.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Proteína Huntingtina/genética , Neuronas/efectos de los fármacos , ARN Interferente Pequeño/administración & dosificación , Animales , Astrocitos/efectos de los fármacos , Astrocitos/patología , Barrera Hematoencefálica/fisiopatología , Encéfalo/efectos de los fármacos , Encéfalo/fisiopatología , Arterias Carótidas/fisiología , Ácidos Docosahexaenoicos/administración & dosificación , Ácidos Docosahexaenoicos/química , Silenciador del Gen , Terapia Genética/métodos , Humanos , Proteína Huntingtina/antagonistas & inhibidores , Interacciones Hidrofóbicas e Hidrofílicas , Manitol/administración & dosificación , Neuronas/patología , Fosforilcolina/administración & dosificación , Fosforilcolina/química , ARN Interferente Pequeño/química , Ratas
11.
J Am Chem Soc ; 139(16): 5640-5643, 2017 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-28406616

RESUMEN

Huntington's disease is a neurodegenerative disorder associated with the expansion of the polyglutamine tract in the exon-1 domain of the huntingtin protein (htte1). Above a threshold of 37 glutamine residues, htte1 starts to aggregate in a nucleation-dependent manner. A 17-residue N-terminal fragment of htte1 (N17) has been suggested to play a crucial role in modulating the aggregation propensity and toxicity of htte1. Here we identify N17 as a potential target for novel therapeutic intervention using the molecular tweezer CLR01. A combination of biochemical experiments and computer simulations shows that binding of CLR01 induces structural rearrangements within the htte1 monomer and inhibits htte1 aggregation, underpinning the key role of N17 in modulating htte1 toxicity.


Asunto(s)
Hidrocarburos Aromáticos con Puentes/farmacología , Proteína Huntingtina/antagonistas & inhibidores , Organofosfatos/farmacología , Hidrocarburos Aromáticos con Puentes/química , Exones , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Simulación de Dinámica Molecular , Estructura Molecular , Organofosfatos/química , Agregado de Proteínas/efectos de los fármacos
12.
Mol Ther ; 24(4): 726-35, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26708003

RESUMEN

Effective gene delivery to the central nervous system (CNS) is vital for development of novel gene therapies for neurological diseases. Adeno-associated virus (AAV) vectors have emerged as an effective platform for in vivo gene transfer, but overall neuronal transduction efficiency of vectors derived from naturally occurring AAV capsids after systemic administration is relatively low. Here, we investigated the possibility of improving CNS transduction of existing AAV capsids by genetically fusing peptides to the N-terminus of VP2 capsid protein. A novel vector AAV-AS, generated by the insertion of a poly-alanine peptide, is capable of extensive gene transfer throughout the CNS after systemic administration in adult mice. AAV-AS is 6- and 15-fold more efficient than AAV9 in spinal cord and cerebrum, respectively. The neuronal transduction profile varies across brain regions but is particularly high in the striatum where AAV-AS transduces 36% of striatal neurons. Widespread neuronal gene transfer was also documented in cat brain and spinal cord. A single intravenous injection of an AAV-AS vector encoding an artificial microRNA targeting huntingtin (Htt) resulted in 33-50% knockdown of Htt across multiple CNS structures in adult mice. This novel AAV-AS vector is a promising platform to develop new gene therapies for neurodegenerative disorders.


Asunto(s)
Proteínas de la Cápside/metabolismo , Sistema Nervioso Central/metabolismo , Péptidos/genética , Transducción Genética , Animales , Células CHO , Proteínas de la Cápside/genética , Gatos , Línea Celular , Cricetulus , Dependovirus/genética , Técnicas de Transferencia de Gen , Terapia Genética , Vectores Genéticos/administración & dosificación , Proteína Huntingtina/antagonistas & inhibidores , Proteína Huntingtina/genética , Ratones , Péptidos/metabolismo , Proteínas Recombinantes de Fusión/metabolismo
13.
Angew Chem Int Ed Engl ; 56(38): 11530-11533, 2017 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-28703441

RESUMEN

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by the aggregation of mutant huntingtin (mHtt), and removal of toxic mHtt is expected to be an effective therapeutic approach. We designed two small hybrid molecules (1 and 2) by linking a ligand for ubiquitin ligase (cellular inhibitor of apoptosis protein 1; cIAP1) with probes for mHtt aggregates, anticipating that these compounds would recruit cIAP1 to mHtt and induce selective degradation by the ubiquitin-proteasome system. The synthesized compounds reduced mHtt levels in HD patient fibroblasts and appear to be promising candidates for the development of a treatment for HD.


Asunto(s)
Descubrimiento de Drogas , Proteína Huntingtina/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Relación Dosis-Respuesta a Droga , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Células HeLa , Humanos , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/metabolismo , Ligandos , Estructura Molecular , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad
14.
Arch Pharm Res ; 47(6): 571-595, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38764004

RESUMEN

Huntington's disease (HD) is a paradigm of a genetic neurodegenerative disorder characterized by the expansion of CAG repeats in the HTT gene. This extensive review investigates the molecular complexities of HD by highlighting the pathogenic mechanisms initiated by the mutant huntingtin protein. Adverse outcomes of HD include mitochondrial dysfunction, compromised protein clearance, and disruption of intracellular signaling, consequently contributing to the gradual deterioration of neurons. Numerous therapeutic strategies, particularly precision medicine, are currently used for HD management. Antisense oligonucleotides, such as Tominersen, play a leading role in targeting and modulating the expression of mutant huntingtin. Despite the promise of these therapies, challenges persist, particularly in improving delivery systems and the necessity for long-term safety assessments. Considering the future landscape, the review delineates promising directions for HD research and treatment. Innovations such as Clustered regularly interspaced short palindromic repeats associated system therapies (CRISPR)-based genome editing and emerging neuroprotective approaches present unprecedented opportunities for intervention. Collaborative interdisciplinary endeavors and a more insightful understanding of HD pathogenesis are on the verge of reshaping the therapeutic landscape. As we navigate the intricate landscape of HD, this review serves as a guide for unraveling the intricacies of this disease and progressing toward transformative treatments.


Asunto(s)
Proteína Huntingtina , Enfermedad de Huntington , Enfermedad de Huntington/terapia , Enfermedad de Huntington/genética , Humanos , Animales , Proteína Huntingtina/genética , Proteína Huntingtina/antagonistas & inhibidores , Proteína Huntingtina/metabolismo , Oligonucleótidos Antisentido/uso terapéutico , Terapia Genética/métodos , Edición Génica/métodos , Fármacos Neuroprotectores/uso terapéutico
15.
J Med Chem ; 64(2): 938-940, 2021 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-33443413

RESUMEN

GPR52 is an orphan G protein-coupled receptor (GPCR) highly expressed in the brain, especially in the striatum, and represents an emerging therapeutic target for Huntington's disease (HD), an incurable monogenic neurodegenerative disorder caused by the mutation of the huntingtin (mHTT) gene. This Viewpoint discusses the discovery, published in this journal, that a highly potent and specific GPR52 antagonist was identified through high-throughput screening and structure-activity relationship study, which diminishes not only mHTT protein levels, but also ameliorates HD-like phenotypes in the animal disease models. This strategy offers intriguing promise as a surprising approach for HD therapy, where nucleic acid medicine approaches such as small interference RNAs have been the main focus and encounter obstacles such as delivery efficiency.


Asunto(s)
Enfermedad de Huntington/tratamiento farmacológico , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Animales , Descubrimiento de Drogas , Ensayos Analíticos de Alto Rendimiento , Humanos , Proteína Huntingtina/antagonistas & inhibidores , Ratones , Fenotipo , ARN Interferente Pequeño/uso terapéutico , Relación Estructura-Actividad
16.
ACS Appl Mater Interfaces ; 13(51): 60894-60906, 2021 Dec 29.
Artículo en Inglés | MEDLINE | ID: mdl-34914364

RESUMEN

Huntington's disease (HD) belongs to protein misfolding disorders associated with polyglutamine (polyQ)-rich mutant huntingtin (mHtt) protein inclusions. Currently, it is indicated that the aggregation of polyQ-rich mHtt participates in neuronal toxicity and dysfunction. Here, we designed and synthesized a polyglutamine-specific gold nanoparticle (AuNP) complex, which specifically targeted mHtt and alleviated its toxicity. The polyglutamine-specific AuNPs were prepared by decorating the surface of AuNPs with an amphiphilic peptide (JLD1) consisting of both polyglutamine-binding sequences and negatively charged sequences. By applying the polyQ aggregation model system, we demonstrated that AuNPs-JLD1 dissociated the fibrillary aggregates from the polyQ peptide and reduced its ß-sheet content in a concentration-dependent manner. By further integrating polyethyleneimine (PEI) onto AuNPs-JLD1, we generated a complex (AuNPs-JLD1-PEI). We showed that this complex could penetrate cells, bind to cytosolic mHtt proteins, dissociate mHtt inclusions, reduce mHtt oligomers, and ameliorate mHtt-induced toxicity. AuNPs-JLD1-PEI was also able to be transported to the brain and improved the functional deterioration in the HD Drosophila larva model. Our results revealed the feasibility of combining AuNPs, JLD1s, and cell-penetrating polymers against mHtt protein aggregation and oligomerization, which hinted on the early therapeutic strategies against HD.


Asunto(s)
Materiales Biocompatibles/farmacología , Proteínas de Drosophila/antagonistas & inhibidores , Oro/farmacología , Proteína Huntingtina/antagonistas & inhibidores , Enfermedad de Huntington/tratamiento farmacológico , Nanopartículas del Metal/química , Compuestos Organometálicos/farmacología , Péptidos/farmacología , Animales , Materiales Biocompatibles/química , Drosophila , Proteínas de Drosophila/metabolismo , Oro/química , Proteína Huntingtina/metabolismo , Enfermedad de Huntington/metabolismo , Ensayo de Materiales , Compuestos Organometálicos/química , Péptidos/química , Agregado de Proteínas/efectos de los fármacos
17.
Eur J Med Chem ; 216: 113247, 2021 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-33652355

RESUMEN

As a newly emerged technology, PROTAC (proteolysis targeting chimera) is a promising therapeutic strategy for varieties of diseases. Unlike small molecule inhibitors, PROTACs catalytically induce target proteins degradation, including currently "undruggable" target proteins. In addition, PROTACs can be a potentially successful strategy to overcome drug resistance. IAPs can inhibit apoptosis by inhibiting caspase, and also exhibits the activity of E3 ubiquitin ligase. Specific and nongenetic IAP-based protein erasers (SNIPERs) are hybrid molecules that designed based on IAPs, and used to degrade the target proteins closely associated with diseases. Their structures consist of three parts, including target protein ligand, E3 ligase ligand and the linker between them. SNIPERs (PROTACs) degrade diseases-associated proteins through human inherent ubiquitin-proteasome system. So far, many SNIPERs have been developed to treat diseases that difficult to handle by traditional methods, such as radiotherapy, chemotherapy and small molecule inhibitors, and showed promising prospects in application. In this paper, the recent advances of SNIPERs were summarized, and the chances and challenges associated with this area were also highlighted.


Asunto(s)
Proteínas Inhibidoras de la Apoptosis/metabolismo , Ligandos , Antineoplásicos/química , Antineoplásicos/metabolismo , Proteínas Fetales/antagonistas & inhibidores , Proteínas Fetales/metabolismo , Proteínas de Fusión bcr-abl/antagonistas & inhibidores , Proteínas de Fusión bcr-abl/metabolismo , Humanos , Proteína Huntingtina/antagonistas & inhibidores , Proteína Huntingtina/metabolismo , Proteínas Inhibidoras de la Apoptosis/antagonistas & inhibidores , Proteínas Asociadas a Microtúbulos/antagonistas & inhibidores , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/metabolismo , Receptor Notch1/antagonistas & inhibidores , Receptor Notch1/metabolismo , Receptores Androgénicos/química , Receptores Androgénicos/metabolismo , Receptores de Estrógenos/metabolismo
18.
JAMA Neurol ; 77(6): 764-772, 2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32202594

RESUMEN

Huntington disease (HD) is caused by a cytosine-adenine-guanine trinucleotide repeat expansion in the huntingtin gene, HTT, that results in expression of variant (mutant) huntingtin protein (HTT). Therapeutic strategies that reduce HTT levels are currently being pursued to slow or stop disease progression in people with HD. These approaches are supported by robust preclinical data indicating that reducing variant huntingtin protein is associated with decreased HD pathology. However, the risk-benefit profile of reducing either variant HTT or both variant and wild-type HTT is currently an open question that is being addressed in ongoing clinical trials. This review aims to examine the current data available regarding altered HTT in humans, normal animals, and animal models of HD. Studies indexed in PubMed were searched using the MeSH term Huntington disease or the text words huntington or huntingtin from August 31, 1999, to August 31, 2019, with no language restrictions. Additional studies were included from the reference lists of relevant studies and the authors' personal files. Articles describing at least 1 aspect of HTT reduction were included, prioritizing those published within the last 10 years. In vivo studies were also prioritized, with a focus on studies that examined the consequences of wild-type HTT reduction in adults. In a recently completed phase 1/2a study of RG6042 in 46 adults with early manifest HD, antisense oligonucleotide-mediated partial reduction of HTT was reported to be generally safe and well tolerated over the course of 4-monthly RG6042 doses. In case studies of people with rare genetic variations in huntingtin alleles, the loss of 1 wild-type allele was not associated with HD. People with homozygous cytosine-adenine-guanine expansions developed normally until the onset of HD, although they may have experienced a more aggressive disease course. In mouse models of HD, partial reduction of HTT was beneficial, with improvements in motor, cognitive, and behavioral phenotypes. The partial reduction of wild-type HTT in normal adult rodents and nonhuman primates was generally safe and well tolerated. The body of evidence reviewed in this article indicates a positive risk-benefit profile for the partial reduction of either variant HTT alone or both variant and wild-type HTT. These strategies target the underlying cause of HD and are currently being tested in several investigational clinical trials.


Asunto(s)
Proteína Huntingtina/antagonistas & inhibidores , Enfermedad de Huntington/terapia , Oligonucleótidos Antisentido/uso terapéutico , Animales , Humanos
19.
Neurotherapeutics ; 17(4): 1645-1659, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32705582

RESUMEN

Huntington's disease (HD) is a monogenic neurodegenerative disorder that presents with progressive motor, behavior, and cognitive symptoms leading to early disability and mortality. HD is caused by an expanded CAG repeats in exon 1 of the huntingtin (HTT) gene. The corresponding genetic test allows a clinical, definite diagnosis in life and the identification of a fully penetrant mutation carrier in a premanifest stage. In addition to the development of symptomatic treatments that attempt to address unmet care needs such as apathy, irritability, and cognition, novel therapies that target pathways specific to HD biology are being developed with the intent of slowing disease progression. Among these approaches, HTT protein lowering therapies hold great promise. There are currently active programs using antisense oligonucleotides (ASOs), RNA interference, small-molecule splicing modulators, and zinc-finger protein transcription factor. Except for ASOs and RNA interference approaches, the remaining therapeutic strategies are at a preclinical stage of development. While the current therapeutic landscape in HD may bring an unparalleled change in the lives of people with HD and their families with the first-ever disease-modifying therapy, the evaluation of these therapies requires novel tools that enable a more efficient and expedited discovery and evaluative process. Examples are biomarkers targeting the HTT protein to measure target engagement or disease progression and rating scales more sensitive to the earliest clinical changes. These tools will be instrumental in the next phase of disease-modifying clinical trials in HD likely to target the phenoconversion period of the disease, including the prodromal HD stage.


Asunto(s)
Desarrollo de Medicamentos/tendencias , Proteína Huntingtina/antagonistas & inhibidores , Proteína Huntingtina/genética , Enfermedad de Huntington/tratamiento farmacológico , Enfermedad de Huntington/genética , Desarrollo de Medicamentos/métodos , Humanos , Factores Inmunológicos/farmacología , Factores Inmunológicos/uso terapéutico , Oligonucleótidos Antisentido/farmacología , Oligonucleótidos Antisentido/uso terapéutico , Interferencia de ARN/efectos de los fármacos , Interferencia de ARN/fisiología , Ensayos Clínicos Controlados Aleatorios como Asunto/métodos
20.
Nucleic Acid Ther ; 29(5): 256-265, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31184975

RESUMEN

Huntington's disease is a neurodegenerative disorder caused by a CAG repeat expansion in the first exon of huntingtin gene (HTT) encoding for a toxic polyglutamine protein. This disease is characterized by motor, psychiatric, and cognitive impairments. Currently, there is no disease modifying treatment. However, reducing the expression of the huntingtin protein (HTT) using antisense oligonucleotides (ASOs) has been shown as a promising therapeutic strategy. In this study, we explore the therapeutic potential of ASO made of tricyclo-DNA (tcDNA), a conformationally constrained DNA analog, to silence HTT. We used a gapmer ASO, containing central DNA nucleotides flanked by tcDNA modifications on 5' and 3' ends, allowing the recruitment of RNAse H and subsequent degradation of the messenger RNA. After transfection of tcDNA-ASO in patient-derived fibroblast cell lines, we show a strong decrease of HTT mRNA and protein levels. As a control, 2'O-methyl-RNA targeting the same region of HTT was also tested and did not induce a significant effect. tcDNA-ASO were also evaluated in vivo in the YAC128 mice, containing the full-length human HTT gene with 128 CAG repeat expansion. Single intracerebroventricular (ICV) injections of tcDNA induce a significant decrease of HTT messenger and protein levels in the cortex, hippocampus, striatum, and cerebellum of treated mice. tcDNA-ASO were found well distributed in the central nervous system (CNS) and show long lasting effect with protein levels still low, 12 weeks after a single ICV injection. This proof of concept study suggests the therapeutic potential of gapmer tcDNA ASO to downregulate huntingtin in vitro and in vivo.


Asunto(s)
Proteína Huntingtina/genética , Enfermedad de Huntington/terapia , Proteínas Mutantes/genética , Oligonucleótidos Antisentido/farmacología , Animales , ADN sin Sentido/farmacología , Modelos Animales de Enfermedad , Exones/genética , Humanos , Proteína Huntingtina/antagonistas & inhibidores , Enfermedad de Huntington/genética , Enfermedad de Huntington/inmunología , Enfermedad de Huntington/patología , Ratones , Proteínas Mutantes/antagonistas & inhibidores , Oligonucleótidos Antisentido/genética , Ribonucleasa H/genética , Expansión de Repetición de Trinucleótido/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA