Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 794
Filtrar
Más filtros

Intervalo de año de publicación
1.
Immunol Rev ; 322(1): 148-156, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38033164

RESUMEN

Severe combined immune deficiency due to adenosine deaminase deficiency (ADA SCID) is an inborn error of immunity with pan-lymphopenia, due to accumulated cytotoxic adenine metabolites. ADA SCID has been treated using gene therapy with a normal human ADA gene added to autologous hematopoietic stem cells (HSC) for over 30 years. Iterative improvements in vector design, HSC processing methods, and clinical HSC transplant procedures have led nearly all ADA SCID gene therapy patients to achieve consistently beneficial immune restoration with stable engraftment of ADA gene-corrected HSC over the duration of observation (as long as 20 years). One gene therapy for ADA SCID is approved by the European Medicines Agency (EMA) in the European Union (EU) and another is being advanced to licensure in the U.S. and U.K. Despite the clear-cut benefits and safety of this curative gene and cell therapy, it remains challenging to achieve sustained availability and access, especially for rare disorders like ADA SCID.


Asunto(s)
Agammaglobulinemia , Trasplante de Células Madre Hematopoyéticas , Inmunodeficiencia Combinada Grave , Humanos , Inmunodeficiencia Combinada Grave/genética , Inmunodeficiencia Combinada Grave/terapia , Adenosina Desaminasa/genética , Terapia Genética/métodos
2.
Trends Genet ; 39(9): 646-648, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37429772

RESUMEN

Recent exciting developments in clustered regularly interspaced short palindromic repeats (CRISPR)-based genome editing showcase its potential to rapidly and efficiently edit genomes in planta, eliminating long processes of tissue culture and extensive breeding for crop improvement. These new methods offer heritable transgene-free edits in one generation, making them an attractive option for improving commercially important crops.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica , Sistemas CRISPR-Cas/genética , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Genoma de Planta/genética , Fitomejoramiento
3.
Annu Rev Genomics Hum Genet ; 24: 255-275, 2023 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-37624668

RESUMEN

Sickle cell disease (SCD) is a monogenic blood disease caused by a point mutation in the gene coding for ß-globin. The abnormal hemoglobin [sickle hemoglobin (HbS)] polymerizes under low-oxygen conditions and causes red blood cells to sickle. The clinical presentation varies from very severe (with acute pain, chronic pain, and early mortality) to normal (few complications and a normal life span). The variability of SCD might be due (in part) to various genetic modulators. First, we review the main genetic factors, polymorphisms, and modifier genes that influence the expression of globin or otherwise modulate the severity of SCD. Considering SCD as a complex, multifactorial disorder is important for the development of appropriate pharmacological and genetic treatments. Second, we review the characteristics, advantages, and disadvantages of the latest advances in gene therapy for SCD, from lentiviral-vector-based approaches to gene-editing strategies.


Asunto(s)
Dolor Agudo , Anemia de Células Falciformes , Dolor Crónico , Hemoglobinas Anormales , Humanos , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Eritrocitos
4.
Mol Ther ; 32(1): 84-102, 2024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-37952087

RESUMEN

So far, the mechanisms that impede AAV transduction, especially in the human heart, are poorly understood, hampering the introduction of new, effective gene therapy strategies. Therefore, the aim of this study was to identify and overcome the main cellular barriers to successful transduction in the heart, using induced pluripotent stem cell (iPSC)-derived cardiomyocytes (iPSC-CMs), iPSC-derived cardiac fibroblasts (iPSC-CFs), and primary endothelial cells to model vector-host interactions. Through phosphoproteome analysis we established that casein kinase 2 (CK2) signaling is one of the most significantly affected pathways upon AAV exposure. Transient inhibition of CK2 activity substantially enhanced the transduction rate of AAV2, AAV6, and AAV9 in all tested cell types. In particular, CK2 inhibition improved the trafficking of AAVs through the cytoplasm, impaired DNA damage response through destabilization of MRE11, and altered the RNA processing pathways, which were also highly responsive to AAV transduction. Also, it augmented transgene expression in already transduced iPSC-CFs, which retain AAV genomes in a functional, but probably silent form. In summary, the present study provides new insights into the current understanding of the host-AAV vector interaction, identifying CK2 activity as a key barrier to efficient transduction and transgene expression, which may translate to improving the outcome of AAV-based therapies in the future.


Asunto(s)
Quinasa de la Caseína II , Células Endoteliales , Humanos , Transducción Genética , Quinasa de la Caseína II/genética , Quinasa de la Caseína II/metabolismo , Terapia Genética , Transgenes , Dependovirus/genética , Dependovirus/metabolismo , Vectores Genéticos/genética
5.
Mol Ther ; 2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-39175195

RESUMEN

A major obstacle in inducing therapeutic angiogenesis in the heart is inefficient gene transfer to endothelial cells (ECs). Here, we identify compounds able to enhance the permissiveness of cardiac ECs to adeno-associated virus (AAV) vectors, which stand as ideal tools for in vivo gene delivery. We screened a library of >1,500 US Food and Drug Administration (FDA)-approved drugs, in combination with AAV vectors, in cardiac ECs. Among the top drugs increasing AAV-mediated transduction, we found vatalanib, an inhibitor of multiple tyrosine kinase receptors. The increased AAV transduction efficiency by vatalanib was paralleled by induction of the endothelial-to-mesenchymal transition, as documented by decreased endothelial and increased mesenchymal marker expression. Induction of the endothelial-to-mesenchymal transition by other strategies similarly increased EC permissiveness to AAV vectors. In vivo injection of AAV vectors in the heart after myocardial infarction resulted in the selective transduction of cells undergoing the endothelial-to-mesenchymal transition, which is known to happen transiently after cardiac ischemia. Collectively, these results point to the endothelial-to-mesenchymal transition as a mechanism for improving AAV transduction in cardiac ECs, with implications for both basic research and the induction of therapeutic angiogenesis in the heart.

6.
Mol Ther ; 32(5): 1202-1218, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38454604

RESUMEN

In recent years, a growing number of clinical trials have been initiated to evaluate gene therapy approaches for the treatment of patients with transfusion-dependent ß-thalassemia and sickle cell disease (SCD). Therapeutic modalities being assessed in these trials utilize different molecular techniques, including lentiviral vectors to add functional copies of the gene encoding the hemoglobin ß subunit in defective cells and CRISPR-Cas9, transcription activator-like effector protein nuclease, and zinc finger nuclease gene editing strategies to either directly address the underlying genetic cause of disease or induce fetal hemoglobin production by gene disruption. Here, we review the mechanisms of action of these various gene addition and gene editing approaches and describe the status of clinical trials designed to evaluate the potentially for these approaches to provide one-time functional cures to patients with transfusion-dependent ß-thalassemia and SCD.


Asunto(s)
Terapia Genética , Hemoglobinopatías , Animales , Humanos , Anemia de Células Falciformes/terapia , Anemia de Células Falciformes/genética , Talasemia beta/terapia , Talasemia beta/genética , Ensayos Clínicos como Asunto , Sistemas CRISPR-Cas , Edición Génica/métodos , Terapia Genética/métodos , Vectores Genéticos/genética , Vectores Genéticos/administración & dosificación , Hemoglobinopatías/terapia , Hemoglobinopatías/genética , Lentivirus/genética
7.
Mol Ther ; 32(10): 3470-3484, 2024 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-39113357

RESUMEN

One of the biggest challenges for in vivo gene therapy are vectors mediating highly selective gene transfer into a defined population of therapy-relevant cells. Here we present DARPin-targeted AAVs (DART-AAVs) displaying DARPins specific for human and murine CD8. Insertion of DARPins into the GH2/GH3 loop of the capsid protein 1 (VP1) of AAV2 and AAV6 resulted in high selectivity for CD8-positive T cells with unimpaired gene delivery activity. Remarkably, the capsid core structure was unaltered with protruding DARPins detectable. In complex primary cell mixtures, including donor blood or systemic injections into mice, the CD8-targeted AAVs were by far superior to unmodified AAV2 and AAV6 in terms of selectivity, target cell viability, and gene transfer rates. In vivo, up to 80% of activated CD8+ T cells were hit upon a single vector injection into conditioned humanized or immunocompetent mice. While gene transfer rates decreased significantly under non-activated conditions, genomic modification selectively in CD8+ T cells was still detectable upon Cre delivery into indicator mice. In both mouse models, selectivity for CD8+ T cells was close to absolute with exceptional detargeting from liver. The CD8-AAVs described here expand strategies for immunological research and in vivo gene therapy options.


Asunto(s)
Linfocitos T CD8-positivos , Dependovirus , Técnicas de Transferencia de Gen , Terapia Genética , Vectores Genéticos , Animales , Dependovirus/genética , Ratones , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Humanos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Terapia Genética/métodos , Transducción Genética , Proteínas de la Cápside/genética
8.
Mol Ther ; 32(5): 1328-1343, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38454603

RESUMEN

Vanishing white matter (VWM) is a fatal leukodystrophy caused by recessive mutations in subunits of the eukaryotic translation initiation factor 2B. Currently, there are no effective therapies for VWM. Here, we assessed the potential of adenine base editing to correct human pathogenic VWM variants in mouse models. Using adeno-associated viral vectors, we delivered intein-split adenine base editors into the cerebral ventricles of newborn VWM mice, resulting in 45.9% ± 5.9% correction of the Eif2b5R191H variant in the cortex. Treatment slightly increased mature astrocyte populations and partially recovered the integrated stress response (ISR) in female VWM animals. This led to notable improvements in bodyweight and grip strength in females; however, locomotor disabilities were not rescued. Further molecular analyses suggest that more precise editing (i.e., lower rates of bystander editing) as well as more efficient delivery of the base editors to deep brain regions and oligodendrocytes would have been required for a broader phenotypic rescue. Our study emphasizes the potential, but also identifies limitations, of current in vivo base-editing approaches for the treatment of VWM or other leukodystrophies.


Asunto(s)
Dependovirus , Modelos Animales de Enfermedad , Factor 2B Eucariótico de Iniciación , Edición Génica , Leucoencefalopatías , Fenotipo , Animales , Ratones , Factor 2B Eucariótico de Iniciación/genética , Factor 2B Eucariótico de Iniciación/metabolismo , Leucoencefalopatías/genética , Leucoencefalopatías/terapia , Leucoencefalopatías/patología , Dependovirus/genética , Humanos , Vectores Genéticos/genética , Vectores Genéticos/administración & dosificación , Femenino , Mutación , Terapia Genética/métodos , Sustancia Blanca/patología , Sustancia Blanca/metabolismo , Astrocitos/metabolismo
9.
J Cell Mol Med ; 28(17): e70056, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39245805

RESUMEN

Disruptions in normal development and the emergence of health conditions often result from the malfunction of vital genes in the human body. Decades of scientific research have focused on techniques to modify or substitute defective genes with healthy alternatives, marking a new era in disease treatment, prevention and cure. Recent strides in science and technology have reshaped our understanding of disorders, medication development and treatment recommendations, with human gene and cell therapy at the forefront of this transformative shift. Its primary objective is the modification of genes or adjustment of cell behaviour for therapeutic purposes. In this review, we focus on the latest advances in gene and cell therapy for treating human genetic diseases, with a particular emphasis on FDA and EMA-approved therapies and the evolving landscape of genome editing. We examine the current state of innovative gene editing technologies, particularly the CRISPR-Cas systems. As we explore the progress, ethical considerations and prospects of these innovations, we gain insight into their potential to revolutionize the treatment of genetic diseases, along with a discussion of the challenges associated with their regulatory pathways. This review traces the origins and evolution of these therapies, from conceptual ideas to practical clinical applications, marking a significant milestone in the field of medical science.


Asunto(s)
Sistemas CRISPR-Cas , Tratamiento Basado en Trasplante de Células y Tejidos , Edición Génica , Enfermedades Genéticas Congénitas , Terapia Genética , Humanos , Terapia Genética/métodos , Enfermedades Genéticas Congénitas/terapia , Enfermedades Genéticas Congénitas/genética , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Tratamiento Basado en Trasplante de Células y Tejidos/tendencias , Edición Génica/métodos , Animales
10.
J Gene Med ; 26(1): e3567, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37455676

RESUMEN

Sickle cell disease (SCD) results from a sequence defect in the ß-globin chain of adult hemoglobin (HbA) leading to expression of sickle hemoglobin (HbS). It is traditionally diagnosed by cellulose-acetate hemoglobin electrophoresis or high-performance liquid chromatography. While clinically useful, these methods have both sensitivity and specificity limitations. We developed a novel mass spectrometry (MS) method for the rapid, sensitive and highly quantitative detection of endogenous human ß-globin and sickle hß-globin, as well as lentiviral-encoded therapeutic hßAS3-globin in cultured cells and small quantities of mouse peripheral blood. The MS methods were used to phenotype homozygous HbA (AA), heterozygous HbA-HbS (AS) and homozygous HbS (SS) Townes SCD mice and detect lentiviral vector-encoded hßAS3-globin in transduced mouse erythroid cell cultures and transduced human CD34+ cells after erythroid differentiation. hßAS3-globin was also detected in peripheral blood 6 weeks post-transplant of transduced Townes SS bone marrow cells into syngeneic Townes SS mice and persisted for over 20 weeks post-transplant. As several genome-editing and gene therapy approaches for severe hemoglobin disorders are currently in clinical trials, this MS method will be useful for patient assessment before treatment and during follow-up.


Asunto(s)
Anemia de Células Falciformes , Lentivirus , Adulto , Ratones , Animales , Humanos , Lentivirus/genética , Vectores Genéticos/genética , Hemoglobinas/genética , Hemoglobinas/metabolismo , Anemia de Células Falciformes/diagnóstico , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/terapia , Globinas beta/genética , Células Cultivadas , Espectrometría de Masas
11.
Exp Eye Res ; 245: 109983, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38942133

RESUMEN

Over the past twenty years, ocular gene therapy has primarily focused on addressing diseases linked to various genetic factors. The eye is an ideal candidate for gene therapy due to its unique characteristics, such as easy accessibility and the ability to target both corneal and retinal conditions, including retinitis pigmentosa (RP), Leber congenital amaurosis (LCA), age-related macular degeneration (AMD), and Stargardt disease. Currently, literature documents 33 clinical trials in this field, with the most promising results emerging from trials focused on LCA. These successes have catalyzed further research into other ocular conditions such as glaucoma, AMD, RP, and choroideremia. The effectiveness of gene therapy relies on the efficient delivery of genetic material to specific cells, ensuring sustained and optimal gene expression over time. Viral vectors have been widely used for this purpose, although concerns about potential risks such as immune reactions and genetic mutations have led to the development of non-viral vector systems. Preliminary laboratory research and clinical investigations have shown a connection between vector dosage and the intensity of immune response and inflammation in the eye. The method of administration significantly influences these reactions, with subretinal delivery resulting in a milder humoral response compared to the intravitreal route. This review discusses various ophthalmic diseases, including both corneal and retinal conditions, and their underlying mechanisms, highlighting recent advances and applications in ocular gene therapies.


Asunto(s)
Terapia Genética , Vectores Genéticos , Humanos , Terapia Genética/métodos , Oftalmopatías/terapia , Oftalmopatías/genética , Técnicas de Transferencia de Gen , Enfermedades de la Retina/terapia , Enfermedades de la Retina/genética , Animales
12.
Anal Biochem ; 694: 115617, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39019206

RESUMEN

Data are presented demonstrating that absorbance detection can be used during high-speed sedimentation velocity analytical ultracentrifugation (hs-SV-AUC) experiments to characterize the size distribution of adeno-associated virus (AAV) drug products accurately. Advantages and limitations of being able to use this detector in this specific type of SV-AUC experiment are discussed.


Asunto(s)
Dependovirus , Ultracentrifugación , Dependovirus/genética , Dependovirus/aislamiento & purificación , Ultracentrifugación/métodos , Humanos
13.
Anal Biochem ; 689: 115482, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38342199

RESUMEN

Simulated SV-AUC data for an adeno-associated virus (AAV) sample consisting of four components having closely spaced sedimentation coefficients were used to develop a high-speed protocol that optimized the size distribution analysis resolution. The resulting high speed (45K rpm) SV-AUC (hs-SV-AUC) protocol poses several experimental challenges: 1) the need for rapid data acquisition, 2) increased potential for optical artifacts from steep and fast moving boundaries and 3) the increased potential for convection. To overcome these challenges the protocol uses interference detection at low temperatures and data that are confined to a limited radial-time window. In addition to providing higher resolution AAV SV-AUC data and very short run times (<20 min after temperature equilibration), the need to match the sample and reference solvent composition and meniscus positions is relaxed making interference detection as simple to employ as absorbance detection. Finally, experimental data comparing hs-SV-AUC (at 45K rpm) with standard low-speed (15K rpm) SV-AUC on the same AAV sample demonstrate the size distribution resolution improvement. These experiments also validate the use of a radial-time window and show how quickly data can be acquired using the hs-SV-AUC protocol.


Asunto(s)
Frío , Dependovirus , Dependovirus/genética , Área Bajo la Curva , Ultracentrifugación/métodos , Temperatura
14.
Eur Biophys J ; 53(3): 111-121, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38329496

RESUMEN

Sedimentation velocity analytical ultracentrifugation (SV-AUC) has long been an important method for characterization of antibody therapeutics. Recently, SV-AUC has experienced a wave of new interest and usage from the gene and cell therapy industry, where SV-AUC has proven itself to be the "gold standard" analytical approach for determining capsid loading ratios for adeno-associated virus (AAV) and other viral vectors. While other more common approaches have existed in the realm of cGMP-compliant techniques for years, SV-AUC has long been used strictly for characterization, but not for release testing. This manuscript describes the challenges faced in bringing SV-AUC to a cGMP environment and describes a new program, "BASIS", which allows for 21 CFR Part 11-compliant data handling and data analysis using the well-known and frequently cited SEDFIT analysis software.


Asunto(s)
Anticuerpos , Programas Informáticos , Área Bajo la Curva , Ultracentrifugación/métodos
15.
Nanotechnology ; 35(41)2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39008958

RESUMEN

The rise of gene therapy has solved many diseases that cannot be effectively treated by conventional methods. Gene vectors is very important to protect and deliver the therapeutic genes to the target site. Polyethyleneimine (PEI) modified with mannitol could enhance the gene transfection efficiency reported by our group previously. In order to further control and improve the effective gene release to action site, disulfide bonds were introduced into mannitol-modified PEI to construct new non-viral gene vectors PeiSM. The degrees of mannitol linking with disulfide bonds were screened. Among them, moderate mannitol-modified PEI with disulfide bonds showed the best transfection efficiency, and significantly enhanced long-term systemic transgene expression for 72 hin vivoeven at a single dose administration, and could promote caveolae-mediated uptake through up-regulating the phosphorylation of caveolin-1 and increase the loaded gene release from the nanocomplexes in high glutathione intracellular environment. This functionalized gene delivery system can be used as an potential and safe non-viral nanovector for further gene therapy.


Asunto(s)
Vectores Genéticos , Glutatión , Polietileneimina , Transfección , Polietileneimina/química , Transfección/métodos , Glutatión/metabolismo , Glutatión/química , Animales , Humanos , Vectores Genéticos/química , Vectores Genéticos/genética , Manitol/química , Ratones , Caveolina 1/metabolismo , Caveolina 1/genética , Terapia Genética/métodos , Técnicas de Transferencia de Gen , Disulfuros/química
16.
Mol Biol Rep ; 51(1): 563, 2024 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-38647711

RESUMEN

Adeno-associated virus (AAV) has emerged as a pivotal tool in neuroscience research, owing to its remarkable versatility and efficiency in delivering genetic material to diverse cell types within the nervous system. This mini review aims to underscore the advanced applications of AAV vectors in neuroscience and their profound potential to revolutionize our understanding of brain function and therapeutic interventions for neurological disorders. By providing a concise overview of the latest developments and strategies employing AAV vectors, this review illuminates the transformative role of AAV technology in unraveling the complexities of neural circuits and paving the way for innovative treatments. Through elucidating the multifaceted capabilities of AAV-mediated gene delivery, this review underscores its pivotal role as a cornerstone in contemporary neuroscience research, promising remarkable insights into the intricacies of brain biology and offering new avenues for therapeutic intervention.


Asunto(s)
Dependovirus , Técnicas de Transferencia de Gen , Terapia Genética , Vectores Genéticos , Neurociencias , Dependovirus/genética , Humanos , Vectores Genéticos/genética , Animales , Neurociencias/métodos , Terapia Genética/métodos , Encéfalo/virología , Encéfalo/metabolismo , Enfermedades del Sistema Nervioso/terapia , Enfermedades del Sistema Nervioso/virología , Enfermedades del Sistema Nervioso/genética
17.
Mol Biol Rep ; 51(1): 408, 2024 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-38460043

RESUMEN

BACKGROUND: To describe an oncolytic adenovirus (OAd) encoding SP-SA-E7-4-1BBL that is capable of inducing tumor regression in therapeutic assays. Herein, we tested whether the antitumor effect is given by the induction of a tumor-specific immune response, as well as the minimum dose needed to elicit antitumor protection and monitor the OAd biodistribution over time. METHODS AND RESULTS: C57BL/6 mice (n = 5) per group were immunized twice with OAds encoding SP-SA-E7-4-1BBL, SA-E7-4-1BBL, or SP-SA-4-1BBL and challenged with TC-1 cancer cells. The DNA construct SP-SA-E7-4-1BBL was employed as a control via biolistic or PBS injection. Groups without tumor development at 47 days were rechallenged with TC-1 cells, and follow-up lasted until day 90. The minimum dose of OAd to induce the antitumor effect was established by immunization using serial dilution doses. The cytometry bead assay and the ELISpot assay were used to evaluate cytokine release in response to ex vivo antigenic stimulation. The distribution profile of the OAd vaccine was evaluated in the different organs by histological, immunohistochemical and qPCR analyses. The OAd SP-SA-E7-4-1BBL-immunized mice did not develop tumors even in a rechallenge. A protective antitumor effect was observed from a dose that is one hundredth of most reports of adenoviral vaccines. Immunization with OAd increases Interferon-gamma-producing cells in response to antigen stimulation. OAd was detected in tumors over time, with significant morphological changes, contrary to nontumor tissues. CONCLUSIONS: The OAd SP-SA-E7-4-1BBL vaccine confers a prophylactic, safe, long-lasting, and antigen-dependent antitumor effect mediated by a Th1 antitumor immune response.


Asunto(s)
Vacunas contra el Cáncer , Neoplasias , Animales , Ratones , Papillomavirus Humano 16 , Ligando 4-1BB/genética , Ligando 4-1BB/farmacología , Distribución Tisular , Ratones Endogámicos C57BL , Adenoviridae/genética , Inmunidad , Neoplasias/terapia
18.
Brain ; 146(1): 149-166, 2023 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-35298632

RESUMEN

Huntington's disease is a fatal neurodegenerative disease characterized by striatal neurodegeneration, aggregation of mutant Huntingtin and the presence of reactive astrocytes. Astrocytes are important partners for neurons and engage in a specific reactive response in Huntington's disease that involves morphological, molecular and functional changes. How reactive astrocytes contribute to Huntington's disease is still an open question, especially because their reactive state is poorly reproduced in experimental mouse models. Here, we show that the JAK2-STAT3 pathway, a central cascade controlling astrocyte reactive response, is activated in the putamen of Huntington's disease patients. Selective activation of this cascade in astrocytes through viral gene transfer reduces the number and size of mutant Huntingtin aggregates in neurons and improves neuronal defects in two complementary mouse models of Huntington's disease. It also reduces striatal atrophy and increases glutamate levels, two central clinical outcomes measured by non-invasive magnetic resonance imaging. Moreover, astrocyte-specific transcriptomic analysis shows that activation of the JAK2-STAT3 pathway in astrocytes coordinates a transcriptional program that increases their intrinsic proteolytic capacity, through the lysosomal and ubiquitin-proteasome degradation systems. This pathway also enhances their production and exosomal release of the co-chaperone DNAJB1, which contributes to mutant Huntingtin clearance in neurons. Together, our results show that the JAK2-STAT3 pathway controls a beneficial proteostasis response in reactive astrocytes in Huntington's disease, which involves bi-directional signalling with neurons to reduce mutant Huntingtin aggregation, eventually improving disease outcomes.


Asunto(s)
Enfermedad de Huntington , Enfermedades Neurodegenerativas , Animales , Ratones , Enfermedad de Huntington/genética , Astrocitos/metabolismo , Proteostasis , Enfermedades Neurodegenerativas/patología , Neuronas/metabolismo , Proteína Huntingtina/genética , Proteína Huntingtina/metabolismo
19.
Mol Ther ; 31(1): 7-23, 2023 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-36196048

RESUMEN

Krabbe disease (KD) is a lysosomal storage disease (LSD) caused by mutations in the galc gene. There are over 50 monogenetic LSDs, which largely impede the normal development of children and often lead to premature death. At present, there are no cures for LSDs and the available treatments are generally insufficient, short acting, and not without co-morbidities or long-term side effects. The last 30 years have seen significant advances in our understanding of LSD pathology as well as treatment options. Two gene therapy-based clinical trials, NCT04693598 and NCT04771416, for KD were recently started based on those advances. This review will discuss how our knowledge of KD got to where it is today, focusing on preclinical investigations, and how what was discovered may prove beneficial for the treatment of other LSDs.


Asunto(s)
Leucodistrofia de Células Globoides , Enfermedades por Almacenamiento Lisosomal , Niño , Humanos , Leucodistrofia de Células Globoides/genética , Leucodistrofia de Células Globoides/terapia , Leucodistrofia de Células Globoides/patología , Terapia Combinada , Mutación , Enfermedades por Almacenamiento Lisosomal/genética , Enfermedades por Almacenamiento Lisosomal/terapia
20.
Mol Ther ; 31(10): 2901-2913, 2023 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-37550965

RESUMEN

Hematopoietic stem cell (HSC) gene therapy is currently performed on CD34+ hematopoietic stem and progenitor cells containing less than 1% true HSCs and requiring a highly specialized infrastructure for cell manufacturing and transplantation. We have previously identified the CD34+CD90+ subset to be exclusively responsible for short- and long-term engraftment. However, purification and enrichment of this subset is laborious and expensive. HSC-specific delivery agents for the direct modification of rare HSCs are currently lacking. Here, we developed novel targeted viral vectors to specifically transduce CD90-expressing HSCs. Anti-CD90 single chain variable fragments (scFvs) were engineered onto measles- and VSV-G-pseudotyped lentiviral vectors that were knocked out for native targeting. We further developed a custom hydrodynamic titration methodology to assess the loading of surface-engineered capsids, measure antigen recognition of the scFv, and predict the performance on cells. Engineered vectors formed with minimal impairment in the functional titer, maintained their ability to fuse with the target cells, and showed highly specific recognition of CD90 on cells ex vivo. Most important, targeted vectors selectively transduced human HSCs with secondary colony-forming potential. Our novel HSC-targeted viral vectors have the potential to significantly enhance the feasibility of ex vivo gene therapy and pave the way for future in vivo applications.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Humanos , Antígenos CD34/genética , Terapia Genética/métodos , Vectores Genéticos/genética , Células Madre Hematopoyéticas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA