Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Blood Cells Mol Dis ; 84: 102456, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32498026

RESUMEN

ß-Hemoglobinopathies are among the most common single-gene disorders and are caused by different mutations in the ß-globin gene. Recent curative therapeutic approaches for these disorders utilize lentiviral vectors (LVs) to introduce a functional copy of the ß-globin gene into the patient's hematopoietic stem cells. Alternatively, fetal hemoglobin (HbF) can reduce or even prevent the symptoms of disease when expressed in adults. Thus, induction of HbF by means of LVs and other molecular approaches has become an alternative treatment of ß-hemoglobinopathies. Here, we performed a head-to-head comparative analysis of HbF-inducing LVs encoding for: 1) IGF2BP1, 2) miRNA-embedded shRNA (shmiR) sequences specific for the γ-globin repressor protein BCL11A, and 3) γ-globin gene. Furthermore, two novel baboon envelope proteins (BaEV)-LVs were compared to the commonly used vesicular-stomatitis-virus glycoprotein (VSV-G)-LVs. Therapeutic levels of HbF were achieved for all VSV-G-LV approaches, from a therapeutic level of 20% using γ-globin LVs to 50% for both IGF2BP1 and BCL11A-shmiR LVs. Contrarily, BaEV-LVs conferred lower HbF expression with a peak level of 13%, however, this could still ameliorate symptoms of disease. From this thorough comparative analysis of independent HbF-inducing LV strategies, we conclude that HbF-inducing VSV-G-LVs represent a promising alternative to ß-globin gene addition for patients with ß-hemoglobinopathies.


Asunto(s)
Hemoglobina Fetal/genética , Vectores Genéticos/genética , Hemoglobinopatías/terapia , Lentivirus/genética , Línea Celular , Células Cultivadas , Expresión Génica , Técnicas de Transferencia de Gen , Terapia Genética , Vectores Genéticos/administración & dosificación , Vectores Genéticos/uso terapéutico , Hemoglobinopatías/genética , Humanos , Transducción Genética , Regulación hacia Arriba , gamma-Globinas/genética
2.
Stem Cell Res ; 69: 103066, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36947995

RESUMEN

Mutations in Colony-stimulating factor 1 receptor (CSF1R) lead to CSF1R-related leukoencephalopathy, also known as Adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP), a rapidly progressing neurodegenerative disease with severe cognitive and motor impairment. In this study, a homozygous and a heterozygous CSF1R knockout induced pluripotent stem cell (iPSC) line were generated by CRISPR/Cas9-based gene editing. These in vitro models will provide a helpful tool for investigating the still largely unknown pathophysiology of CSF1R-related leukoencephalopathy.


Asunto(s)
Células Madre Pluripotentes Inducidas , Leucoencefalopatías , Enfermedades Neurodegenerativas , Adulto , Humanos , Enfermedades Neurodegenerativas/genética , Sistemas CRISPR-Cas/genética , Neuroglía , Leucoencefalopatías/genética , Mutación
3.
CRISPR J ; 6(1): 5-16, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36662546

RESUMEN

Cellular therapies hold enormous potential for the cure of severe hematological and oncological disorders. The forefront of innovative gene therapy approaches including therapeutic gene editing and hematopoietic stem cell transplantation needs to be processed by good manufacturing practice to ensure safe application in patients. In the present study, an effective transfection protocol for automated clinical-scale production of genetically modified hematopoietic stem and progenitor cells (HSPCs) using the CliniMACS Prodigy® system including the CliniMACS Electroporator (Miltenyi Biotec) was established. As a proof-of-concept, the enhancer of the BCL11A gene, clustered regularly interspaced short palindromic repeat (CRISPR) target in ongoing clinical trials for ß-thalassemia and sickle-cell disease treatment, was disrupted by the CRISPR-Cas9 system simulating a large-scale clinical scenario, yielding 100 million HSPCs with high editing efficiency. In vitro erythroid differentiation and high-performance liquid chromatography analyses corroborated fetal hemoglobin resurgence in edited samples, supporting the feasibility of running the complete process of HSPC gene editing in an automated closed system.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Hemoglobinopatías , Humanos , Edición Génica/métodos , Sistemas CRISPR-Cas/genética , Hemoglobinopatías/genética , Hemoglobinopatías/terapia , Células Madre Hematopoyéticas
4.
CRISPR J ; 5(1): 66-79, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34882002

RESUMEN

Metachromatic leukodystrophy (MLD) is a rare genetic disorder caused by mutations in the Arylsulfatase-A (ARSA) gene. The enzyme plays a key role in sulfatide metabolism in brain cells, and its deficiency leads to neurodegeneration. The clinical manifestations of MLD include stagnation and decline of motor and cognitive function, leading to premature death with limited standard treatment options. Here, we describe a mutation-agnostic hematopoietic stem and progenitor cell (HSPC) gene therapy using CRISPR-Cas9 and AAV6 repair template as a prospective treatment option for MLD. Our strategy achieved efficient insertions and deletions (>87%) and a high level of gene integration (>47%) at the ARSA locus in human bone marrow-derived HSPCs, with no detectable off-target editing. As a proof of concept, we tested our mutation-agnostic therapy in HSPCs derived from two MLD patients with distinct mutations and demonstrated restoration of ARSA enzyme activity (>30-fold improvement) equivalent to healthy adults. In summary, our investigation enabled a mutation-agnostic therapy for MLD patients with proven efficacy and strong potential for clinical translation.


Asunto(s)
Leucodistrofia Metacromática , Sistemas CRISPR-Cas/genética , Edición Génica , Terapia Genética , Células Madre Hematopoyéticas/metabolismo , Humanos , Leucodistrofia Metacromática/genética , Leucodistrofia Metacromática/terapia , Mutación , Estudios Prospectivos
5.
Genes (Basel) ; 13(12)2022 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-36553615

RESUMEN

X-linked severe combined immunodeficiency (X-SCID) is a primary immunodeficiency that is caused by mutations in the interleukin-2 receptor gamma (IL2RG) gene. Some patients present atypical X-SCID with mild clinical symptoms due to somatic revertant mosaicism. CRISPR/Cas9 and prime editing are two advanced genome editing tools that paved the way for treating immune deficiency diseases. Prime editing overcomes the limitations of the CRISPR/Cas9 system, as it does not need to induce double-strand breaks (DSBs) or exogenous donor DNA templates to modify the genome. Here, we applied CRISPR/Cas9 with single-stranded oligodeoxynucleotides (ssODNs) and prime editing methods to generate an in vitro model of the disease in K-562 cells and healthy donors' T cells for the c. 458T>C point mutation in the IL2RG gene, which also resulted in a useful way to optimize the gene correction approach for subsequent experiments in patients' cells. Both methods proved to be successful and were able to induce the mutation of up to 31% of treated K-562 cells and 26% of treated T cells. We also applied similar strategies to correct the IL2RG c. 458T>C mutation in patient T cells that carry the mutation with revertant somatic mosaicism. However, both methods failed to increase the frequency of the wild-type sequence in the mosaic T cells of patients due to limited in vitro proliferation of mutant cells and the presence of somatic reversion. To the best of our knowledge, this is the first attempt to treat mosaic cells from atypical X-SCID patients employing CRISPR/Cas9 and prime editing. We showed that prime editing can be applied to the formation of specific-point IL2RG mutations without inducing nonspecific on-target modifications. We hypothesize that the feasibility of the nucleotide substitution of the IL2RG gene using gene therapy, especially prime editing, could provide an alternative strategy to treat X-SCID patients without revertant mutations, and further technological improvements need to be developed to correct somatic mosaicism mutations.


Asunto(s)
Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X , Humanos , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia , Sistemas CRISPR-Cas/genética , Mosaicismo , Edición Génica/métodos , Terapia Genética/métodos
6.
CRISPR J ; 4(2): 207-222, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33876951

RESUMEN

Mutations in the human ß-globin gene are the cause of ß-hemoglobinopathies, one of the most common inherited single-gene blood disorders in the world. Novel therapeutic approaches are based on lentiviral vectors (LVs) or CRISPR-Cas9-mediated gene disruption to express adult hemoglobin (HbA), or to reactivate the completely functional fetal hemoglobin, respectively. Nonetheless, LVs present a risk of insertional mutagenesis, while gene-disrupting transcription factors (BCL11A, KLF1) involved in the fetal-to-adult hemoglobin switch might generate dysregulation of other cellular processes. Therefore, universal gene addition/correction approaches combining CRISPR-Cas9 and homology directed repair (HDR) by delivering a DNA repair template through adeno-associated virus could mitigate the limitations of both lentiviral gene transfer and gene disruption strategies, ensuring targeted integration and controlled transgene expression. In this study, we attained high rates of gene addition (up to 12%) and gene correction (up to 38%) in hematopoietic stem and progenitor cells from healthy donors without any cell sorting/enrichment or the application of HDR enhancers. Furthermore, these approaches were tested in heterozygous (ß0/ß+) and homozygous (ß0/ß0, ß+/ß+) ß-thalassemia patients, achieving a significant increase in HbA and demonstrating the universal therapeutic potential of this study for the treatment of ß-hemoglobinopathies.


Asunto(s)
Sistemas CRISPR-Cas , Dependovirus/genética , Terapia Genética , Hemoglobinopatías/genética , Hemoglobinopatías/terapia , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Dependovirus/metabolismo , Hemoglobina Fetal/genética , Hemoglobina Fetal/metabolismo , Edición Génica , Células Madre Hematopoyéticas , Humanos , Globinas beta/genética , Globinas beta/metabolismo , Talasemia beta/genética , Talasemia beta/metabolismo , Talasemia beta/terapia
7.
Ann N Y Acad Sci ; 1497(1): 27-38, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33682151

RESUMEN

In Chagas disease (ChD) caused by Trypanosoma cruzi, new biomarkers to predict chronic cardiac pathology are urgently needed. Previous studies in chagasic patients with mild symptomatology showed that antibodies against the immunodominant R3 epitope of sCha, a fragment of the human basic helix-loop-helix transcription factor like 5, correlated with cardiac pathology. To validate sCha as a biomarker and to understand the origin of anti-sCha antibodies, we conducted a multicenter study with several cohorts of chagasic patients with severe cardiac symptomatology. We found that levels of antibodies against sCha discriminated the high risk of sudden death, indicating they could be useful for ChD prognosis. We investigated the origin of the antibodies and performed an alanine scan of the R3 epitope. We identified a minimal epitope MRQLD, and a BLAST search retrieved several T. cruzi antigens. Five of the hits had known or putative functions, of which phosphonopyruvate decarboxylase showed the highest cross-reactivity with sCha, confirming the role of molecular mimicry in the development of anti-sCha antibodies. Altogether, we demonstrate that the development of antibodies against sCha, which originated by molecular mimicry with T. cruzi antigens, could discriminate electrocardiographic alterations associated with a high risk of sudden death.


Asunto(s)
Autoanticuerpos/inmunología , Cardiomiopatía Chagásica/etiología , Cardiomiopatía Chagásica/metabolismo , Enfermedad de Chagas/complicaciones , Enfermedad de Chagas/inmunología , Muerte Súbita/etiología , Epítopos Inmunodominantes/inmunología , Anticuerpos Antiprotozoarios/inmunología , Biomarcadores , Cardiomiopatía Chagásica/diagnóstico , Enfermedad de Chagas/parasitología , Enfermedad Crónica , Reacciones Cruzadas , Susceptibilidad a Enfermedades , Humanos , Trypanosoma cruzi/inmunología
8.
Blood Rev ; 40: 100641, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31761379

RESUMEN

Due to pioneering in vitro investigations on gene modification, gene engineering platforms have incredibly improved to a safer and more powerful tool for the treatment of multiple blood and immune disorders. Likewise, several clinical trials have been initiated combining autologous hematopoietic stem cell transplantation (auto-HSCT) with gene therapy (GT) tools. As several GT modalities such as lentivirus and gene editing tools have a long developmental path ahead to diminish its negative side effects, it is hard to decide which modality is optimal for treating a specific disease. Gene transfer by lentiviruses is the platform of choice for loss-of-mutation diseases, whereas gene correction/addition or gene disruption by gene editing tools, mainly CRISPR/Cas9, is likely to be more efficient in diseases where tight regulation is needed. Therefore, in this review, we compiled pertinent information about lentiviral gene transfer and CRISPR/Cas9 gene editing, their evolution to a safer platform for HSCT, and their applications on other types of gene disorders based on the etiology of the disease and cell fitness.


Asunto(s)
Edición Génica , Terapia Genética , Enfermedades Hematológicas , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas , Enfermedades del Sistema Inmune , Lentivirus , Autoinjertos , Enfermedades Hematológicas/genética , Enfermedades Hematológicas/terapia , Humanos , Enfermedades del Sistema Inmune/genética , Enfermedades del Sistema Inmune/terapia
9.
Sci Rep ; 10(1): 10133, 2020 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-32576837

RESUMEN

ß-hemoglobinopathies are caused by abnormal or absent production of hemoglobin in the blood due to mutations in the ß-globin gene (HBB). Imbalanced expression of adult hemoglobin (HbA) induces strong anemia in patients suffering from the disease. However, individuals with natural-occurring mutations in the HBB cluster or related genes, compensate this disparity through γ-globin expression and subsequent fetal hemoglobin (HbF) production. Several preclinical and clinical studies have been performed in order to induce HbF by knocking-down genes involved in HbF repression (KLF1 and BCL11A) or disrupting the binding sites of several transcription factors in the γ-globin gene (HBG1/2). In this study, we thoroughly compared the different CRISPR/Cas9 gene-disruption strategies by gene editing analysis and assessed their safety profile by RNA-seq and GUIDE-seq. All approaches reached therapeutic levels of HbF after gene editing and showed similar gene expression to the control sample, while no significant off-targets were detected by GUIDE-seq. Likewise, all three gene editing platforms were established in the GMP-grade CliniMACS Prodigy, achieving similar outcome to preclinical devices. Based on this gene editing comparative analysis, we concluded that BCL11A is the most clinically relevant approach while HBG1/2 could represent a promising alternative for the treatment of ß-hemoglobinopathies.


Asunto(s)
Anemia de Células Falciformes/genética , Sistemas CRISPR-Cas , Hemoglobina Fetal/genética , Edición Génica/métodos , Factores de Transcripción de Tipo Kruppel/genética , Proteínas Represoras/genética , gamma-Globinas/genética , Anemia de Células Falciformes/terapia , Antígenos CD34 , Células Cultivadas , Expresión Génica/genética , Humanos , Terapia Molecular Dirigida , Mutación
10.
Brief Funct Genomics ; 19(3): 191-200, 2020 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-31844895

RESUMEN

Chimeric antigen receptor (CAR)-modified T cells have raised among other immunotherapies for cancer treatment, being implemented against B-cell malignancies. Despite the promising outcomes of this innovative technology, CAR-T cells are not exempt from limitations that must yet to be overcome in order to provide reliable and more efficient treatments against other types of cancer. The purpose of this review is to shed light on the field of CAR-T cell gene editing for therapy universalization and further enhancement of antitumor function. Several studies have proven that the disruption of certain key genes is essential to boost immunosuppressive resistance, prevention of fratricide, and clinical safety. Due to its unparalleled simplicity, feasibility to edit multiple gene targets simultaneously, and affordability, CRISPR/CRISPR-associated protein 9 system has been proposed in different clinical trials for such CAR-T cell improvement. The combination of such powerful technologies is expected to provide a new generation of CAR-T cell-based immunotherapies for clinical application.


Asunto(s)
Sistemas CRISPR-Cas/genética , Edición Génica/métodos , Inmunoterapia/métodos , Humanos
11.
Bone Marrow Transplant ; 54(12): 1940-1950, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-30903024

RESUMEN

Allogeneic hematopoietic stem cell transplantation (HSCT) is a standard therapeutic intervention for hematological malignancies and several monogenic diseases. However, this approach has limitations related to lack of a suitable donor, graft-versus-host disease and infectious complications due to immune suppression. On the contrary, autologous HSCT diminishes the negative effects of allogeneic HSCT. Despite the good efficacy, earlier gene therapy trials with autologous HSCs and viral vectors have raised serious safety concerns. However, the CRISPR/Cas9-edited autologous HSCs have been proposed to be an alternative option with a high safety profile. In this review, we summarized the possibility of CRISPR/Cas9-mediated autologous HSCT as a potential treatment option for various diseases supported by preclinical gene-editing studies. Furthermore, we discussed future clinical perspectives and possible clinical grade improvements of CRISPR/cas9-mediated autologous HSCT.


Asunto(s)
Sistemas CRISPR-Cas/genética , Trasplante de Células Madre Hematopoyéticas/métodos , Trasplante de Células Madre/métodos , Acondicionamiento Pretrasplante/métodos , Trasplante Homólogo/métodos , Humanos
12.
Mol Cell Pediatr ; 5(1): 9, 2018 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-30430274

RESUMEN

BACKGROUND: ß-Thalassemia is an inherited hematological disorder caused by mutations in the human hemoglobin beta (HBB) gene that reduce or abrogate ß-globin expression. Although lentiviral-mediated expression of ß-globin and autologous transplantation is a promising therapeutic approach, the risk of insertional mutagenesis or low transgene expression is apparent. However, targeted gene correction of HBB mutations with programmable nucleases such as CRISPR/Cas9, TALENs, and ZFNs with non-viral repair templates ensures a higher safety profile and endogenous expression control. METHODS: We have compared three different gene-editing tools (CRISPR/Cas9, TALENs, and ZFNs) for their targeting efficiency of the HBB gene locus. As a proof of concept, we studied the personalized gene-correction therapy for a common ß-thalassemia splicing variant HBBIVS1-110 using Cas9 mRNA and several optimally designed single-stranded oligonucleotide (ssODN) donors in K562 and CD34+ hematopoietic stem cells (HSCs). RESULTS: Our results exhibited that indel frequency of CRISPR/Cas9 was superior to TALENs and ZFNs (P < 0.0001). Our designed sgRNA targeting the site of HBBIVS1-110 mutation showed indels in both K562 cells (up to 77%) and CD34+ hematopoietic stem cells-HSCs (up to 87%). The absolute quantification by next-generation sequencing showed that up to 8% site-specific insertion of the NheI tag was achieved using Cas9 mRNA and a chemically modified ssODN in CD34+ HSCs. CONCLUSION: Our approach provides guidance on non-viral gene correction in CD34+ HSCs using Cas9 mRNA and chemically modified ssODN. However, further optimization is needed to increase the homology directed repair (HDR) to attain a real clinical benefit for ß-thalassemia.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA