Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
Mol Biotechnol ; 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38300454

ABSTRACT

Lentiviral vector (LVV) has been used as one of the common carriers for gene therapy in clinical trials. LVV-mediated clinical trials have being reported in successfully treating hundreds of ß-thalassemia cases. These LVVs bear an inversely placed ß-hemoglobin (HBB) gene expression cassette for preserving introns during the viral RNA packaging. Consequently, these LVVs often produce a small amount of negatively orientated transcript driven by its internal gene promoter and would lower the viral titer by the minus-strand complemented with the viral backbone. To overcome this problem, we designed shRNAs specifically target the minus-strand RNA driven by the LVV internal promoter that resulted in a notable increase in the viral titer. This report demonstrates a simple and positive mean for increasing the effectiveness for gene therapy with the LVV system.

2.
J Gene Med ; 26(1): e3640, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37989259

ABSTRACT

BACKGROUND: LentiGlobin BB305 is a self-inactivating lentiviral vector carrying a human ß-globin expressing cassette for treating ß-thalassemia. Initially, a 2 × 250 bp chicken Locus Control Region fragment of cHS4, functioning as an insulator, was placed at its ΔU3, which was removed after the first clinical trial led by a French team to avoid abnormal splicing, etc. This action could potentially lead to an increasing risk of the transcriptional read-through rate driven by the ß-globin promoter to a significant level, posing a biosafety risk in clinical trials. METHODS: In the present study, a read-through reducing agent (C-U+ or WPRE) was designed to be placed at the 3' UTR of the ß-globin gene. The Enhancer Activities and/or Transcriptional Read-Through (EATRT) rate at the mRNA level and the protein expression level regarding lentiviral preparation titer were examined. RESULTS: We found that the insertion of the element (C-U+ or WPRE) reduced the EATRT effectively by 53% or 41%, respectively. C-U+ has less impact on virus package efficiency. Furthermore, there was no significant difference in the protein expression level after the C-U+ or WPRE insertion. CONCLUSIONS: The results of the present study show that inserting C-U+ or WPRE before the polyA sequence of the BB305 would reduce the EATRT rate at no cost of its expressing efficacy and viral preparation titers. Thus, we present an alternative improvement for a safer lentiviral vector for ß-thalassemia clinical trials.


Subject(s)
beta-Thalassemia , Humans , beta-Thalassemia/genetics , beta-Thalassemia/therapy , Lentivirus/genetics , Genetic Vectors/genetics , Genetic Therapy/methods , beta-Globins/genetics
3.
Nat Commun ; 14(1): 6489, 2023 10 14.
Article in English | MEDLINE | ID: mdl-37838801

ABSTRACT

The hippocampus plays major roles in learning and memory, and its formation requires precise coordination of patterning, cell proliferation, differentiation, and migration. Here we removed the chromatin-association capability of KDM2B in the progenitors of developing dorsal telencephalon (Kdm2b∆CxxC) to discover that Kdm2b∆CxxC hippocampus, particularly the dentate gyrus, became drastically smaller with disorganized cellular components and structure. Kdm2b∆CxxC mice display prominent defects in spatial memory, motor learning and fear conditioning, resembling patients with KDM2B mutations. The migration and differentiation of neural progenitor cells is greatly impeded in the developing Kdm2b∆CxxC hippocampus. Mechanism studies reveal that Wnt signaling genes in developing Kdm2b∆CxxC hippocampi are de-repressed due to reduced enrichment of repressive histone marks by polycomb repressive complexes. Activating the Wnt signaling disturbs hippocampal neurogenesis, recapitulating the effect of KDM2B loss. Together, we unveil a previously unappreciated gene repressive program mediated by KDM2B that controls progressive fate specifications and cell migration, hence morphogenesis of the hippocampus.


Subject(s)
Hippocampus , Wnt Signaling Pathway , Animals , Humans , Mice , Cell Differentiation/genetics , Neurogenesis/genetics , Polycomb-Group Proteins , Wnt Signaling Pathway/genetics
4.
Neurosci Bull ; 37(2): 183-200, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33196962

ABSTRACT

Long non-coding RNAs (lncRNAs) regulate transcription to control development and homeostasis in a variety of tissues and organs. However, their roles in the development of the cerebral cortex have not been well elucidated. Here, a bioinformatics pipeline was applied to delineate the dynamic expression and potential cis-regulating effects of mouse lncRNAs using transcriptome data from 8 embryonic time points and sub-regions of the developing cerebral cortex. We further characterized a sense lncRNA, SenZfp536, which is transcribed downstream of and partially overlaps with the protein-coding gene Zfp536. Both SenZfp536 and Zfp536 were predominantly expressed in the proliferative zone of the developing cortex. Zfp536 was cis-regulated by SenZfp536, which facilitates looping between the promoter of Zfp536 and the genomic region that transcribes SenZfp536. Surprisingly, knocking down or activating the expression of SenZfp536 increased or compromised the proliferation of cortical neural progenitor cells (NPCs), respectively. Finally, overexpressing Zfp536 in cortical NPCs reversed the enhanced proliferation of cortical NPCs caused by SenZfp536 knockdown. The study deepens our understanding of how lncRNAs regulate the propagation of cortical NPCs through cis-regulatory mechanisms.


Subject(s)
Neural Stem Cells , RNA, Long Noncoding , Animals , Cerebral Cortex , Gene Expression Profiling , Mice , RNA, Long Noncoding/genetics , Transcriptome
5.
J Mol Cell Biol ; 12(5): 345-358, 2020 06 11.
Article in English | MEDLINE | ID: mdl-31504682

ABSTRACT

The release and nuclear translocation of the intracellular domain of Notch receptor (NICD) is the prerequisite for Notch signaling-mediated transcriptional activation. NICD is subjected to various posttranslational modifications including ubiquitination. Here, we surprisingly found that NUMB proteins stabilize the intracellular domain of NOTCH1 receptor (N1ICD) by regulating the ubiquitin-proteasome machinery, which is independent of NUMB's role in modulating endocytosis. BAP1, a deubiquitinating enzyme (DUB), was further identified as a positive N1ICD regulator, and NUMB facilitates the association between N1ICD and BAP1 to stabilize N1ICD. Intriguingly, BAP1 stabilizes N1ICD independent of its DUB activity but relying on the BRCA1-inhibiting function. BAP1 strengthens Notch signaling and maintains stem-like properties of cortical neural progenitor cells. Thus, NUMB enhances Notch signaling by regulating the ubiquitinating activity of the BAP1-BRCA1 complex.


Subject(s)
Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Receptor, Notch1/chemistry , Receptor, Notch1/metabolism , Signal Transduction , Ubiquitination , BRCA1 Protein/metabolism , Cell Self Renewal , HEK293 Cells , HeLa Cells , Humans , Membrane Proteins/chemistry , Nerve Tissue Proteins/chemistry , Neural Stem Cells/metabolism , Protein Domains , Protein Stability , Tumor Suppressor Proteins/metabolism , Ubiquitin Thiolesterase/metabolism
6.
Protein Cell ; 11(3): 161-186, 2020 03.
Article in English | MEDLINE | ID: mdl-31317506

ABSTRACT

The mechanisms underlying spatial and temporal control of cortical neurogenesis of the brain are largely elusive. Long non-coding RNAs (lncRNAs) have emerged as essential cell fate regulators. Here we found LncKdm2b (also known as Kancr), a lncRNA divergently transcribed from a bidirectional promoter of Kdm2b, is transiently expressed during early differentiation of cortical projection neurons. Interestingly, Kdm2b's transcription is positively regulated in cis by LncKdm2b, which has intrinsic-activating function and facilitates a permissive chromatin environment at the Kdm2b's promoter by associating with hnRNPAB. Lineage tracing experiments and phenotypic analyses indicated LncKdm2b and Kdm2b are crucial in proper differentiation and migration of cortical projection neurons. These observations unveiled a lncRNA-dependent machinery in regulating cortical neuronal differentiation.


Subject(s)
Cerebral Cortex/cytology , F-Box Proteins/physiology , Jumonji Domain-Containing Histone Demethylases/physiology , Neurogenesis , Neurons/metabolism , RNA, Long Noncoding/physiology , Animals , Cell Lineage , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Neural Stem Cells/cytology , Neurons/cytology
7.
J Neurosci Res ; 94(9): 814-24, 2016 09.
Article in English | MEDLINE | ID: mdl-27317429

ABSTRACT

In utero electroporation (IUE) is commonly used to study cortical development of cerebrum by downregulating or overexpressing genes of interest in neural progenitor cells (NPCs) of small mammals. However, exogenous plasmids are lost or diluted over time. Furthermore, gene knockdown based on short-hairpin RNAs may exert nonspecific effects that lead to aberrant neuronal migration. Genomic engineering by the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated 9 (Cas9) system has great research and therapeutic potentials. Here we integrate the CRISPR/Cas9 components into the piggyBac (PB) transposon system (the CRISPR/Cas9-PB toolkit) for cortical IUEs. The mouse Sry-related HMG box-2 (Sox2) gene was selected as the target for its application. Most transduced cortical NPCs were depleted of SOX2 protein as early as 3 days post-IUE, whereas expressions of SOX1 and PAX6 remained intact. Furthermore, both the WT Cas9 and the D10A nickase mutant Cas9n showed comparable knockout efficiency. Transduced cortical cells were purified with fluorescence-activated cell sorting, and effective gene editing at the Sox2 loci was confirmed. Thus, application of the CRISPR/Cas9-PB toolkit in IUE is a promising strategy to study gene functions in cortical NPCs and their progeny. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cerebral Cortex/growth & development , Clustered Regularly Interspaced Short Palindromic Repeats , DNA Transposable Elements/genetics , Electroporation/methods , Gene Knockout Techniques/methods , Neurology/methods , Animals , Cerebral Cortex/embryology , Female , Fetal Development , Fetus , Mice , Mice, Inbred ICR , Mice, Knockout , Neural Stem Cells , PAX6 Transcription Factor/biosynthesis , PAX6 Transcription Factor/genetics , Plasmids , Pregnancy , Protein Engineering , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...