Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 2343, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38491008

ABSTRACT

The intermitochondrial cement (IMC) and chromatoid body (CB) are posited as central sites for piRNA activity in mice, with MIWI initially assembling in the IMC for piRNA processing before translocating to the CB for functional deployment. The regulatory mechanism underpinning MIWI translocation, however, has remained elusive. We unveil that piRNA loading is the trigger for MIWI translocation from the IMC to CB. Mechanistically, piRNA loading facilitates MIWI release from the IMC by weakening its ties with the mitochondria-anchored TDRKH. This, in turn, enables arginine methylation of MIWI, augmenting its binding affinity for TDRD6 and ensuring its integration within the CB. Notably, loss of piRNA-loading ability causes MIWI entrapment in the IMC and its destabilization in male germ cells, leading to defective spermatogenesis and male infertility in mice. Collectively, our findings establish the critical role of piRNA loading in MIWI translocation during spermatogenesis, offering new insights into piRNA biology in mammals.


Subject(s)
Argonaute Proteins , Germ Cell Ribonucleoprotein Granules , Piwi-Interacting RNA , Animals , Male , Mice , Argonaute Proteins/metabolism , Germ Cells/metabolism , Mammals/genetics , Mitochondria/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Spermatogenesis/genetics , Testis/metabolism
2.
Sci China Life Sci ; 66(7): 1459-1481, 2023 07.
Article in English | MEDLINE | ID: mdl-37335463

ABSTRACT

PIWI-clade proteins harness piRNAs of 24-33 nt in length. Of great puzzles are how PIWI-clade proteins incorporate piRNAs of different sizes and whether the size matters to PIWI/piRNA function. Here we report that a PIWI-Ins module unique in PIWI-clade proteins helps define the length of piRNAs. Deletion of PIWI-Ins in Miwi shifts MIWI to load with shorter piRNAs and causes spermiogenic failure in mice, demonstrating the functional importance of this regulatory module. Mechanistically, we show that longer piRNAs provide additional complementarity to target mRNAs, thereby enhancing the assembly of the MIWI/eIF3f/HuR super-complex for translational activation. Importantly, we identify a c.1108C>T (p.R370W) mutation of HIWI (human PIWIL1) in infertile men and demonstrate in Miwi knock-in mice that this genetic mutation impairs male fertility by altering the property of PIWI-Ins in selecting longer piRNAs. These findings reveal a critical role of PIWI-Ins-ensured longer piRNAs in fine-tuning MIWI/piRNA targeting capacity, proven essential for spermatid development and male fertility.


Subject(s)
Piwi-Interacting RNA , Testis , Humans , Male , Mice , Animals , Testis/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Spermatogenesis/genetics , Proteins/metabolism , Fertility/genetics , Argonaute Proteins/genetics , Argonaute Proteins/metabolism
4.
Nat Commun ; 14(1): 207, 2023 01 13.
Article in English | MEDLINE | ID: mdl-36639729

ABSTRACT

In ß-thalassemia, either γ-globin induction to form fetal hemoglobin (α2γ2) or ß-globin repair to restore adult hemoglobin (α2ß2) could be therapeutic. ABE8e, a recently evolved adenine base editor variant, can achieve efficient adenine conversion, yet its application in patient-derived hematopoietic stem cells needs further exploration. Here, we purified ABE8e for ribonucleoprotein electroporation of ß-thalassemia patient CD34+ hematopoietic stem and progenitor cells to introduce nucleotide substitutions that upregulate γ-globin expression in the BCL11A enhancer or in the HBG promoter. We observed highly efficient on-target adenine base edits at these two regulatory regions, resulting in robust γ-globin induction. Moreover, we developed ABE8e-SpRY, a near-PAMless ABE variant, and successfully applied ABE8e-SpRY RNP to directly correct HbE and IVS II-654 mutations in patient-derived CD34+ HSPCs. Finally, durable therapeutic editing was produced in self-renewing repopulating human HSCs as assayed in primary and secondary recipients. Together, these results support the potential of ABE-mediated base editing in HSCs to treat inherited monogenic blood disorders.


Subject(s)
Gene Editing , beta-Thalassemia , Humans , Adenine/metabolism , Antigens, CD34/genetics , Antigens, CD34/metabolism , beta-Thalassemia/genetics , CRISPR-Cas Systems/genetics , Fetal Hemoglobin/genetics , gamma-Globins/genetics , Gene Editing/methods , Hematopoietic Stem Cells/metabolism
5.
Adv Exp Med Biol ; 1442: 177-199, 2023.
Article in English | MEDLINE | ID: mdl-38228965

ABSTRACT

Hematopoietic stem cells (HSCs) can be isolated and collected from the body, genetically modified, and expanded ex vivo. The invention of innovative and powerful gene editing tools has provided researchers with great convenience in genetically modifying a wide range of cells, including hematopoietic stem and progenitor cells (HSPCs). In addition to being used to modify genes to study the functional role that specific genes play in the hematopoietic system, the application of gene editing platforms in HSCs is largely focused on the development of cell-based gene editing therapies to treat diseases such as immune deficiency disorders and inherited blood disorders. Here, we review the application of gene editing tools in HSPCs. In particular, we provide a broad overview of the development of gene editing tools, multiple strategies for the application of gene editing tools in HSPCs, and exciting clinical advances in HSPC gene editing therapies. We also outline the various challenges integral to clinical translation of HSPC gene editing and provide the possible corresponding solutions.


Subject(s)
Hematologic Diseases , Hematopoietic Stem Cell Transplantation , Humans , Gene Editing , Hematopoietic Stem Cells
6.
Nat Med ; 28(8): 1573-1580, 2022 08.
Article in English | MEDLINE | ID: mdl-35922667

ABSTRACT

Gene editing to disrupt the GATA1-binding site at the +58 BCL11A erythroid enhancer could induce γ-globin expression, which is a promising therapeutic strategy to alleviate ß-hemoglobinopathy caused by HBB gene mutation. In the present study, we report the preliminary results of an ongoing phase 1/2 trial (NCT04211480) evaluating safety and efficacy of gene editing therapy in children with blood transfusion-dependent ß-thalassemia (TDT). We transplanted BCL11A enhancer-edited, autologous, hematopoietic stem and progenitor cells into two children, one carrying the ß0/ß0 genotype, classified as the most severe type of TDT. Primary endpoints included engraftment, overall survival and incidence of adverse events (AEs). Both patients were clinically well with multilineage engraftment, and all AEs to date were considered unrelated to gene editing and resolved after treatment. Secondary endpoints included achieving transfusion independence, editing rate in bone marrow cells and change in hemoglobin (Hb) concentration. Both patients achieved transfusion independence for >18 months after treatment, and their Hb increased from 8.2 and 10.8 g dl-1 at screening to 15.0 and 14.0 g dl-1 at the last visit, respectively, with 85.46% and 89.48% editing persistence in bone marrow cells. Exploratory analysis of single-cell transcriptome and indel patterns in edited peripheral blood mononuclear cells showed no notable side effects of the therapy.


Subject(s)
Gene Editing , beta-Thalassemia , CRISPR-Cas Systems/genetics , Child , Gene Editing/methods , Humans , Leukocytes, Mononuclear/metabolism , Repressor Proteins/genetics , beta-Globins/genetics , beta-Thalassemia/genetics , beta-Thalassemia/therapy , gamma-Globins/genetics
7.
Cell Rep ; 31(3): 107521, 2020 04 21.
Article in English | MEDLINE | ID: mdl-32320667

ABSTRACT

Recent studies have revealed an essential role for embryonic cortical development in the pathophysiology of neurodevelopmental disorders, including autism spectrum disorder (ASD). However, the genetic basis and underlying mechanisms remain unclear. Here, we generate mutant human embryonic stem cell lines (Mut hESCs) carrying an NR2F1-R112K mutation that has been identified in a patient with ASD features and investigate their neurodevelopmental alterations. Mut hESCs overproduce ventral telencephalic neuron progenitors (ventral NPCs) and underproduce dorsal NPCs, causing the imbalance of excitatory/inhibitory neurons. These alterations can be mainly attributed to the aberrantly activated Hedgehog signaling pathway. Moreover, the corresponding Nr2f1 point-mutant mice display a similar excitatory/inhibitory neuron imbalance and abnormal behaviors. Antagonizing the increased inhibitory synaptic transmission partially alleviates their behavioral deficits. Together, our results suggest that the NR2F1-dependent imbalance of excitatory/inhibitory neuron differentiation caused by the activated Hedgehog pathway is one precursor of neurodevelopmental disorders and may enlighten the therapeutic approaches.


Subject(s)
Autism Spectrum Disorder/metabolism , COUP Transcription Factor I/metabolism , Hedgehog Proteins/metabolism , Neurodevelopmental Disorders/metabolism , Neurons/metabolism , Neurons/pathology , Point Mutation , Animals , Autism Spectrum Disorder/genetics , COUP Transcription Factor I/genetics , Cell Differentiation/physiology , Humans , Mice , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/pathology , Signal Transduction
9.
Cell Res ; 29(11): 911-926, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31591447

ABSTRACT

Primary germ layers have the potential to form all tissues in the mature organism, and their formation during gastrulation requires precise epigenetic modulation of both proximal and distal regulatory elements. Previous studies indicated that spatial and temporal patterns of gene expression in the gastrula predispose individual regions to distinct cell fates. However, the underlying epigenetic mechanisms remain largely unexplored. Here, we profile the spatiotemporal landscape of the epigenome and transcriptome of the mouse gastrula. We reveal the asynchronous dynamics of proximal chromatin states during germ layer formation as well as unique gastrula-specific epigenomic features of regulatory elements, which have strong usage turnover dynamics and clear germ layer-specific signatures. Importantly, we also find that enhancers around organogenetic genes, which are weakly expressed at the gastrulation stage, are frequently pre-marked by histone H3 lysine 27 acetylation (H3K27ac) in the gastrula. By using the transgenic mice and genome editing system, we demonstrate that a pre-marked enhancer, which is located in the intron of a brain-specific gene 2510009E07Rik, exhibits specific enhancer activity in the ectoderm and future brain tissue, and also executes important function during mouse neural differentiation. Taken together, our study provides the comprehensive epigenetic information for embryonic patterning during mouse gastrulation, demonstrates the importance of gastrula pre-marked enhancers in regulating the correct development of the mouse embryo, and thus broadens the current understanding of mammalian embryonic development and related diseases.


Subject(s)
Enhancer Elements, Genetic/physiology , Epigenesis, Genetic , Gastrula/embryology , Gastrulation/genetics , Gene Expression Regulation, Developmental , Germ Layers/embryology , Animals , Brain/embryology , Brain/metabolism , Cells, Cultured , Embryo, Mammalian , Embryonic Stem Cells , Female , Gastrula/cytology , Gastrula/metabolism , Germ Layers/cytology , Germ Layers/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurogenesis/genetics , Transcriptome
10.
PLoS Biol ; 17(7): e3000350, 2019 07.
Article in English | MEDLINE | ID: mdl-31265461

ABSTRACT

Mutagenic screening is powerful for identifying key genes involved in developmental processes. However, such screens are successful only in lower organisms. Here, we develop a targeted genetic screening approach in mice through combining androgenetic haploid embryonic stem cells (AG-haESCs) and clustered regularly interspaced palindromic repeats/CRISPR-associated protein 9 (CRISPR-Cas9) technology. We produced a mutant semi-cloned (SC) mice pool by oocyte injection of AG-haESCs carrying constitutively expressed Cas9 and an single guide RNA (sgRNA) library targeting 72 preselected genes in one step and screened for bone-development-related genes through skeletal analysis at birth. This yielded 4 genes: Zic1 and Clec11a, which are required for bone development, and Rln1 and Irx5, which had not been previously considered. Whereas Rln1-/- mice exhibited small skeletal size only at birth, Irx5-/- mice showed skeletal abnormalities both in postnatal and adult phases due to decreased bone mass and increased bone marrow adipogenesis. Mechanistically, iroquois homeobox 5 (IRX5) promotes osteoblastogenesis and inhibits adipogenesis by suppressing peroxisome proliferator activated receptor γ (PPARγ) activation. Thus, AG-haESC-mediated functional mutagenic screening opens new avenues for genetic interrogation of developmental processes in mice.


Subject(s)
Bone Development/genetics , Gene Expression Regulation, Developmental , Gene Targeting/methods , Genetic Testing/methods , Mouse Embryonic Stem Cells/metabolism , Animals , CRISPR-Cas Systems , Cells, Cultured , Haploidy , Hematopoietic Cell Growth Factors/genetics , Hematopoietic Cell Growth Factors/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Mice , Mice, Knockout , Relaxin/genetics , Relaxin/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
11.
Sci Rep ; 8(1): 7629, 2018 May 11.
Article in English | MEDLINE | ID: mdl-29752475

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

12.
Sci Rep ; 7(1): 5282, 2017 07 13.
Article in English | MEDLINE | ID: mdl-28706241

ABSTRACT

The paraventricular nucleus of hypothalamus plays important roles in the regulation of energy balance and fetal growth. However, the molecular mechanisms underlying its formation and function have not been clearly elucidated. Various mutations in the human COUP-TFII gene, which encodes a nuclear receptor, result in growth retardation, congenital diaphragmatic hernia and congenital heart defects. Here, we show that COUP-TFII gene is expressed in the developing hypothalamus in mouse. The ventral forebrain-specific RXCre/+; COUP-TFII F/F mutant mice display growth retardation. The development of the paraventricular nucleus of hypothalamus is compromised in the COUP-TFII mutant mainly because of increased apoptosis and mis-migration of the Brn2+ neurons. Moreover, hypoplastic anterior pituitary with blood cell clusters and shrunken posterior pituitary lacking AVP/OT neuron innervations are observed in the mutant, indicating the failure of formation of the hypothalamic-pituitary axis. Mechanistic studies show that the expression of Bdnf and Nrp1 genes is reduced in the mutant embryo, and that Bdnf is a direct downstream target of the COUP-TFII protein. Thus, our findings provide a novel functional validation that COUP-TFII gene promotes the expression of Bdnf and Nrp1 genes to ensure the appropriate morphogenesis of the hypothalamic-pituitary axis, especially the paraventricular nucleus of hypothalamus, and to prevent growth retardation.


Subject(s)
COUP Transcription Factor II/physiology , Gene Expression Regulation, Developmental , Growth Disorders/pathology , Nerve Tissue Proteins/metabolism , Neurons/pathology , POU Domain Factors/metabolism , Paraventricular Hypothalamic Nucleus/pathology , Animals , Growth Disorders/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Morphogenesis , Nerve Tissue Proteins/genetics , Neurons/metabolism , POU Domain Factors/genetics , Paraventricular Hypothalamic Nucleus/metabolism
13.
Cell Res ; 27(6): 815-829, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28429771

ABSTRACT

Studying the early function of essential genes is an important and challenging problem in developmental biology. Here, we established a method for rapidly inducing CRISPR-Cas9-mediated mutations in one blastomere of two-cell stage embryos, termed 2-cell embryo-CRISPR-Cas9 injection (2CC), to study the in vivo function of essential (or unknown) genes in founder chimeric mice. By injecting both Cre mRNA and CRISPR-Cas9 targeting the gene of interest into fluorescent reporter mice, the 2CC method can trace both wild-type and mutant cells at different developmental stages, offering internal control for phenotypic analyses of mutant cells. Using this method, we identified novel functions of the essential gene Tet3 in regulating excitatory and inhibitory synaptic transmission in the developing mouse cerebral cortex. By generating chimeric mutant mice, the 2CC method allows for the rapid screening of gene function in multiple tissues and cell types in founder chimeric mice, significantly expanding the current armamentarium of genetic tools.


Subject(s)
Blastomeres/metabolism , CRISPR-Cas Systems/physiology , Gene Editing/methods , Animals , CRISPR-Cas Systems/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dioxygenases , Embryo, Mammalian/metabolism , Genetic Engineering/methods , Male , Mice , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism
15.
Cell Res ; 25(1): 67-79, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25475058

ABSTRACT

Spermatogonial stem cells (SSCs) can produce numerous male gametes after transplantation into recipient testes, presenting a valuable approach for gene therapy and continuous production of gene-modified animals. However, successful genetic manipulation of SSCs has been limited, partially due to complexity and low efficiency of currently available genetic editing techniques. Here, we show that efficient genetic modifications can be introduced into SSCs using the CRISPR-Cas9 system. We used the CRISPR-Cas9 system to mutate an EGFP transgene or the endogenous Crygc gene in SCCs. The mutated SSCs underwent spermatogenesis after transplantation into the seminiferous tubules of infertile mouse testes. Round spermatids were generated and, after injection into mature oocytes, supported the production of heterozygous offspring displaying the corresponding mutant phenotypes. Furthermore, a disease-causing mutation in Crygc (Crygc(-/-)) that pre-existed in SSCs could be readily repaired by CRISPR-Cas9-induced nonhomologous end joining (NHEJ) or homology-directed repair (HDR), resulting in SSC lines carrying the corrected gene with no evidence of off-target modifications as shown by whole-genome sequencing. Fertilization using round spermatids generated from these lines gave rise to offspring with the corrected phenotype at an efficiency of 100%. Our results demonstrate efficient gene editing in mouse SSCs by the CRISPR-Cas9 system, and provide the proof of principle of curing a genetic disease via gene correction in SSCs.


Subject(s)
Adult Stem Cells/metabolism , Adult Stem Cells/transplantation , CRISPR-Cas Systems , Infertility, Male/genetics , Infertility, Male/therapy , Mutagenesis , Animals , Cells, Cultured , DNA End-Joining Repair , Female , Genes, erbB-1 , Genetic Therapy , Male , Mice , Mice, Inbred BALB C , Spermatogenesis , Transgenes , gamma-Crystallins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...