Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Am J Hum Genet ; 2024 Aug 24.
Article in English | MEDLINE | ID: mdl-39226899

ABSTRACT

The BAF chromatin remodeler regulates lineage commitment including cranial neural crest cell (CNCC) specification. Variants in BAF subunits cause Coffin-Siris syndrome (CSS), a congenital disorder characterized by coarse craniofacial features and intellectual disability. Approximately 50% of individuals with CSS harbor variants in one of the mutually exclusive BAF subunits, ARID1A/ARID1B. While Arid1a deletion in mouse neural crest causes severe craniofacial phenotypes, little is known about the role of ARID1A in CNCC specification. Using CSS-patient-derived ARID1A+/- induced pluripotent stem cells to model CNCC specification, we discovered that ARID1A-haploinsufficiency impairs epithelial-to-mesenchymal transition (EMT), a process necessary for CNCC delamination and migration from the neural tube. Furthermore, wild-type ARID1A-BAF regulates enhancers associated with EMT genes. ARID1A-BAF binding at these enhancers is impaired in heterozygotes while binding at promoters is unaffected. At the sequence level, these EMT enhancers contain binding motifs for ZIC2, and ZIC2 binding at these sites is ARID1A-dependent. When excluded from EMT enhancers, ZIC2 relocates to neuronal enhancers, triggering aberrant neuronal gene activation. In mice, deletion of Zic2 impairs NCC delamination, while ZIC2 overexpression in chick embryos at post-migratory neural crest stages elicits ectopic delamination from the neural tube. These findings reveal an essential ARID1A-ZIC2 axis essential for EMT and CNCC delamination.

2.
Genesis ; 55(10)2017 10.
Article in English | MEDLINE | ID: mdl-28875532

ABSTRACT

The canonical Wnt signalling pathway has been implicated in organogenesis and self-renewal of essentially all stem cell systems. In vivo reporter systems are crucial to assess the role of Wnt signalling in the biology and pathology of stem cell systems. We set out to develop a Turquoise (TQ) fluorescent protein based Wnt reporter. We used a CRISPR-Cas9 approach to insert a TQ fluorescent protein encoding gene into the general Wnt target gene Axin2, thereby establishing a Wnt reporter mouse similar to previously generated Wnt reporter mice but with the mTurquoise2 gene instead of E. coli-ß-galactosidase (LacZ). The use of mTurquoise2 is especially important in organ systems in which cells need to a be alive for further experimentation such as in vitro activation or transplantation studies. We here report successful generation of Axin2-TQ mice and show that cells from these mice faithfully respond to Wnt signals. High Wnt signals were detected in the intestinal crypts, a classical Wnt signalling site in vivo, and by flow cytometry in the thymus. These mice are an improved tool to further elucidate the role of Wnt signalling in vivo.


Subject(s)
Axin Protein/metabolism , Genes, Reporter , Green Fluorescent Proteins/genetics , Wnt Signaling Pathway , Animals , Axin Protein/genetics , CRISPR-Cas Systems , Gene Targeting/methods , Green Fluorescent Proteins/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism
4.
Stem Cell Reports ; 18(9): 1793-1810, 2023 09 12.
Article in English | MEDLINE | ID: mdl-37541258

ABSTRACT

CRB1 gene mutations can cause early- or late-onset retinitis pigmentosa, Leber congenital amaurosis, or maculopathy. Recapitulating human CRB1 phenotypes in animal models has proven challenging, necessitating the development of alternatives. We generated human induced pluripotent stem cell (iPSC)-derived retinal organoids of patients with retinitis pigmentosa caused by biallelic CRB1 mutations and evaluated them against autologous gene-corrected hiPSCs and hiPSCs from healthy individuals. Patient organoids show decreased levels of CRB1 and NOTCH1 expression at the retinal outer limiting membrane. Proximity ligation assays show that human CRB1 and NOTCH1 can interact via their extracellular domains. CRB1 patient organoids feature increased levels of WDFY1+ vesicles, fewer RAB11A+ recycling endosomes, decreased VPS35 retromer complex components, and more degradative endolysosomal compartments relative to isogenic control organoids. Taken together, our data demonstrate that patient-derived retinal organoids enable modeling of retinal degeneration and highlight the importance of CRB1 in early endosome maturation receptor recycling in the retina.


Subject(s)
Induced Pluripotent Stem Cells , Retinal Degeneration , Retinitis Pigmentosa , Animals , Humans , Induced Pluripotent Stem Cells/metabolism , Retina/metabolism , Retinal Degeneration/genetics , Retinitis Pigmentosa/genetics , Mutation , Organoids/metabolism , Eye Proteins/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism
6.
Stem Cell Res ; 53: 102374, 2021 05.
Article in English | MEDLINE | ID: mdl-34088003

ABSTRACT

Combined Oxidative Phosphorylation Deficiency 8 (COXPD8) is an autosomal recessive disorder causing lethal childhood-onset hypertrophic cardiomyopathy. Homozygous or compound heterozygous mutations in the nuclear-encoded mitochondrial alanyl-tRNA synthetase 2 (AARS2) gene underly the pathology. We generated induced pluripotent stem cells (hiPSCs) from two patients carrying the heterozygous compound c.1774 C>T, c.2188 G>A and c.2872 C>T AARS2 mutations, as well as a related healthy control carrying the c.2872 C>T AARS2 mutation. All hiPSC-lines expressed pluripotency markers, maintained a normal karyotype, and differentiated towards the three germ layer derivatives in vitro. These lines can be used to model COXPD8 or mitochondrial dysfunction.


Subject(s)
Cardiomyopathy, Hypertrophic , Induced Pluripotent Stem Cells , Mitochondrial Diseases , Child , Heterozygote , Homozygote , Humans , Mutation
7.
Nat Commun ; 12(1): 6469, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34753942

ABSTRACT

Subunit switches in the BAF chromatin remodeler are essential during development. ARID1B and its paralog ARID1A encode for mutually exclusive BAF subunits. De novo ARID1B haploinsufficient mutations cause neurodevelopmental disorders, including Coffin-Siris syndrome, which is characterized by neurological and craniofacial features. Here, we leveraged ARID1B+/- Coffin-Siris patient-derived iPSCs and modeled cranial neural crest cell (CNCC) formation. We discovered that ARID1B is active only during the first stage of this process, coinciding with neuroectoderm specification, where it is part of a lineage-specific BAF configuration (ARID1B-BAF). ARID1B-BAF regulates exit from pluripotency and lineage commitment by attenuating thousands of enhancers and genes of the NANOG and SOX2 networks. In iPSCs, these enhancers are maintained active by ARID1A-containing BAF. At the onset of differentiation, cells transition from ARID1A- to ARID1B-BAF, eliciting attenuation of the NANOG/SOX2 networks and triggering pluripotency exit. Coffin-Siris patient cells fail to perform the ARID1A/ARID1B switch, and maintain ARID1A-BAF at the pluripotency enhancers throughout all stages of CNCC formation. This leads to persistent NANOG/SOX2 activity which impairs CNCC formation. Despite showing the typical neural crest signature (TFAP2A/SOX9-positive), ARID1B-haploinsufficient CNCCs are also aberrantly NANOG-positive. These findings suggest a connection between ARID1B mutations, neuroectoderm specification and a pathogenic mechanism for Coffin-Siris syndrome.


Subject(s)
Chromatin/metabolism , DNA-Binding Proteins/metabolism , Nanog Homeobox Protein/metabolism , Neural Crest/metabolism , Transcription Factors/metabolism , Blotting, Western , DNA-Binding Proteins/genetics , Flow Cytometry , HEK293 Cells , Humans , Mutation/genetics , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Transcription Factors/genetics
8.
Bio Protoc ; 10(23): e3845, 2020 Dec 05.
Article in English | MEDLINE | ID: mdl-33659493

ABSTRACT

Natural killer (NK) cells are innate immune cells, characterized by their cytotoxic capacity, and chemokine and cytokine secretion upon activation. Human NK cells are identified by CD56 expression. Circulating NK cells can be further subdivided into the CD56bright (~10%) and CD56dim NK cell subsets (~90%). NK cell-like cells can also be derived from human induced pluripotent stem cells (iPSC). To study the chemokine and cytokine secretion profile of the distinct heterogenous NK cell subsets, intracellular flow cytometry staining can be performed. However, this assay is challenging when the starting material is limited. Alternatively, NK cell subsets can be enriched, sorted, stimulated, and functionally profiled by measuring secreted effector molecules in the supernatant by Luminex. Here, we provide a rapid and straightforward protocol for the isolation and stimulation of primary NK cells or iPSC-derived NK cell-like cells, and subsequent detection of secreted cytokines and chemokines, which is also applicable for a low number of cells.

9.
Stem Cell Reports ; 14(2): 300-311, 2020 02 11.
Article in English | MEDLINE | ID: mdl-31956083

ABSTRACT

RAG2 severe combined immune deficiency (RAG2-SCID) is a lethal disorder caused by the absence of functional T and B cells due to a differentiation block. Here, we generated induced pluripotent stem cells (iPSCs) from a RAG2-SCID patient to study the nature of the T cell developmental blockade. We observed a strongly reduced capacity to differentiate at every investigated stage of T cell development, from early CD7-CD5- to CD4+CD8+. The impaired differentiation was accompanied by an increase in CD7-CD56+CD33+ natural killer (NK) cell-like cells. T cell receptor D rearrangements were completely absent in RAG2SCID cells, whereas the rare T cell receptor B rearrangements were likely the result of illegitimate rearrangements. Repair of RAG2 restored the capacity to induce T cell receptor rearrangements, normalized T cell development, and corrected the NK cell-like phenotype. In conclusion, we succeeded in generating an iPSC-based RAG2-SCID model, which enabled the identification of previously unrecognized disorder-related T cell developmental roadblocks.


Subject(s)
DNA-Binding Proteins/metabolism , Induced Pluripotent Stem Cells/metabolism , Models, Biological , Severe Combined Immunodeficiency/immunology , Severe Combined Immunodeficiency/pathology , T-Lymphocytes/immunology , Animals , Antigens, CD/metabolism , Cell Differentiation , Cell Lineage , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Hematopoiesis , Humans , Killer Cells, Natural/immunology , Mice, SCID
10.
Sci Rep ; 10(1): 5499, 2020 03 26.
Article in English | MEDLINE | ID: mdl-32218519

ABSTRACT

Research on acute and chronic lung diseases would greatly benefit from reproducible availability of alveolar epithelial cells (AEC). Primary alveolar epithelial cells can be derived from human lung tissue but the quality of these cells is highly donor dependent. Here, we demonstrated that culture of EpCAM+ cells derived from human induced pluripotent stem cells (hiPSC) at the physiological air-liquid interface (ALI) resulted in type 2 AEC-like cells (iAEC2) with alveolar characteristics. iAEC2 cells expressed native AEC2 markers (surfactant proteins and LPCAT-1) and contained lamellar bodies. ALI-iAEC2 were used to study alveolar repair over a period of 2 weeks following mechanical wounding of the cultures and the responses were compared with those obtained using primary AEC2 (pAEC2) isolated from resected lung tissue. Addition of the Wnt/ß-catenin activator CHIR99021 reduced wound closure in the iAEC2 cultures but not pAEC2 cultures. This was accompanied by decreased surfactant protein expression and accumulation of podoplanin-positive cells at the wound edge. These results demonstrated the feasibility of studying alveolar repair using hiPSC-AEC2 cultured at the ALI and indicated that this model can be used in the future to study modulation of alveolar repair by (pharmaceutical) compounds.


Subject(s)
Alveolar Epithelial Cells/physiology , Induced Pluripotent Stem Cells/physiology , Models, Biological , Alveolar Epithelial Cells/cytology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Cells, Cultured , Humans , In Vitro Techniques , Induced Pluripotent Stem Cells/cytology , Pulmonary Alveoli/injuries , Pulmonary Alveoli/physiology , Pulmonary Alveoli/physiopathology , Regeneration/physiology , Wound Healing/physiology
11.
Stem Cell Reports ; 12(5): 906-919, 2019 05 14.
Article in English | MEDLINE | ID: mdl-30956116

ABSTRACT

Human retinal organoids from induced pluripotent stem cells (hiPSCs) can be used to confirm the localization of proteins in retinal cell types and to test transduction and expression patterns of gene therapy vectors. Here, we compared the onset of CRB protein expression in human fetal retina with human iPSC-derived retinal organoids. We show that CRB2 protein precedes the expression of CRB1 in the developing human retina. Our data suggest the presence of CRB1 and CRB2 in human photoreceptors and Müller glial cells. Thus the fetal CRB complex formation is replicated in hiPSC-derived retina. CRB1 patient iPSC retinal organoids showed disruptions at the outer limiting membrane as found in Crb1 mutant mice. Furthermore, AAV serotype 5 (AAV5) is potent in infecting human Müller glial cells and photoreceptors in hiPSC-derived retinas and retinal explants. Our data suggest that human photoreceptors can be efficiently transduced by AAVs in the presence of photoreceptor segments.


Subject(s)
Carrier Proteins/metabolism , Ependymoglial Cells/metabolism , Eye Proteins/metabolism , Induced Pluripotent Stem Cells/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Organoids/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Retina/metabolism , Adult , Carrier Proteins/genetics , Cells, Cultured , Dependovirus/genetics , Ependymoglial Cells/cytology , Ependymoglial Cells/ultrastructure , Eye Proteins/genetics , Female , Fetus , Humans , Immunohistochemistry , Induced Pluripotent Stem Cells/cytology , Membrane Proteins/genetics , Microscopy, Immunoelectron , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Nerve Tissue Proteins/genetics , Organoids/cytology , Photoreceptor Cells, Vertebrate/ultrastructure , Pregnancy , Retina/cytology , Retina/embryology
12.
Methods Mol Biol ; 1715: 261-273, 2018.
Article in English | MEDLINE | ID: mdl-29188520

ABSTRACT

In vitro retinal organoid modeling from human pluripotent stem cells is becoming more common place in many ophthalmic laboratories worldwide. These organoids mimic human retinogenesis through formation of organized layered retinal structures that display markers for typical retinal cell types. Pivotally these humanized retinal models provide a stepping stone to the clinic as therapeutic tools and are expected to provide a promising alternative to current animal models. Thus pluripotent stem cell based healthy as well as diseased human retinal organoids are attractive for use in drug potency assays and gene augmentation therapeutics. Here we outline an established protocol for generation of these retinal organoids and how they can be used in conjunction with adeno-associated virus vectors for transgene expression assays.


Subject(s)
Dependovirus/genetics , Genetic Vectors , Induced Pluripotent Stem Cells/metabolism , Organoids/metabolism , Retina/metabolism , Transgenes/physiology , Humans , Induced Pluripotent Stem Cells/cytology , Organoids/growth & development , Retina/growth & development
13.
Sci Rep ; 8(1): 386, 2018 01 10.
Article in English | MEDLINE | ID: mdl-29321583

ABSTRACT

The transcription factor Sox2 controls the fate of pluripotent stem cells and neural stem cells. This gatekeeper function requires well-regulated Sox2 levels. We postulated that Sox2 regulation is partially controlled by the Sox2 overlapping long non-coding RNA (lncRNA) gene Sox2ot. Here we show that the RNA levels of Sox2ot and Sox2 are inversely correlated during neural differentiation of mouse embryonic stem cells (ESCs). Through allele-specific enhanced transcription of Sox2ot in mouse Sox2eGFP knockin ESCs we demonstrate that increased Sox2ot transcriptional activity reduces Sox2 RNA levels in an allele-specific manner. Enhanced Sox2ot transcription, yielding lower Sox2 RNA levels, correlates with a decreased chromatin interaction of the upstream regulatory sequence of Sox2 and the ESC-specific Sox2 super enhancer. Our study indicates that, in addition to previously reported in trans mechanisms, Sox2ot can regulate Sox2 by an allele-specific mechanism, in particular during development.


Subject(s)
Mouse Embryonic Stem Cells/cytology , Neurogenesis , RNA, Long Noncoding/genetics , SOXB1 Transcription Factors/genetics , Alleles , Animals , Cell Differentiation , Gene Expression Regulation, Developmental , Gene Knock-In Techniques , Mice , Transcription, Genetic
14.
J Invest Dermatol ; 138(4): 826-835, 2018 04.
Article in English | MEDLINE | ID: mdl-29179949

ABSTRACT

Systemic sclerosis is an autoimmune disease characterized by fibrosis of skin and multiple organs of which the pathogenesis is poorly understood. We studied differentially expressed coding and non-coding genes in relation to systemic sclerosis pathogenesis with a specific focus on antisense non-coding RNAs. Skin biopsy-derived RNAs from 14 early systemic sclerosis patients and six healthy individuals were sequenced with ion-torrent and analyzed using DEseq2. Overall, 4,901 genes with a fold change >1.5 and a false discovery rate <5% were detected in patients versus controls. Upregulated genes clustered in immunologic, cell adhesion, and keratin-related processes. Interestingly, 676 deregulated non-coding genes were detected, 257 of which were classified as antisense genes. Sense genes expressed opposite of these antisense genes were also deregulated in 42% of the observed sense-antisense gene pairs. The majority of the antisense genes had a similar effect sizes in an independent North American dataset with three genes (CTBP1-AS2, OTUD6B-AS1, and AGAP2-AS1) exceeding the study-wide Bonferroni-corrected P-value (PBonf < 0.0023, Pcombined = 1.1 × 10-9, 1.4 × 10-8, 1.7 × 10-6, respectively). In this study, we highlight that together with coding genes, (antisense) long non-coding RNAs are deregulated in skin tissue of systemic sclerosis patients suggesting a novel class of genes involved in pathogenesis of systemic sclerosis.


Subject(s)
RNA, Long Noncoding/genetics , Scleroderma, Systemic/genetics , Skin/metabolism , Up-Regulation , Cells, Cultured , Humans , RNA, Long Noncoding/biosynthesis , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/pathology , Skin/pathology , Transcription Factors , Transcriptional Activation
15.
Nat Commun ; 8(1): 657, 2017 09 22.
Article in English | MEDLINE | ID: mdl-28939824

ABSTRACT

Precise genome editing involves homologous recombination between donor DNA and chromosomal sequences subjected to double-stranded DNA breaks made by programmable nucleases. Ideally, genome editing should be efficient, specific, and accurate. However, besides constituting potential translocation-initiating lesions, double-stranded DNA breaks (targeted or otherwise) are mostly repaired through unpredictable and mutagenic non-homologous recombination processes. Here, we report that the coordinated formation of paired single-stranded DNA breaks, or nicks, at donor plasmids and chromosomal target sites by RNA-guided nucleases based on CRISPR-Cas9 components, triggers seamless homology-directed gene targeting of large genetic payloads in human cells, including pluripotent stem cells. Importantly, in addition to significantly reducing the mutagenicity of the genome modification procedure, this in trans paired nicking strategy achieves multiplexed, single-step, gene targeting, and yields higher frequencies of accurately edited cells when compared to the standard double-stranded DNA break-dependent approach.CRISPR-Cas9-based gene editing involves double-strand breaks at target sequences, which are often repaired by mutagenic non-homologous end-joining. Here the authors use Cas9 nickases to generate coordinated single-strand breaks in donor and target DNA for precise homology-directed gene editing.


Subject(s)
DNA/genetics , Gene Editing , CRISPR-Cas Systems , Cell Line , DNA/metabolism , DNA Breaks, Double-Stranded , DNA Breaks, Single-Stranded , DNA End-Joining Repair , Genome, Human , Humans , RNA, Guide, Kinetoplastida/genetics , RNA, Guide, Kinetoplastida/metabolism
16.
Hum Gene Ther ; 25(10): 866-74, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25141889

ABSTRACT

Hematopoietic stem cell transplantations have become a very successful therapeutic approach to treat otherwise life-threatening blood disorders. It is thought that stem cell transplantation may also become a feasible treatment option for many non-blood-related diseases. So far, however, the limited availability of human leukocyte antigen-matched donors has hindered development of some cell replacement therapies. The Nobel-prize rewarded finding that pluripotency can be induced in somatic cells via expression of a few transcription factors has led to a revolution in stem cell biology. The possibility to change the fate of somatic cells by expressing key transcription factors has been used not only to generate pluripotent stem cells, but also for directly converting somatic cells into fully differentiated cells of another lineage or more committed progenitor cells. These approaches offer the prospect of generating cell types with a specific genotype de novo, which would circumvent the problems associated with allogeneic cell transplantations. This technology has generated a plethora of new disease-specific research efforts, from studying disease pathogenesis to therapeutic interventions. Here we will discuss the opportunities in this booming field of cell biology and summarize how the scientists in the Netherlands have joined efforts in one area to exploit the new technology.


Subject(s)
Cell- and Tissue-Based Therapy , Cellular Reprogramming , Animals , Cell Differentiation , Cell- and Tissue-Based Therapy/adverse effects , Cell- and Tissue-Based Therapy/methods , Cell- and Tissue-Based Therapy/trends , Humans , Netherlands , Stem Cell Research , Stem Cell Transplantation/adverse effects , Stem Cell Transplantation/methods , Stem Cell Transplantation/trends , Stem Cells/cytology , Stem Cells/metabolism
17.
Ann N Y Acad Sci ; 1266: 179-88, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22901269

ABSTRACT

Many life-threatening hematological diseases are now treated by bone marrow transplantations, i.e., infusion of hematopoietic stem cells (HSCs). HSC transplantations are a valid option for the treatment of a variety of metabolic disorders, and even for solid tumors and some refractory severe autoimmune diseases. Unfortunately, the frequency and outcome of HSC transplantations are limited by a shortage of suitable donors. Induced pluripotent stem cells (iPSCs)--somatic cells that have acquired pluripotent stem cell characteristics by the ectopic expression of pluripotency-inducing factors--have been proposed as an alternative source of HSCs. Possible applications include cells of autologous, of autologous and genetically modified, or of allogeneic origin. Here, we provide a perspective on the distinct opportunities of iPSCs and discuss the challenges that lie ahead.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/transplantation , Cell Differentiation , Hematologic Diseases/therapy , Hematologic Neoplasms/therapy , Hematopoiesis , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Safety , Transplantation, Autologous , Transplantation, Homologous
18.
Ned Tijdschr Geneeskd ; 155(47): A3565, 2011.
Article in Dutch | MEDLINE | ID: mdl-22129808

ABSTRACT

UNLABELLED: Stem cells are being used more frequently for research and experimental therapy, but as yet the clinical applications of stem cells are limited. Pluripotent stem cells, with embryonic stem cells as the most well know example, can differentiate into each cell type; in contrast, tissue specific stem cells can only form one or more cell types within one type of tissue. It has been possible for some time to reprogram different types of somatic cells into pluripotent stem cells. Such stem cells are termed induced pluripotent stem cells (iPS cells). iPS cells can also be created from cells of patients with genetic conditions. Research into mechanisms of pathology and new medicines can be carried out with these against a specific genetic background. Clinical application of such iPS cells is not to be expected in the short term. Facilities are being established in different Dutch academic centres to create iPS cells for scientific research. CONFLICT OF INTEREST: none declared.


Subject(s)
Cell Culture Techniques , Cell Transplantation , Embryonic Stem Cells/cytology , Pluripotent Stem Cells/cytology , Regenerative Medicine , Cell Differentiation , Embryonic Stem Cells/physiology , Genetic Engineering , Humans , Pluripotent Stem Cells/physiology
SELECTION OF CITATIONS
SEARCH DETAIL