Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 12 de 12
1.
Neuron ; 112(7): 1117-1132.e9, 2024 Apr 03.
Article En | MEDLINE | ID: mdl-38266647

Mitochondria account for essential cellular pathways, from ATP production to nucleotide metabolism, and their deficits lead to neurological disorders and contribute to the onset of age-related diseases. Direct neuronal reprogramming aims at replacing neurons lost in such conditions, but very little is known about the impact of mitochondrial dysfunction on the direct reprogramming of human cells. Here, we explore the effects of mitochondrial dysfunction on the neuronal reprogramming of induced pluripotent stem cell (iPSC)-derived astrocytes carrying mutations in the NDUFS4 gene, important for Complex I and associated with Leigh syndrome. This led to the identification of the unfolded protein response as a major hurdle in the direct neuronal conversion of not only astrocytes and fibroblasts from patients but also control human astrocytes and fibroblasts. Its transient inhibition potently improves reprogramming by influencing the mitochondria-endoplasmic-reticulum-stress-mediated pathways. Taken together, disease modeling using patient cells unraveled novel general hurdles and ways to overcome these in human astrocyte-to-neuron reprogramming.


Induced Pluripotent Stem Cells , Mitochondrial Diseases , Humans , Neurons/physiology , Mitochondria/metabolism , Induced Pluripotent Stem Cells/metabolism , Unfolded Protein Response , Astrocytes/metabolism , Mitochondrial Diseases/metabolism , Cellular Reprogramming , Electron Transport Complex I/genetics , Electron Transport Complex I/metabolism
2.
Stem Cell Res ; 72: 103197, 2023 10.
Article En | MEDLINE | ID: mdl-37689041

Mitochondrial membrane Protein-Associated Neurodegeneration (MPAN) is a lethal neurodegenerative disorder caused by mutations in the human gene C19orf12. The molecular mechanisms underlying the disorder are still unclear, and no established therapy is available. Here, we describe the generation and characterization of two human induced pluripotent stem cell (iPSC) lines derived from skin fibroblasts of two MPAN patients carrying homozygous recessive mutations in C19orf12. These iPSC lines represent a useful resource for future investigations on the pathology of MPAN, as well as for the development of successful treatments.


Induced Pluripotent Stem Cells , Humans , Mitochondrial Proteins/genetics , Mutation/genetics , Membrane Proteins/genetics , Fibroblasts
3.
Nucleic Acids Res ; 51(3): 1297-1316, 2023 02 22.
Article En | MEDLINE | ID: mdl-36651277

The RNA-binding protein PURA has been implicated in the rare, monogenetic, neurodevelopmental disorder PURA Syndrome. PURA binds both DNA and RNA and has been associated with various cellular functions. Only little is known about its main cellular roles and the molecular pathways affected upon PURA depletion. Here, we show that PURA is predominantly located in the cytoplasm, where it binds to thousands of mRNAs. Many of these transcripts change abundance in response to PURA depletion. The encoded proteins suggest a role for PURA in immune responses, mitochondrial function, autophagy and processing (P)-body activity. Intriguingly, reduced PURA levels decrease the expression of the integral P-body components LSM14A and DDX6 and strongly affect P-body formation in human cells. Furthermore, PURA knockdown results in stabilization of P-body-enriched transcripts, whereas other mRNAs are not affected. Hence, reduced PURA levels, as reported in patients with PURA Syndrome, influence the formation and composition of this phase-separated RNA processing machinery. Our study proposes PURA Syndrome as a new model to study the tight connection between P-body-associated RNA regulation and neurodevelopmental disorders.


RNA-Binding Proteins , Transcription Factors , Humans , DNA-Binding Proteins/genetics , Epilepsy , Processing Bodies , RNA, Messenger/metabolism , Transcription Factors/metabolism
4.
Blood ; 141(6): 645-658, 2023 02 09.
Article En | MEDLINE | ID: mdl-36223592

The mechanisms of coordinated changes in proteome composition and their relevance for the differentiation of neutrophil granulocytes are not well studied. Here, we discover 2 novel human genetic defects in signal recognition particle receptor alpha (SRPRA) and SRP19, constituents of the mammalian cotranslational targeting machinery, and characterize their roles in neutrophil granulocyte differentiation. We systematically study the proteome of neutrophil granulocytes from patients with variants in the SRP genes, HAX1, and ELANE, and identify global as well as specific proteome aberrations. Using in vitro differentiation of human induced pluripotent stem cells and in vivo zebrafish models, we study the effects of SRP deficiency on neutrophil granulocyte development. In a heterologous cell-based inducible protein expression system, we validate the effects conferred by SRP dysfunction for selected proteins that we identified in our proteome screen. Thus, SRP-dependent protein processing, intracellular trafficking, and homeostasis are critically important for the differentiation of neutrophil granulocytes.


Induced Pluripotent Stem Cells , Proteome , Animals , Humans , Zebrafish , Human Genetics , Mammals , Adaptor Proteins, Signal Transducing
5.
Stem Cell Res ; 61: 102773, 2022 05.
Article En | MEDLINE | ID: mdl-35397396

Phosphopantothenoylcysteine synthetase (PPCS) catalyzes the second step of the de novo coenzyme A (CoA) synthesis starting from pantothenate. Mutations in PPCS cause autosomal-recessive dilated cardiomyopathy, often fatal, without apparent neurodegeneration, whereas pathogenic variants in PANK2 and COASY, two other genes involved in the CoA synthesis, cause Neurodegeneration with Brain Iron Accumulation (NBIA). PPCS-deficiency is a relatively new disease with unclear pathogenesis and no targeted therapy. Here, we report the generation of induced pluripotent stem cells from fibroblasts of two PPCS-deficient patients. These cellular models could represent a platform for pathophysiological studies and testing of therapeutic compounds for PPCS-deficiency.


Cardiomyopathy, Dilated , Induced Pluripotent Stem Cells , Coenzyme A , Fibroblasts , Humans , Mutation/genetics
6.
Sci Rep ; 12(1): 3100, 2022 03 08.
Article En | MEDLINE | ID: mdl-35260583

The northern white rhinoceros (NWR) is probably the earth's most endangered mammal. To rescue the functionally extinct species, we aim to employ induced pluripotent stem cells (iPSCs) to generate gametes and subsequently embryos in vitro. To elucidate the regulation of pluripotency and differentiation of NWR PSCs, we generated iPSCs from a deceased NWR female using episomal reprogramming, and observed surprising similarities to human PSCs. NWR iPSCs exhibit a broad differentiation potency into the three germ layers and trophoblast, and acquire a naïve-like state of pluripotency, which is pivotal to differentiate PSCs into primordial germ cells (PGCs). Naïve culturing conditions induced a similar expression profile of pluripotency related genes in NWR iPSCs and human ESCs. Furthermore, naïve-like NWR iPSCs displayed increased expression of naïve and PGC marker genes, and a higher integration propensity into developing mouse embryos. As the conversion process was aided by ectopic BCL2 expression, and we observed integration of reprogramming factors, the NWR iPSCs presented here are unsuitable for gamete production. However, the gained insights into the developmental potential of both primed and naïve-like NWR iPSCs are fundamental for in future PGC-specification in order to rescue the species from extinction using cryopreserved somatic cells.


Induced Pluripotent Stem Cells , Animals , Cell Differentiation/genetics , Female , Germ Cells/metabolism , Germ Layers , Mice , Perissodactyla/genetics
7.
CRISPR J ; 4(2): 178-190, 2021 04.
Article En | MEDLINE | ID: mdl-33876960

STAT3-hyper IgE syndrome (STAT3-HIES) is a primary immunodeficiency presenting with destructive lung disease along with other symptoms. CRISPR-Cas9-mediated adenine base editors (ABEs) have the potential to correct one of the most common STAT3-HIES causing heterozygous STAT3 mutations (c.1144C>T/p.R382W). As a proof-of-concept, we successfully applied ABEs to correct STAT3 p.R382W in patient fibroblasts and induced pluripotent stem cells (iPSCs). Treated primary STAT3-HIES patient fibroblasts showed a correction efficiency of 29% ± 7% without detectable off-target effects evaluated through whole-genome and high-throughput sequencing. Compared with untreated patient fibroblasts, corrected single-cell clones showed functional rescue of STAT3 signaling with significantly increased STAT3 DNA-binding activity and target gene expression of CCL2 and SOCS3. Patient-derived iPSCs were corrected with an efficiency of 30% ± 6% and differentiated to alveolar organoids showing preserved plasticity in treated cells. In conclusion, our results are supportive for ABE-based gene correction as a potential causative treatment of STAT3-HIES.


Gene Editing/methods , Job Syndrome/genetics , Job Syndrome/therapy , STAT3 Transcription Factor/genetics , Adenine , CRISPR-Cas Systems , Cell Differentiation , Clustered Regularly Interspaced Short Palindromic Repeats , Fibroblasts , Heterozygote , High-Throughput Nucleotide Sequencing , Humans , Immunoglobulin E/genetics , Induced Pluripotent Stem Cells , Mutation , Whole Genome Sequencing
8.
BMC Biol ; 18(1): 42, 2020 04 22.
Article En | MEDLINE | ID: mdl-32321486

BACKGROUND: Many long noncoding RNAs (lncRNAs) have been implicated in general and cell type-specific molecular regulation. Here, we asked what underlies the fundamental basis for the seemingly random appearance of nuclear lncRNA condensates in cells, and we sought compounds that can promote the disintegration of lncRNA condensates in vivo. RESULTS: As a basis for comparing lncRNAs and cellular properties among different cell types, we screened lncRNAs in human pluripotent stem cells (hPSCs) that were differentiated to an atlas of cell lineages. We found that paraspeckles, which form by aggregation of the lncRNA NEAT1, are scaled by the size of the nucleus, and that small DNA-binding molecules promote the disintegration of paraspeckles and other lncRNA condensates. Furthermore, we found that paraspeckles regulate the differentiation of hPSCs. CONCLUSIONS: Positive correlation between the size of the nucleus and the number of paraspeckles exist in numerous types of human cells. The tethering and structure of paraspeckles, as well as other lncRNAs, to the genome can be disrupted by small molecules that intercalate in DNA. The structure-function relationship of lncRNAs that regulates stem cell differentiation is likely to be determined by the dynamics of nucleus size and binding site accessibility.


Cell Differentiation , Pluripotent Stem Cells/physiology , RNA, Long Noncoding/metabolism , Cell Nucleus/genetics , Cell Nucleus/physiology , DNA/genetics , DNA/physiology , Humans
9.
Mol Cell ; 74(5): 951-965.e13, 2019 06 06.
Article En | MEDLINE | ID: mdl-31047794

RNA-binding proteins (RBPs) and long non-coding RNAs (lncRNAs) are key regulators of gene expression, but their joint functions in coordinating cell fate decisions are poorly understood. Here we show that the expression and activity of the RBP TDP-43 and the long isoform of the lncRNA Neat1, the scaffold of the nuclear compartment "paraspeckles," are reciprocal in pluripotent and differentiated cells because of their cross-regulation. In pluripotent cells, TDP-43 represses the formation of paraspeckles by enhancing the polyadenylated short isoform of Neat1. TDP-43 also promotes pluripotency by regulating alternative polyadenylation of transcripts encoding pluripotency factors, including Sox2, which partially protects its 3' UTR from miR-21-mediated degradation. Conversely, paraspeckles sequester TDP-43 and other RBPs from mRNAs and promote exit from pluripotency and embryonic patterning in the mouse. We demonstrate that cross-regulation between TDP-43 and Neat1 is essential for their efficient regulation of a broad network of genes and, therefore, of pluripotency and differentiation.


Cell Differentiation/genetics , DNA-Binding Proteins/genetics , Mouse Embryonic Stem Cells/metabolism , RNA, Long Noncoding/genetics , Animals , Cell Nucleus/genetics , Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Humans , Mice , MicroRNAs/genetics , Pluripotent Stem Cells/metabolism , Polyadenylation/genetics , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
10.
Stem Cell Reports ; 12(5): 861-868, 2019 05 14.
Article En | MEDLINE | ID: mdl-31006630

The neural crest (NC) gives rise to a multitude of fetal tissues, and its misregulation is implicated in congenital malformations. Here, we investigated molecular mechanisms pertaining to NC-related symptoms in Bohring-Opitz syndrome (BOS), a developmental disorder linked to mutations in the Polycomb group factor Additional sex combs-like 1 (ASXL1). Genetically edited human pluripotent stem cell lines that were differentiated to NC progenitors and then xenotransplanted into chicken embryos demonstrated an impairment of NC delamination and emigration. Molecular analysis showed that ASXL1 mutations correlated with reduced activation of the transcription factor ZIC1 and the NC gene regulatory network. These findings were supported by differentiation experiments using BOS patient-derived induced pluripotent stem cell lines. Expression of truncated ASXL1 isoforms (amino acids 1-900) recapitulated the NC phenotypes in vitro and in ovo, raising the possibility that truncated ASXL1 variants contribute to BOS pathology. Collectively, we expand the understanding of truncated ASXL1 in BOS and in the human NC.


Cell Differentiation/genetics , Craniosynostoses/genetics , Gene Expression Profiling/methods , Intellectual Disability/genetics , Mutation , Neural Crest/metabolism , Pluripotent Stem Cells/metabolism , Repressor Proteins/genetics , Animals , Cell Line , Cells, Cultured , Chick Embryo , Craniosynostoses/metabolism , Craniosynostoses/pathology , Gene Regulatory Networks , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Intellectual Disability/metabolism , Intellectual Disability/pathology , Neural Crest/cytology , Pluripotent Stem Cells/cytology , Repressor Proteins/metabolism , Transplantation, Heterologous
11.
Nat Med ; 25(4): 561-568, 2019 04.
Article En | MEDLINE | ID: mdl-30858616

Malformations of the human cortex represent a major cause of disability1. Mouse models with mutations in known causal genes only partially recapitulate the phenotypes and are therefore not unlimitedly suited for understanding the molecular and cellular mechanisms responsible for these conditions2. Here we study periventricular heterotopia (PH) by analyzing cerebral organoids derived from induced pluripotent stem cells (iPSCs) of patients with mutations in the cadherin receptor-ligand pair DCHS1 and FAT4 or from isogenic knockout (KO) lines1,3. Our results show that human cerebral organoids reproduce the cortical heterotopia associated with PH. Mutations in DCHS1 and FAT4 or knockdown of their expression causes changes in the morphology of neural progenitor cells and result in defective neuronal migration dynamics only in a subset of neurons. Single-cell RNA-sequencing (scRNA-seq) data reveal a subpopulation of mutant neurons with dysregulated genes involved in axon guidance, neuronal migration and patterning. We suggest that defective neural progenitor cell (NPC) morphology and an altered navigation system in a subset of neurons underlie this form of PH.


Cell Movement , Cerebrum/pathology , Neurons/pathology , Organoids/pathology , Periventricular Nodular Heterotopia/pathology , Cadherin Related Proteins , Cadherins/genetics , Cell Line , Humans , Infant, Newborn , Mutation/genetics , Sequence Analysis, RNA , Single-Cell Analysis , Time-Lapse Imaging , Tumor Suppressor Proteins/genetics
12.
Glia ; 66(2): 413-427, 2018 02.
Article En | MEDLINE | ID: mdl-29119608

Astrocytes, the most abundant cells in the mammalian brain, perform key functions and are involved in several neurodegenerative diseases. The human immunodeficiency virus (HIV) can persist in astrocytes, contributing to the HIV burden and neurological dysfunctions in infected individuals. While a comprehensive approach to HIV cure must include the targeting of HIV-1 in astrocytes, dedicated tools for this purpose are still lacking. Here we report a novel Adeno-associated virus-based vector (AAV9P1) with a synthetic surface peptide for transduction of astrocytes. Analysis of AAV9P1 transduction efficiencies with single brain cell populations, including primary human brain cells, as well as human brain organoids demonstrated that AAV9P1 targeted terminally differentiated human astrocytes much more efficiently than neurons. We then investigated whether AAV9P1 can be used to deliver HIV-inhibitory genes to astrocytes. To this end we generated AAV9P1 vectors containing genes for HIV-1 proviral editing by CRISPR/Cas9. Latently HIV-1 infected astrocytes transduced with these vectors showed significantly diminished reactivation of proviruses, compared with untransduced cultures. Sequence analysis identified mutations/deletions in key HIV-1 transcriptional control regions. We conclude that AAV9P1 is a promising tool for gene delivery to astrocytes and may facilitate inactivation/destruction of persisting HIV-1 proviruses in astrocyte reservoirs.


Astrocytes/physiology , Clustered Regularly Interspaced Short Palindromic Repeats/physiology , Dependovirus/physiology , Gene Expression Regulation, Viral/physiology , Genetic Vectors/administration & dosage , HIV-1/physiology , Astrocytes/drug effects , Astrocytes/virology , Cell Line, Transformed , Cells, Cultured , Clustered Regularly Interspaced Short Palindromic Repeats/drug effects , Foreskin/cytology , Gene Expression Regulation, Viral/drug effects , HEK293 Cells , HIV-1/drug effects , Humans , Male
...