Logo: to the web site of Uppsala University

uu.sePublications from Uppsala University
Change search
Link to record
Permanent link

Direct link
Schuster, Jens, Assistant ProfessorORCID iD iconorcid.org/0000-0002-4383-9880
Alternative names
Publications (10 of 44) Show all publications
Schuster, J., Tripathi, R., Klar, J. & Dahl, N. (2022). Generation of a human iPSC line (UUIGPi015-A) from a patient with Dravet syndrome and a 2.9 Mb deletion spanning SCN1A on chromosome 2. Stem Cell Research, 60, Article ID 102712.
Open this publication in new window or tab >>Generation of a human iPSC line (UUIGPi015-A) from a patient with Dravet syndrome and a 2.9 Mb deletion spanning SCN1A on chromosome 2
2022 (English)In: Stem Cell Research, ISSN 1873-5061, E-ISSN 1876-7753, Vol. 60, article id 102712Article in journal (Refereed) Published
Abstract [en]

Dravet syndrome is an early onset devastating epilepsy syndrome usually caused by heterozygous mutations in SCN1A. We generated a human iPSC line (UUIGPi015A) from dermal fibroblasts of a patient with Dravet syndrome carrying a deletion on chromosome 2 encompassing SCN1A and 9 flanking genes. Characterization of the iPSC line confirmed expression of pluripotency markers, tri-lineage differentiation capacity and absence of exogenous reprogramming factors. The iPSC line retained the deletion and was genomically stable. The iPSC line UUIGPi015-A provides a useful resource for studies on the pathophysiology of Dravet syndrome and seizures caused by haploinsufficiency of SCN1A and flanking gene products.

Place, publisher, year, edition, pages
ElsevierElsevier BV, 2022
National Category
Cell and Molecular Biology
Identifiers
urn:nbn:se:uu:diva-474704 (URN)10.1016/j.scr.2022.102712 (DOI)000789840000006 ()35203050 (PubMedID)
Funder
Swedish Research Council, 2020-01947The Swedish Brain Foundation, FO2019-0210The Swedish Brain Foundation, FO2020-0171
Available from: 2022-05-24 Created: 2022-05-24 Last updated: 2024-01-15Bibliographically approved
Schuster, J., Klar, J., Khalfallah, A., Laan, L., Hoeber, J., Fatima, A., . . . Dahl, N. (2022). ZEB2 haploinsufficient Mowat-Wilson syndrome induced pluripotent stem cells show disrupted GABAergic transcriptional regulation and function. Frontiers in Molecular Neuroscience, 15, Article ID 988993.
Open this publication in new window or tab >>ZEB2 haploinsufficient Mowat-Wilson syndrome induced pluripotent stem cells show disrupted GABAergic transcriptional regulation and function
Show others...
2022 (English)In: Frontiers in Molecular Neuroscience, ISSN 1662-5099, Vol. 15, article id 988993Article in journal (Refereed) Published
Abstract [en]

Mowat-Wilson syndrome (MWS) is a severe neurodevelopmental disorder caused by heterozygous variants in the gene encoding transcription factor ZEB2. Affected individuals present with structural brain abnormalities, speech delay and epilepsy. In mice, conditional loss of Zeb2 causes hippocampal degeneration, altered migration and differentiation of GABAergic interneurons, a heterogeneous population of mainly inhibitory neurons of importance for maintaining normal excitability. To get insights into GABAergic development and function in MWS we investigated ZEB2 haploinsufficient induced pluripotent stem cells (iPSC) of MWS subjects together with iPSC of healthy donors. Analysis of RNA-sequencing data at two time points of GABAergic development revealed an attenuated interneuronal identity in MWS subject derived iPSC with enrichment of differentially expressed genes required for transcriptional regulation, cell fate transition and forebrain patterning. The ZEB2 haploinsufficient neural stem cells (NSCs) showed downregulation of genes required for ventral telencephalon specification, such as FOXG1, accompanied by an impaired migratory capacity. Further differentiation into GABAergic interneuronal cells uncovered upregulation of transcription factors promoting pallial and excitatory neurons whereas cortical markers were downregulated. The differentially expressed genes formed a neural protein-protein network with extensive connections to well-established epilepsy genes. Analysis of electrophysiological properties in ZEB2 haploinsufficient GABAergic cells revealed overt perturbations manifested as impaired firing of repeated action potentials. Our iPSC model of ZEB2 haploinsufficient GABAergic development thus uncovers a dysregulated gene network leading to immature interneurons with mixed identity and altered electrophysiological properties, suggesting mechanisms contributing to the neuropathogenesis and seizures in MWS.

Place, publisher, year, edition, pages
Frontiers Media S.A., 2022
Keywords
ZEB2, Mowat-Wilson syndrome, FOXG1, epilepsy, neurodevelopmental disease, GABAergic interneurons, transcriptional network, electrophysiology
National Category
Neurosciences
Identifiers
urn:nbn:se:uu:diva-490531 (URN)10.3389/fnmol.2022.988993 (DOI)000889996300001 ()36353360 (PubMedID)
Available from: 2022-12-19 Created: 2022-12-19 Last updated: 2022-12-19Bibliographically approved
Reimegård, J., Tarbier, M., Danielsson, M., Schuster, J., Baskaran, S., Panagiotou, S., . . . Gallant, C. J. (2021). A combined approach for single-cell mRNA and intracellular protein expression analysis. Communications Biology, 4(1), Article ID 624.
Open this publication in new window or tab >>A combined approach for single-cell mRNA and intracellular protein expression analysis
Show others...
2021 (English)In: Communications Biology, E-ISSN 2399-3642, Vol. 4, no 1, article id 624Article in journal (Refereed) Published
Abstract [en]

Combined measurements of mRNA and protein expression in single cells enable in-depth analysis of cellular states. We present SPARC, an approach that combines single-cell RNA-sequencing with proximity extension essays to simultaneously measure global mRNA and 89 intracellular proteins in individual cells. We show that mRNA expression fails to accurately reflect protein abundance at the time of measurement, although the direction of changes is in agreement during neuronal differentiation. Moreover, protein levels of transcription factors better predict their downstream effects than do their corresponding transcripts. Finally, we highlight that protein expression variation is overall lower than mRNA variation, but relative protein variation does not reflect the mRNA level. Our results demonstrate that mRNA and protein measurements in single cells provide different and complementary information regarding cell states. SPARC presents a state-of-the-art co-profiling method that overcomes current limitations in throughput and protein localization, including removing the need for cell fixation.

Place, publisher, year, edition, pages
Springer Nature, 2021
National Category
Cell and Molecular Biology
Identifiers
urn:nbn:se:uu:diva-446884 (URN)10.1038/s42003-021-02142-w (DOI)000658522400002 ()34035432 (PubMedID)
Funder
Swedish Research Council, 2017-05229Swedish Research Council, 2015-02424Knut and Alice Wallenberg FoundationEU, European Research Council, 758397Swedish Research Council, 2019-05320Science for Life Laboratory - a national resource center for high-throughput molecular bioscience
Note

De tre första författarna delar förstaförfattarskapet

Available from: 2021-06-29 Created: 2021-06-29 Last updated: 2024-01-15Bibliographically approved
Vasylovska, S., Schuster, J., Brboric, A., Carlsson, P.-O., Dahl, N. & Lau, J. (2021). Generation of human induced pluripotent stem cell (iPSC) lines (UUMCBi001-A, UUMCBi002-A) from two healthy donors. Stem Cell Research, 50, Article ID 102114.
Open this publication in new window or tab >>Generation of human induced pluripotent stem cell (iPSC) lines (UUMCBi001-A, UUMCBi002-A) from two healthy donors
Show others...
2021 (English)In: Stem Cell Research, ISSN 1873-5061, E-ISSN 1876-7753, Vol. 50, article id 102114Article in journal (Refereed) Published
Abstract [en]

Availability of numerous high-quality iPSC lines is needed to overcome donor-associated variability caused by genetic background effects. We generated two human iPSC lines from dermal fibroblasts of two healthy females using Sendai virus reprogramming. Quality assessment of the iPSC lines confirmed the expression of pluripotency markers, trilineage differentiation capacity and absence of exogenous expression of reprogramming factors. Both iPSC lines were genetically stable with a genotype that matched the fibroblast lines of donors. These iPSC lines add to available reference lines as a resource for disease modeling of polygenic and multifactorial diseases, for evaluation of differentiation protocols and toxicology screening.

Place, publisher, year, edition, pages
ElsevierElsevier BV, 2021
National Category
Cell and Molecular Biology Cell Biology Medical Biotechnology (with a focus on Cell Biology (including Stem Cell Biology), Molecular Biology, Microbiology, Biochemistry or Biopharmacy)
Identifiers
urn:nbn:se:uu:diva-437092 (URN)10.1016/j.scr.2020.102114 (DOI)000614260000006 ()33340796 (PubMedID)
Funder
Swedish Research Council, 2015-02424Swedish Research Council, 2020-01947Swedish Research Council, 2017-01343
Note

Svitlana Vasylovska and Jens Schuster contributed equally to the study.

Available from: 2021-03-08 Created: 2021-03-08 Last updated: 2024-01-15Bibliographically approved
Fatima, A., Hoeber, J., Schuster, J., Koshimizu, E., Gonzalez, C. M., Keren, B., . . . Dahl, N. (2021). Monoallelic and bi-allelic variants in NCDN cause neurodevelopmental delay, intellectual disability, and epilepsy. American Journal of Human Genetics, 108(4), 739-748
Open this publication in new window or tab >>Monoallelic and bi-allelic variants in NCDN cause neurodevelopmental delay, intellectual disability, and epilepsy
Show others...
2021 (English)In: American Journal of Human Genetics, ISSN 0002-9297, E-ISSN 1537-6605, Vol. 108, no 4, p. 739-748Article in journal (Refereed) Published
Abstract [en]

Neurochondrin (NCDN) is a cytoplasmatic neural protein of importance for neural growth, glutamate receptor (mGluR) signaling, and synaptic plasticity. Conditional loss of Ncdn in mice neural tissue causes depressive-like behaviors, impaired spatial learning, and epileptic seizures. We report on NCDN missense variants in six affected individuals with variable degrees of developmental delay, intellectual disability (ID), and seizures. Three siblings were found homozygous for a NCDN missense variant, whereas another three unrelated individuals carried different de novo missense variants in NCDN. We assayed the missense variants for their capability to rescue impaired neurite formation in human neuroblastoma (SH-SY5Y) cells depleted of NCDN. Overexpression of wild-type NCDN rescued the neurite-phenotype in contrast to expression of NCDN containing the variants of affected individuals. Two missense variants, associated with severe neurodevelopmental features and epilepsy, were unable to restore mGluR5-induced ERK phosphorylation. Electrophysiological analysis of SH-SY5Y cells depleted of NCDN exhibited altered membrane potential and impaired action potentials at repolarization, suggesting NCDN to be required for normal biophysical properties. Using available transcriptome data from human fetal cortex, we show that NCDN is highly expressed in maturing excitatory neurons. In combination, our data provide evidence that bi-allelic and de novo variants in NCDN cause a clinically variable form of neurodevelopmental delay and epilepsy, highlighting a critical role for NCDN in human brain development.

Place, publisher, year, edition, pages
Cell PressCELL PRESS, 2021
National Category
Neurosciences
Identifiers
urn:nbn:se:uu:diva-442185 (URN)10.1016/j.ajhg.2021.02.015 (DOI)000636658200015 ()33711248 (PubMedID)
Funder
Swedish Research Council, 2015-02424Swedish Research Council, 2020-01947The Swedish Brain Foundation, FO2019-0210The Swedish Brain Foundation, FO2020-0171Science for Life Laboratory - a national resource center for high-throughput molecular bioscienceM Borgströms stiftelse för ärftlighetsforskning
Available from: 2021-05-17 Created: 2021-05-17 Last updated: 2024-01-15Bibliographically approved
Schuster, J., de Guidi, C., Fatima, A., Sobol, M. & Dahl, N. (2021). Syndromic RNA polymerase II insufficiency: Generation of a human induced pluripotent stem cell line (UUIGPi002A-5) with a heterozygous disruption of POLR2A. Stem Cell Research, 57, Article ID 102577.
Open this publication in new window or tab >>Syndromic RNA polymerase II insufficiency: Generation of a human induced pluripotent stem cell line (UUIGPi002A-5) with a heterozygous disruption of POLR2A
Show others...
2021 (English)In: Stem Cell Research, ISSN 1873-5061, E-ISSN 1876-7753, Vol. 57, article id 102577Article in journal (Refereed) Published
Abstract [en]

Heterozygous variants in POLR2A, encoding the largest subunit of RNA polymerase II, cause severe neurodevelopmental and multisystem abnormalities in humans. Using CRISPR/Cas9 we generated the human iPSC line KICRi002A-5 with a heterozygous truncating 4 bp insertion in exon 5 of the POLR2A gene. Analysis using qRTPCR confirmed reduced POLR2A mRNA in KICRi002A-5 vs. the isogenic WT iPSC line. The edited iPSC line expressed pluripotency markers and exhibited differentiation capacity into the three germ layers. Assessment of genomic integrity revealed a normal karyotype and OFF-target editing was excluded. The iPSC line KICRi002A-5 provides a useful resource to study mechanisms underlying developmental defects caused by RBP1 insufficiency.

Place, publisher, year, edition, pages
ElsevierElsevier BV, 2021
National Category
Cell and Molecular Biology Medical Genetics
Identifiers
urn:nbn:se:uu:diva-458693 (URN)10.1016/j.scr.2021.102577 (DOI)000712140900003 ()34688129 (PubMedID)
Funder
Swedish Research Council, 2020-01947The Swedish Brain Foundation, FO 2020-0171
Available from: 2021-12-01 Created: 2021-12-01 Last updated: 2024-01-15Bibliographically approved
Akram, T., Fatima, A., Klar, J., Hoeber, J., Zakaria, M., Tariq, M., . . . Dahl, N. (2020). Aberrant splicing due to a novel RPS7 variant causes Diamond-Blackfan Anemia associated with spontaneous remission and meningocele. International Journal of Hematology, 112(6), 894-899
Open this publication in new window or tab >>Aberrant splicing due to a novel RPS7 variant causes Diamond-Blackfan Anemia associated with spontaneous remission and meningocele
Show others...
2020 (English)In: International Journal of Hematology, ISSN 0925-5710, E-ISSN 1865-3774, Vol. 112, no 6, p. 894-899Article in journal (Refereed) Published
Abstract [en]

Diamond-Blackfan Anemia (DBA) is a congenital pure red cell aplasia caused by heterozygous variants in ribosomal protein genes. The hematological features associated with DBA are highly variable and non-hematological abnormalities are common. We report herein on an affected mother and her daughter presenting with transfusion-dependent anemia. The mother showed mild physical abnormalities and entered spontaneous remission at age 13 years. Her daughter was born with occipital meningocele. Exome sequencing of DNA from the mother revealed a heterozygous novel splice site variant (NM_001011.4:c.508-3T > G) in the Ribosomal Protein S7 gene (RPS7) inherited by the daughter. Functional analysis of the RPS7 variant expressed from a mini-gene construct revealed that the exon 7 acceptor splice site was replaced by a cryptic splice resulting in a transcript missing 64 bp of exon 7 (p.Val170Serfs*8). Our study confirms a pathogenic effect of a novel RPS7 variant in DBA associated with spontaneous remission in the mother and meningocele in her daughter, thus adding to the genotype-phenotype correlations in DBA.

Keywords
Diamond-Blackfan Anemia (DBA), In vitro splicing, Meningocele, RPS7 gene variant, Spontaneous remission
National Category
Hematology Medical Genetics
Identifiers
urn:nbn:se:uu:diva-428346 (URN)10.1007/s12185-020-02950-6 (DOI)000557744400001 ()32772263 (PubMedID)
Funder
Swedish Research Council, 2015-02424Science for Life Laboratory - a national resource center for high-throughput molecular bioscience
Available from: 2020-12-14 Created: 2020-12-14 Last updated: 2021-01-26Bibliographically approved
Laan, L., Klar, J., Sobol, M., Hoeber, J., Shahsavan, M., Kele, M., . . . Dahl, N. (2020). DNA methylation changes in Down syndrome derived neural iPSCs uncover co-dysregulation of ZNF and HOX3 families of transcription factors. Clinical Epigenetics, 12, Article ID 9.
Open this publication in new window or tab >>DNA methylation changes in Down syndrome derived neural iPSCs uncover co-dysregulation of ZNF and HOX3 families of transcription factors
Show others...
2020 (English)In: Clinical Epigenetics, E-ISSN 1868-7083, Vol. 12, article id 9Article in journal (Refereed) Published
Abstract [en]

Background: Down syndrome (DS) is characterized by neurodevelopmental abnormalities caused by partial or complete trisomy of human chromosome 21 (T21). Analysis of Down syndrome brain specimens has shown global epigenetic and transcriptional changes but their interplay during early neurogenesis remains largely unknown. We differentiated induced pluripotent stem cells (iPSCs) established from two DS patients with complete T21 and matched euploid donors into two distinct neural stages corresponding to early- and mid-gestational ages.

Results: Using the Illumina Infinium 450K array, we assessed the DNA methylation pattern of known CpG regions and promoters across the genome in trisomic neural iPSC derivatives, and we identified a total of 500 stably and differentially methylated CpGs that were annotated to CpG islands of 151 genes. The genes were enriched within the DNA binding category, uncovering 37 factors of importance for transcriptional regulation and chromatin structure. In particular, we observed regional epigenetic changes of the transcription factor genes ZNF69, ZNF700 and ZNF763 as well as the HOXA3, HOXB3 and HOXD3 genes. A similar clustering of differential methylation was found in the CpG islands of the HIST1 genes suggesting effects on chromatin remodeling.

Conclusions: The study shows that early established differential methylation in neural iPSC derivatives with T21 are associated with a set of genes relevant for DS brain development, providing a novel framework for further studies on epigenetic changes and transcriptional dysregulation during T21 neurogenesis.

Keywords
Down syndrome, induced pluripotent stem cells, DNA-methylation, neurogenesis, transcription factors, gene expression
National Category
Medical Genetics
Identifiers
urn:nbn:se:uu:diva-398619 (URN)10.1186/s13148-019-0803-1 (DOI)000512048300002 ()31915063 (PubMedID)
Funder
Swedish Research Council, 2015-02424The Swedish Brain Foundation, FO2018-0100The Swedish Brain Foundation, FO2019-0210Knut and Alice Wallenberg Foundation, Bioinformatic supportAstraZeneca
Available from: 2019-12-08 Created: 2019-12-08 Last updated: 2020-03-20Bibliographically approved
Fatima, A., Schuster, J., Akram, T., Sobol, M., Hoeber, J. & Dahl, N. (2020). Generation of a human Neurochondrin deficient iPSC line KICRi002-A-3 using CRISPR/Cas9. Stem Cell Research, 44, Article ID 101758.
Open this publication in new window or tab >>Generation of a human Neurochondrin deficient iPSC line KICRi002-A-3 using CRISPR/Cas9
Show others...
2020 (English)In: Stem Cell Research, ISSN 1873-5061, E-ISSN 1876-7753, Vol. 44, article id 101758Article in journal, Editorial material (Other academic) Published
Abstract [en]

The role of Neurochondrin (NCDN) in humans is not well understood. Mice with a conditional Ncdn knock-out show epileptic seizures, depressive-like behaviours and impaired spatial learning. Using CRISPR/Cas9, we generated a Neurochondrin deficient human iPSC line KICRi002-A-3 carrying a homozygous 752 bp deletion / 2 bp insertion in the NCDN gene. The iPSC line maintained a normal 46,XY karyotype, expressed pluripotency markers and exhibited capability to differentiate into the three germ layers in vitro. Off-target editing was excluded and Neurochondrin expression was not detectable. The iPSC line offers a valuable resource to study the role of Neurochondrin during human neurogenesis.

National Category
Cell and Molecular Biology
Identifiers
urn:nbn:se:uu:diva-412045 (URN)10.1016/j.scr.2020.101758 (DOI)000528893500017 ()32203915 (PubMedID)
Funder
Swedish Research Council, 2015-02424The Swedish Brain Foundation, FO2019-0210
Available from: 2020-06-08 Created: 2020-06-08 Last updated: 2020-06-08Bibliographically approved
Schuster, J., Hoeber, J., Sobol, M., Fatima, A., Annerén, G. & Dahl, N. (2020). Generation of two human iPSC lines (UUIGPi013-A and UUIPGi014-A) from cases with Down syndrome and full trisomy for chromosome 21 (T21). Stem Cell Research, 49, Article ID 102081.
Open this publication in new window or tab >>Generation of two human iPSC lines (UUIGPi013-A and UUIPGi014-A) from cases with Down syndrome and full trisomy for chromosome 21 (T21)
Show others...
2020 (English)In: Stem Cell Research, ISSN 1873-5061, E-ISSN 1876-7753, Vol. 49, article id 102081Article in journal (Refereed) Published
Abstract [en]

Down syndrome (DS) is caused by trisomy for chromosome 21 (T21). We generated two induced pluripotent stem cell (iPSC) lines from skin fibroblasts of two males with DS using Sendai virus delivery of OCT4, SOX2, KLF4, and c-MYC. Characterization of the two iPSC lines, UUIGPi013-A and UUIPGi014-A, showed that they are genetically stable with a 47,XY,+21 karyotype. Both lines displayed expression of pluripotency markers and trilineage differentiation capacity. These two iPSC lines provide a useful resource for DS modeling and pharmacological interventions.

Place, publisher, year, edition, pages
AMSTERDAM, NETHERLANDS: , 2020
Keywords
Cell Biology; Biotechnology & Applied Microbiology
National Category
Medical Genetics
Identifiers
urn:nbn:se:uu:diva-428382 (URN)10.1016/j.scr.2020.102081 (DOI)000600952800080 ()33220594 (PubMedID)
Funder
Swedish Research Council, 2015-02424AstraZenecaThe Swedish Brain Foundation, FO2020-0171The Swedish Brain Foundation, FO2019-0210Science for Life Laboratory - a national resource center for high-throughput molecular bioscience
Available from: 2021-01-12 Created: 2021-01-12 Last updated: 2021-02-17Bibliographically approved
Organisations
Identifiers
ORCID iD: ORCID iD iconorcid.org/0000-0002-4383-9880

Search in DiVA

Show all publications