65 results on '"Hockemeyer D"'
Search Results
2. A suite of enhancer AAVs and transgenic mouse lines for genetic access to cortical cell types.
- Author
-
Ben-Simon Y, Hooper M, Narayan S, Daigle T, Dwivedi D, Way SW, Oster A, Stafford DA, Mich JK, Taormina MJ, Martinez RA, Opitz-Araya X, Roth JR, Allen S, Ayala A, Bakken TE, Barcelli T, Barta S, Bendrick J, Bertagnolli D, Bowlus J, Boyer G, Brouner K, Casian B, Casper T, Chakka AB, Chakrabarty R, Chance RK, Chavan S, Departee M, Donadio N, Dotson N, Egdorf T, Gabitto M, Garcia J, Gary A, Gasperini M, Goldy J, Gore BB, Graybuck L, Greisman N, Haeseleer F, Halterman C, Helback O, Hockemeyer D, Huang C, Huff S, Hunker A, Johansen N, Juneau Z, Kalmbach B, Khem S, Kussick E, Kutsal R, Larsen R, Lee C, Lee AY, Leibly M, Lenz GH, Liang E, Lusk N, Malone J, Mollenkopf T, Morin E, Newman D, Ng L, Ngo K, Omstead V, Oyama A, Pham T, Pom CA, Potekhina L, Ransford S, Rette D, Rimorin C, Rocha D, Ruiz A, Sanchez REA, Sedeno-Cortes A, Sevigny JP, Shapovalova N, Shulga L, Sigler AR, Siverts LA, Somasundaram S, Stewart K, Szelenyi E, Tieu M, Trader C, van Velthoven CTJ, Walker M, Weed N, Wirthlin M, Wood T, Wynalda B, Yao Z, Zhou T, Ariza J, Dee N, Reding M, Ronellenfitch K, Mufti S, Sunkin SM, Smith KA, Esposito L, Waters J, Thyagarajan B, Yao S, Lein ES, Zeng H, Levi BP, Ngai J, Ting J, and Tasic B
- Abstract
The mammalian cortex is comprised of cells classified into types according to shared properties. Defining the contribution of each cell type to the processes guided by the cortex is essential for understanding its function in health and disease. We used transcriptomic and epigenomic cortical cell type taxonomies from mouse and human to define marker genes and putative enhancers and created a large toolkit of transgenic lines and enhancer AAVs for selective targeting of cortical cell populations. We report evaluation of fifteen new transgenic driver lines, two new reporter lines, and >800 different enhancer AAVs covering most subclasses of cortical cells. The tools reported here as well as the scaled process of tool creation and modification enable diverse experimental strategies towards understanding mammalian cortex and brain function.
- Published
- 2024
- Full Text
- View/download PDF
3. Dissecting the oncogenic mechanisms of POT1 cancer mutations through deep scanning mutagenesis.
- Author
-
Martin A, Schabort J, Bartke-Croughan R, Tran S, Preetham A, Lu R, Ho R, Gao J, Jenkins S, Boyle J, Ghanim GE, Jagota M, Song YS, Li H, and Hockemeyer D
- Abstract
Mutations in the shelterin protein POT1 are associated with diverse cancers, but their role in cancer progression remains unclear. To resolve this, we performed deep scanning mutagenesis in POT1 locally haploid human stem cells to assess the impact of POT1 variants on cellular viability and cancer-associated telomeric phenotypes. Though POT1 is essential, frame-shift mutants are rescued by chemical ATR inhibition, indicating that POT1 is not required for telomere replication or lagging strand synthesis. In contrast, a substantial fraction of clinically-validated pathogenic mutations support normal cellular proliferation, but still drive ATR-dependent telomeric DNA damage signaling and ATR-independent telomere elongation. Moreover, this class of cancer-associated POT1 variants elongates telomeres more rapidly than POT1 frame-shifts, indicating they actively drive oncogenesis and are not simple loss-of-function mutations.
- Published
- 2024
- Full Text
- View/download PDF
4. Digital telomere measurement by long-read sequencing distinguishes healthy aging from disease.
- Author
-
Sanchez SE, Gu Y, Wang Y, Golla A, Martin A, Shomali W, Hockemeyer D, Savage SA, and Artandi SE
- Subjects
- Humans, Adult, Healthy Aging genetics, Middle Aged, Male, Aged, Female, Telomere Shortening genetics, Aging genetics, Nanopore Sequencing methods, Young Adult, Telomere genetics, Telomere metabolism, Telomere Homeostasis genetics, Machine Learning
- Abstract
Telomere length is an important biomarker of organismal aging and cellular replicative potential, but existing measurement methods are limited in resolution and accuracy. Here, we deploy digital telomere measurement (DTM) by nanopore sequencing to understand how distributions of human telomere length change with age and disease. We measure telomere attrition and de novo elongation with up to 30 bp resolution in genetically defined populations of human cells, in blood cells from healthy donors and in blood cells from patients with genetic defects in telomere maintenance. We find that human aging is accompanied by a progressive loss of long telomeres and an accumulation of shorter telomeres. In patients with defects in telomere maintenance, the accumulation of short telomeres is more pronounced and correlates with phenotypic severity. We apply machine learning to train a binary classification model that distinguishes healthy individuals from those with telomere biology disorders. This sequencing and bioinformatic pipeline will advance our understanding of telomere maintenance mechanisms and the use of telomere length as a clinical biomarker of aging and disease., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
5. Distinct senescence mechanisms restrain progression of dysplastic nevi.
- Author
-
Lorbeer FK, Rieser G, Goel A, Wang M, Oh A, Yeh I, Bastian BC, and Hockemeyer D
- Abstract
Telomerase reverse transcriptase (TERT) promoter mutations (TPMs) are frequently found in different cancer types, including ∼70% of sun-exposed skin melanomas. In melanoma, TPMs are among the earliest mutations and can be present during the transition from nevus to melanoma. However, the specific factors that contribute to the selection of TPMs in certain nevi subsets are not well understood. To investigate this, we analyzed a group of dysplastic nevi (DN) by sequencing genes commonly mutated in melanocytic neoplasms. We examined the relationship between the identified mutations, patient age, telomere length, histological features, and the expression of p16. Our findings reveal that TPMs are more prevalent in DN from older patients and are associated with shorter telomeres. Importantly, these TPMs were not found in nevi with BRAF V600E mutations. Conversely, DN with BRAF V600E mutations were observed in younger patients, had longer telomeres and a higher proportion of p16-positive cells. This suggests that these nevi arrest growth independently of telomere shortening through a mechanism known as oncogene-induced senescence (OIS). These characteristics extend to melanoma-sequencing datasets, where melanomas with BRAF V600E mutations were more likely to have a CDKN2A inactivation, overriding OIS. In contrast, melanomas without BRAF V600E mutations showed a higher frequency of TPMs. Our data imply that TPMs are selected to bypass replicative senescence (RS) in cells that were not arrested by OIS. Overall, our results indicate that a subset of melanocytic neoplasms face constraints from RS, while others encounter OIS and RS. The order in which these barriers are overcome during progression to melanoma depends on the mutational context., (© The Author(s) 2024. Published by Oxford University Press on behalf of National Academy of Sciences.)
- Published
- 2024
- Full Text
- View/download PDF
6. iSCORE-PD: an isogenic stem cell collection to research Parkinson's Disease.
- Author
-
Busquets O, Li H, Mohieddin Syed K, Jerez PA, Dunnack J, Bu RL, Verma Y, Pangilinan GR, Martin A, Straub J, Du Y, Simon VM, Poser S, Bush Z, Diaz J, Sahagun A, Gao J, Hernandez DG, Levine KS, Booth EO, Bateup HS, Rio DC, Hockemeyer D, Blauwendraat C, and Soldner F
- Abstract
Parkinson's disease (PD) is a neurodegenerative disorder caused by complex genetic and environmental factors. Genome-edited human pluripotent stem cells (hPSCs) offer the uniique potential to advance our understanding of PD etiology by providing disease-relevant cell-types carrying patient mutations along with isogenic control cells. To facilitate this experimental approach, we generated a collection of 55 cell lines genetically engineered to harbor mutations in genes associated with monogenic PD ( SNCA A53T, SNCA A30P, PRKN Ex3del, PINK1 Q129X, DJ1/PARK7 Ex1-5del, LRRK2 G2019S, ATP13A2 FS, FBXO7 R498X/FS, DNAJC6 c.801 A>G+FS, SYNJ1 R258Q/FS, VPS13C A444P, VPS13C W395C, GBA1 IVS2+1). All mutations were generated in a fully characterized and sequenced female human embryonic stem cell (hESC) line (WIBR3; NIH approval number NIHhESC-10-0079) using CRISPR/Cas9 or prime editing-based approaches. We implemented rigorous quality controls, including high density genotyping to detect structural variants and confirm the genomic integrity of each cell line. This systematic approach ensures the high quality of our stem cell collection, highlights differences between conventional CRISPR/Cas9 and prime editing and provides a roadmap for how to generate gene-edited hPSCs collections at scale in an academic setting. We expect that our isogenic stem cell collection will become an accessible platform for the study of PD, which can be used by investigators to understand the molecular pathophysiology of PD in a human cellular setting.
- Published
- 2024
- Full Text
- View/download PDF
7. Functional annotation of variants of the BRCA2 gene via locally haploid human pluripotent stem cells.
- Author
-
Li H, Bartke R, Zhao L, Verma Y, Horacek A, Rechav Ben-Natan A, Pangilinan GR, Krishnappa N, Nielsen R, and Hockemeyer D
- Subjects
- Humans, Genes, BRCA2, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Haploidy, Amino Acids, BRCA2 Protein genetics, Neoplasms, Pluripotent Stem Cells
- Abstract
Mutations in the BRCA2 gene are associated with sporadic and familial cancer, cause genomic instability and sensitize cancer cells to inhibition by the poly(ADP-ribose) polymerase (PARP). Here we show that human pluripotent stem cells (hPSCs) with one copy of BRCA2 deleted can be used to annotate variants of this gene and to test their sensitivities to PARP inhibition. By using Cas9 to edit the functional BRCA2 allele in the locally haploid hPSCs and in fibroblasts differentiated from them, we characterized essential regions in the gene to identify permissive and loss-of-function mutations. We also used Cas9 to directly test the function of individual amino acids, including amino acids encoded by clinical BRCA2 variants of uncertain significance, and identified alleles that are sensitive to PARP inhibitors used as a standard of care in BRCA2-deficient cancers. Locally haploid human pluripotent stem cells can facilitate detailed structure-function analyses of genes and the rapid functional evaluation of clinically observed mutations., (© 2023. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
8. Conserved chromatin and repetitive patterns reveal slow genome evolution in frogs.
- Author
-
Bredeson JV, Mudd AB, Medina-Ruiz S, Mitros T, Smith OK, Miller KE, Lyons JB, Batra SS, Park J, Berkoff KC, Plott C, Grimwood J, Schmutz J, Aguirre-Figueroa G, Khokha MK, Lane M, Philipp I, Laslo M, Hanken J, Kerdivel G, Buisine N, Sachs LM, Buchholz DR, Kwon T, Smith-Parker H, Gridi-Papp M, Ryan MJ, Denton RD, Malone JH, Wallingford JB, Straight AF, Heald R, Hockemeyer D, Harland RM, and Rokhsar DS
- Subjects
- Animals, Genome genetics, Anura genetics, Xenopus genetics, Centromere genetics, Chromatin genetics, Evolution, Molecular
- Abstract
Frogs are an ecologically diverse and phylogenetically ancient group of anuran amphibians that include important vertebrate cell and developmental model systems, notably the genus Xenopus. Here we report a high-quality reference genome sequence for the western clawed frog, Xenopus tropicalis, along with draft chromosome-scale sequences of three distantly related emerging model frog species, Eleutherodactylus coqui, Engystomops pustulosus, and Hymenochirus boettgeri. Frog chromosomes have remained remarkably stable since the Mesozoic Era, with limited Robertsonian (i.e., arm-preserving) translocations and end-to-end fusions found among the smaller chromosomes. Conservation of synteny includes conservation of centromere locations, marked by centromeric tandem repeats associated with Cenp-a binding surrounded by pericentromeric LINE/L1 elements. This work explores the structure of chromosomes across frogs, using a dense meiotic linkage map for X. tropicalis and chromatin conformation capture (Hi-C) data for all species. Abundant satellite repeats occupy the unusually long (~20 megabase) terminal regions of each chromosome that coincide with high rates of recombination. Both embryonic and differentiated cells show reproducible associations of centromeric chromatin and of telomeres, reflecting a Rabl-like configuration. Our comparative analyses reveal 13 conserved ancestral anuran chromosomes from which contemporary frog genomes were constructed., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
9. Digital telomere measurement by long-read sequencing distinguishes healthy aging from disease.
- Author
-
Sanchez SE, Gu J, Golla A, Martin A, Shomali W, Hockemeyer D, Savage SA, and Artandi SE
- Abstract
Telomere length is an important biomarker of organismal aging and cellular replicative potential, but existing measurement methods are limited in resolution and accuracy. Here, we deploy digital telomere measurement by nanopore sequencing to understand how distributions of human telomere length change with age and disease. We measure telomere attrition and de novo elongation with unprecedented resolution in genetically defined populations of human cells, in blood cells from healthy donors and in blood cells from patients with genetic defects in telomere maintenance. We find that human aging is accompanied by a progressive loss of long telomeres and an accumulation of shorter telomeres. In patients with defects in telomere maintenance, the accumulation of short telomeres is more pronounced and correlates with phenotypic severity. We apply machine learning to train a binary classification model that distinguishes healthy individuals from those with telomere biology disorders. This sequencing and bioinformatic pipeline will advance our understanding of telomere maintenance mechanisms and the use of telomere length as a clinical biomarker of aging and disease., Competing Interests: Competing interests: Authors declare that they have no competing interests.
- Published
- 2023
- Full Text
- View/download PDF
10. Tracing cancer evolution and heterogeneity using Hi-C.
- Author
-
Erdmann-Pham DD, Batra SS, Turkalo TK, Durbin J, Blanchette M, Yeh I, Shain H, Bastian BC, Song YS, Rokhsar DS, and Hockemeyer D
- Subjects
- Humans, DNA Copy Number Variations, Chromosomes, Translocation, Genetic, Chromosome Aberrations, Melanoma genetics
- Abstract
Chromosomal rearrangements can initiate and drive cancer progression, yet it has been challenging to evaluate their impact, especially in genetically heterogeneous solid cancers. To address this problem we developed HiDENSEC, a new computational framework for analyzing chromatin conformation capture in heterogeneous samples that can infer somatic copy number alterations, characterize large-scale chromosomal rearrangements, and estimate cancer cell fractions. After validating HiDENSEC with in silico and in vitro controls, we used it to characterize chromosome-scale evolution during melanoma progression in formalin-fixed tumor samples from three patients. The resulting comprehensive annotation of the genomic events includes copy number neutral translocations that disrupt tumor suppressor genes such as NF1, whole chromosome arm exchanges that result in loss of CDKN2A, and whole-arm copy-number neutral loss of homozygosity involving PTEN. These findings show that large-scale chromosomal rearrangements occur throughout cancer evolution and that characterizing these events yields insights into drivers of melanoma progression., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
11. Distinct senescence mechanisms restrain progression of dysplastic nevi.
- Author
-
Lorbeer FK, Rieser G, Goel A, Wang M, Oh A, Yeh I, Bastian BC, and Hockemeyer D
- Abstract
TERT promoter mutations (TPMs) are frequently found in different cancer types, including approximately 70% of sun-exposed skin melanomas. In melanoma, TPMs are among the earliest mutations and can be present during the transition from nevus to melanoma. However, the specific factors that contribute to the selection of TPMs in certain nevi subsets are not well understood. To investigate this, we analyzed a group of dysplastic nevi (DN) by sequencing genes commonly mutated in melanocytic neoplasms. We examined the relationship between the identified mutations, patient age, telomere length, histological features, and the expression of p16. Our findings reveal that TPMs are more prevalent in DN from older patients and are associated with shorter telomeres. Importantly, these TPMs were not found in nevi with BRAF V600E mutations. Conversely, DN with BRAF V600E mutations were observed in younger patients, had longer telomeres, and a higher proportion of p16-positive cells. This suggests that these nevi arrest growth independently of telomere shortening through a mechanism known as oncogene-induced senescence (OIS). These characteristics extend to melanoma sequencing data sets, where melanomas with BRAF V600E mutations were more likely to have CDKN2A inactivation, overriding OIS. In contrast, melanomas without BRAF V600E mutations showed a higher frequency of TPMs. Our data imply that TPMs are selected to bypass replicative senescence (RS) in cells that were not arrested by OIS. Overall, our results indicate that a subset of melanocytic neoplasms face constraints from RS, while others encounter OIS and RS. The order in which these barriers are overcome during progression to melanoma depends on the mutational context.
- Published
- 2023
- Full Text
- View/download PDF
12. A non-genetic switch triggers alternative telomere lengthening and cellular immortalization in ATRX deficient cells.
- Author
-
Turkalo TK, Maffia A, Schabort JJ, Regalado SG, Bhakta M, Blanchette M, Spierings DCJ, Lansdorp PM, and Hockemeyer D
- Subjects
- Humans, Telomere Homeostasis genetics, Telomere genetics, Cell Differentiation genetics, X-linked Nuclear Protein genetics, Telomerase genetics, Pluripotent Stem Cells
- Abstract
Alternative Lengthening of Telomeres (ALT) is an aberrant DNA recombination pathway which grants replicative immortality to approximately 10% of all cancers. Despite this high prevalence of ALT in cancer, the mechanism and genetics by which cells activate this pathway remain incompletely understood. A major challenge in dissecting the events that initiate ALT is the extremely low frequency of ALT induction in human cell systems. Guided by the genetic lesions that have been associated with ALT from cancer sequencing studies, we genetically engineered primary human pluripotent stem cells to deterministically induce ALT upon differentiation. Using this genetically defined system, we demonstrate that disruption of the p53 and Rb pathways in combination with ATRX loss-of-function is sufficient to induce all hallmarks of ALT and results in functional immortalization in a cell type-specific manner. We further demonstrate that ALT can be induced in the presence of telomerase, is neither dependent on telomere shortening nor crisis, but is rather driven by continuous telomere instability triggered by the induction of differentiation in ATRX-deficient stem cells., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
13. Highly efficient generation of isogenic pluripotent stem cell models using prime editing.
- Author
-
Li H, Busquets O, Verma Y, Syed KM, Kutnowski N, Pangilinan GR, Gilbert LA, Bateup HS, Rio DC, Hockemeyer D, and Soldner F
- Subjects
- Humans, Gene Editing methods, Deoxyribonuclease I genetics, Deoxyribonuclease I metabolism, RNA, Messenger metabolism, RNA-Directed DNA Polymerase, Ribonucleoproteins metabolism, CRISPR-Cas Systems, RNA, Guide, CRISPR-Cas Systems, Pluripotent Stem Cells metabolism
- Abstract
The recent development of prime editing (PE) genome engineering technologies has the potential to significantly simplify the generation of human pluripotent stem cell (hPSC)-based disease models. PE is a multicomponent editing system that uses a Cas9-nickase fused to a reverse transcriptase (nCas9-RT) and an extended PE guide RNA (pegRNA). Once reverse transcribed, the pegRNA extension functions as a repair template to introduce precise designer mutations at the target site. Here, we systematically compared the editing efficiencies of PE to conventional gene editing methods in hPSCs. This analysis revealed that PE is overall more efficient and precise than homology-directed repair of site-specific nuclease-induced double-strand breaks. Specifically, PE is more effective in generating heterozygous editing events to create autosomal dominant disease-associated mutations. By stably integrating the nCas9-RT into hPSCs we achieved editing efficiencies equal to those reported for cancer cells, suggesting that the expression of the PE components, rather than cell-intrinsic features, limit PE in hPSCs. To improve the efficiency of PE in hPSCs, we optimized the delivery modalities for the PE components. Delivery of the nCas9-RT as mRNA combined with synthetically generated, chemically-modified pegRNAs and nicking guide RNAs improved editing efficiencies up to 13-fold compared with transfecting the PE components as plasmids or ribonucleoprotein particles. Finally, we demonstrated that this mRNA-based delivery approach can be used repeatedly to yield editing efficiencies exceeding 60% and to correct or introduce familial mutations causing Parkinson's disease in hPSCs., Competing Interests: HL, OB, YV, KS, NK, GP, LG, HB, DR, DH, FS No competing interests declared, (© 2022, Li, Busquets et al.)
- Published
- 2022
- Full Text
- View/download PDF
14. Editing TINF2 as a potential therapeutic approach to restore telomere length in dyskeratosis congenita.
- Author
-
Choo S, Lorbeer FK, Regalado SG, Short SB, Wu S, Rieser G, Bertuch AA, and Hockemeyer D
- Subjects
- Humans, Mutation, Telomere genetics, Telomere metabolism, Telomere Shortening genetics, Telomere-Binding Proteins genetics, Telomere-Binding Proteins metabolism, Dyskeratosis Congenita genetics, Dyskeratosis Congenita therapy, Telomerase genetics, Telomerase metabolism
- Abstract
Mutations in the TINF2 gene, encoding the shelterin protein TIN2, cause telomere shortening and the inherited bone marrow (BM) failure syndrome dyskeratosis congenita (DC). A lack of suitable model systems limits the mechanistic understanding of telomere shortening in the stem cells and thus hinders the development of treatment options for BM failure. Here, we endogenously introduced TIN2-DC mutations in human embryonic stem cells (hESCs) and human hematopoietic stem and progenitor cells (HSPCs) to dissect the disease mechanism and identify a gene-editing strategy that rescued the disease phenotypes. The hESCs with the T284R disease mutation exhibited the short telomere phenotype observed in DC patients. Yet, telomeres in mutant hESCs did not trigger DNA damage responses at telomeres or show exacerbated telomere shortening when differentiated into telomerase-negative cells. Disruption of the mutant TINF2 allele by introducing a frameshift mutation in exon 2 restored telomere length in stem cells and the replicative potential of differentiated cells. Similarly, we introduced TIN2-DC disease variants in human HSPCs to assess the changes in telomere length and proliferative capacity. Lastly, we showed that editing at exon 2 of TINF2 that restored telomere length in hESCs could be generated in TINF2-DC patient HSPCs. Our study demonstrates a simple genetic intervention that rescues the TIN2-DC disease phenotype in stem cells and provides a versatile platform to assess the efficacy of potential therapeutic approaches in vivo., (© 2022 by The American Society of Hematology.)
- Published
- 2022
- Full Text
- View/download PDF
15. Cancer-associated POT1 mutations lead to telomere elongation without induction of a DNA damage response.
- Author
-
Kim WT, Hennick K, Johnson J, Finnerty B, Choo S, Short SB, Drubin C, Forster R, McMaster ML, and Hockemeyer D
- Subjects
- Animals, DNA Damage, Female, Humans, K562 Cells, Male, Mice, Mutation, Shelterin Complex, Stem Cells, Neoplasms genetics, Telomere, Telomere-Binding Proteins genetics
- Abstract
Mutations in the shelterin protein POT1 are associated with chronic lymphocytic leukemia (CLL), Hodgkin lymphoma, angiosarcoma, melanoma, and other cancers. These cancer-associated POT1 (caPOT1) mutations are generally heterozygous, missense, or nonsense mutations occurring throughout the POT1 reading frame. Cancers with caPOT1 mutations have elongated telomeres and show increased genomic instability, but which of the two phenotypes promotes tumorigenesis is unclear. We tested the effects of CAS9-engineered caPOT1 mutations in human embryonic and hematopoietic stem cells (hESCs and HSCs, respectively). HSCs with caPOT1 mutations did not show overt telomere damage. In vitro and in vivo competition experiments showed the caPOT1 mutations did not confer a selective disadvantage. Since DNA damage signaling is known to affect the fitness of HSCs, the data argue that caPOT1 mutations do not cause significant telomere damage. Furthermore, hESC lines with caPOT1 mutations showed no detectable telomere damage response while showing consistent telomere elongation. Thus, caPOT1 mutations are likely selected for during cancer progression because of their ability to elongate telomeres and extend the proliferative capacity of the incipient cancer cells., (© 2021 The Authors. Published under the terms of the CC BY NC ND 4.0 license.)
- Published
- 2021
- Full Text
- View/download PDF
16. Launching a saliva-based SARS-CoV-2 surveillance testing program on a university campus.
- Author
-
Ehrenberg AJ, Moehle EA, Brook CE, Doudna Cate AH, Witkowsky LB, Sachdeva R, Hirsh A, Barry K, Hamilton JR, Lin-Shiao E, McDevitt S, Valentin-Alvarado L, Letourneau KN, Hunter L, Keller A, Pestal K, Frankino PA, Murley A, Nandakumar D, Stahl EC, Tsuchida CA, Gildea HK, Murdock AG, Hochstrasser ML, O'Brien E, Ciling A, Tsitsiklis A, Worden K, Dugast-Darzacq C, Hays SG, Barber CC, McGarrigle R, Lam EK, Ensminger DC, Bardet L, Sherry C, Harte A, Nicolette G, Giannikopoulos P, Hockemeyer D, Petersen M, Urnov FD, Ringeisen BR, Boots M, and Doudna JA
- Subjects
- Adult, Aged, COVID-19 epidemiology, COVID-19 virology, COVID-19 Testing methods, Female, Humans, Male, Middle Aged, RNA, Viral metabolism, Reverse Transcriptase Polymerase Chain Reaction, SARS-CoV-2 genetics, SARS-CoV-2 isolation & purification, Social Norms, Surveys and Questionnaires, Universities, Young Adult, COVID-19 diagnosis, Program Evaluation, Saliva virology
- Abstract
Regular surveillance testing of asymptomatic individuals for SARS-CoV-2 has been center to SARS-CoV-2 outbreak prevention on college and university campuses. Here we describe the voluntary saliva testing program instituted at the University of California, Berkeley during an early period of the SARS-CoV-2 pandemic in 2020. The program was administered as a research study ahead of clinical implementation, enabling us to launch surveillance testing while continuing to optimize the assay. Results of both the testing protocol itself and the study participants' experience show how the program succeeded in providing routine, robust testing capable of contributing to outbreak prevention within a campus community and offer strategies for encouraging participation and a sense of civic responsibility., Competing Interests: The authors have read the journal’s policy and the authors of this manuscript have the following competing interests: The Regents of the University of California have patents issued and pending for CRISPR technologies on which JAD is an inventor. JAD is a co-founder of Caribou Biosciences, Editas Medicine, Scribe Therapeutics, Intellia Therapeutics, and Mammoth Biosciences. JAD is a scientific advisory board member of Caribou Biosciences, Intellia Therapeutics, eFFECTOR Therapeutics, Scribe Therapeutics, Mammoth Biosciences, Synthego, Algen Biotechnologies, Felix Biosciences, and Inari. JAD is a Director at Johnson & Johnson and Tempus Labs and has research projects sponsored by Biogen, Pfizer, AppleTree Partners, and Roche. FDU is a co-founder of Tune Therapeutics. PG is a co-founder and Director at NewCo Health. PG is the CLIA Laboratory Director for Coral Genomics and 3DMed. This does not alter our adherence to PLOS ONE policies on sharing data and materials.
- Published
- 2021
- Full Text
- View/download PDF
17. Generation of a DAT-P2A-Flpo mouse line for intersectional genetic targeting of dopamine neuron subpopulations.
- Author
-
Kramer DJ, Aisenberg EE, Kosillo P, Friedmann D, Stafford DA, Lee AY, Luo L, Hockemeyer D, Ngai J, and Bateup HS
- Subjects
- Animals, Cell Line, Humans, Mice, Dopamine Plasma Membrane Transport Proteins metabolism, Dopaminergic Neurons metabolism
- Abstract
Dopaminergic projections exert widespread influence over multiple brain regions and modulate various behaviors including movement, reward learning, and motivation. It is increasingly appreciated that dopamine neurons are heterogeneous in their gene expression, circuitry, physiology, and function. Current approaches to target dopamine neurons are largely based on single gene drivers, which either label all dopamine neurons or mark a subset but concurrently label non-dopaminergic neurons. Here, we establish a mouse line with Flpo recombinase expressed from the endogenous Slc6a3 (dopamine active transporter [DAT]) locus. DAT-P2A-Flpo mice can be used together with Cre-expressing mouse lines to efficiently and selectively label dopaminergic subpopulations using Cre/Flp-dependent intersectional strategies. We demonstrate the utility of this approach by generating DAT-P2A-Flpo;NEX-Cre mice that specifically label Neurod6-expressing dopamine neurons, which project to the nucleus accumbens medial shell. DAT-P2A-Flpo mice add to a growing toolbox of genetic resources that will help parse the diverse functions mediated by dopaminergic circuits., Competing Interests: Declaration of interests The authors declare no financial or non-financial competing interests., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
18. TINF2 is a haploinsufficient tumor suppressor that limits telomere length.
- Author
-
Schmutz I, Mensenkamp AR, Takai KK, Haadsma M, Spruijt L, de Voer RM, Choo SS, Lorbeer FK, van Grinsven EJ, Hockemeyer D, Jongmans MC, and de Lange T
- Subjects
- Cell Line, Female, HEK293 Cells, Heterozygote, Humans, Loss of Function Mutation, Male, Neoplasms genetics, Telomere pathology, Telomere-Binding Proteins genetics, Telomeric Repeat Binding Protein 1 metabolism, Tumor Suppressor Proteins, Genes, Tumor Suppressor, Telomere genetics, Telomere Shortening genetics, Telomere-Binding Proteins physiology
- Abstract
Telomere shortening is a presumed tumor suppressor pathway that imposes a proliferative barrier (the Hayflick limit) during tumorigenesis. This model predicts that excessively long somatic telomeres predispose to cancer. Here, we describe cancer-prone families with two unique TINF2 mutations that truncate TIN2, a shelterin subunit that controls telomere length. Patient lymphocyte telomeres were unusually long. We show that the truncated TIN2 proteins do not localize to telomeres, suggesting that the mutations create loss-of-function alleles. Heterozygous knock-in of the mutations or deletion of one copy of TINF2 resulted in excessive telomere elongation in clonal lines, indicating that TINF2 is haploinsufficient for telomere length control. In contrast, telomere protection and genome stability were maintained in all heterozygous clones. The data establish that the TINF2 truncations predispose to a tumor syndrome. We conclude that TINF2 acts as a haploinsufficient tumor suppressor that limits telomere length to ensure a timely Hayflick limit., Competing Interests: IS, AM, KT, MH, LS, Rd, SC, FL, Ev, DH, MJ No competing interests declared, Td Member of the SAB of Calico Life Sciences LLC, (© 2020, Schmutz et al.)
- Published
- 2020
- Full Text
- View/download PDF
19. Telomere length set point regulation in human pluripotent stem cells critically depends on the shelterin protein TPP1.
- Author
-
Boyle JM, Hennick KM, Regalado SG, Vogan JM, Zhang X, Collins K, and Hockemeyer D
- Subjects
- HeLa Cells, Humans, Mutation, Protein Isoforms, Shelterin Complex, Telomerase metabolism, Telomere genetics, Telomere metabolism, Telomere Homeostasis genetics, Telomere-Binding Proteins physiology, Pluripotent Stem Cells metabolism, Telomere Homeostasis physiology, Telomere-Binding Proteins metabolism
- Abstract
Telomere maintenance is essential for the long-term proliferation of human pluripotent stem cells, while their telomere length set point determines the proliferative capacity of their differentiated progeny. The shelterin protein TPP1 is required for telomere stability and elongation, but its role in establishing a telomere length set point remains elusive. Here, we characterize the contribution of the shorter isoform of TPP1 (TPP1S) and the amino acid L104 outside the TEL patch, TPP1's telomerase interaction domain, to telomere length control. We demonstrate that cells deficient for TPP1S (TPP1S knockout [KO]), as well as the complete TPP1 KO cell lines, undergo telomere shortening. However, TPP1S KO cells are able to stabilize short telomeres, while TPP1 KO cells die. We compare these phenotypes with those of TPP1
L104A/L104A mutant cells, which have short and stable telomeres similar to the TPP1S KO. In contrast to TPP1S KO cells, TPP1L104A/L104A cells respond to increased telomerase levels and maintain protected telomeres. However, TPP1L104A/L104A shows altered sensitivity to expression changes of shelterin proteins suggesting the mutation causes a defect in telomere length feedback regulation. Together this highlights TPP1L104A/L104A as the first shelterin mutant engineered at the endogenous locus of human stem cells with an altered telomere length set point.- Published
- 2020
- Full Text
- View/download PDF
20. Controlled Cycling and Quiescence Enables Efficient HDR in Engraftment-Enriched Adult Hematopoietic Stem and Progenitor Cells.
- Author
-
Shin JJ, Schröder MS, Caiado F, Wyman SK, Bray NL, Bordi M, Dewitt MA, Vu JT, Kim WT, Hockemeyer D, Manz MG, and Corn JE
- Subjects
- Humans, CRISPR-Cas Systems genetics, GATA3 Transcription Factor metabolism, Gene Editing methods, Genetic Therapy methods, Hematopoietic Stem Cells metabolism, Recombinational DNA Repair genetics, Stem Cells metabolism
- Abstract
Genome editing often takes the form of either error-prone sequence disruption by non-homologous end joining (NHEJ) or sequence replacement by homology-directed repair (HDR). Although NHEJ is generally effective, HDR is often difficult in primary cells. Here, we use a combination of immunophenotyping, next-generation sequencing, and single-cell RNA sequencing to investigate and reprogram genome editing outcomes in subpopulations of adult hematopoietic stem and progenitor cells. We find that although quiescent stem-enriched cells mostly use NHEJ, non-quiescent cells with the same immunophenotype use both NHEJ and HDR. Inducing quiescence before editing results in a loss of HDR in all cell subtypes. We develop a strategy of controlled cycling and quiescence that yields a 6-fold increase in the HDR/NHEJ ratio in quiescent stem cells ex vivo and in vivo. Our results highlight the tension between editing and cellular physiology and suggest strategies to manipulate quiescent cells for research and therapeutic genome editing., Competing Interests: Declaration of Interests The authors declare no competing interests., (Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
21. Analysis of muntjac deer genome and chromatin architecture reveals rapid karyotype evolution.
- Author
-
Mudd AB, Bredeson JV, Baum R, Hockemeyer D, and Rokhsar DS
- Subjects
- Animals, Chromosomes, Mammalian genetics, Female, Phylogeny, Chromatin genetics, Evolution, Molecular, Genome, Karyotype, Muntjacs genetics
- Abstract
Closely related muntjac deer show striking karyotype differences. Here we describe chromosome-scale genome assemblies for Chinese and Indian muntjacs, Muntiacus reevesi (2n = 46) and Muntiacus muntjak vaginalis (2n = 6/7), and analyze their evolution and architecture. The genomes show extensive collinearity with each other and with other deer and cattle. We identified numerous fusion events unique to and shared by muntjacs relative to the cervid ancestor, confirming many cytogenetic observations with genome sequence. One of these M. muntjak fusions reversed an earlier fission in the cervid lineage. Comparative Hi-C analysis showed that the chromosome fusions on the M. muntjak lineage altered long-range, three-dimensional chromosome organization relative to M. reevesi in interphase nuclei including A/B compartment structure. This reshaping of multi-megabase contacts occurred without notable change in local chromatin compaction, even near fusion sites. A few genes involved in chromosome maintenance show evidence for rapid evolution, possibly associated with the dramatic changes in karyotype.
- Published
- 2020
- Full Text
- View/download PDF
22. TERT promoter mutations and telomeres during tumorigenesis.
- Author
-
Lorbeer FK and Hockemeyer D
- Subjects
- Carcinogenesis genetics, Carcinogenesis metabolism, Gene Expression Regulation, Neoplastic, Humans, Neoplasms enzymology, Neoplasms genetics, Carcinogenesis pathology, DNA Methylation, Neoplasms pathology, Promoter Regions, Genetic, Telomerase genetics, Telomere, Telomere Homeostasis
- Abstract
Telomerase regulation and telomere shortening act as a strong tumor suppressor mechanism in human somatic cells. Point mutations in the promoter of telomerase reverse transcriptase (TERT) are the most frequent non-coding mutation in cancer. These TERT promoter mutations (TPMs) create de novo ETS factor binding sites upstream of the start codon of the gene, which can be bound by different ETS factors. TPMs can occur early during tumorigenesis and are thought to be among the first mutations in melanoma, glioblastoma and hepatocellular carcinoma. Despite their association with increased TERT levels, TPMs do not prohibit telomere shortening and TPM-harboring cancers present with short telomeres. Their short telomere length combined with their high prevalence and specificity for cancer makes TPMs an attractive target for future therapeutic exploitation of telomerase inhibition and telomere deprotection-induced cell death., (Copyright © 2020. Published by Elsevier Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
23. Transient activation of the UPR ER is an essential step in the acquisition of pluripotency during reprogramming.
- Author
-
Simic MS, Moehle EA, Schinzel RT, Lorbeer FK, Halloran JJ, Heydari K, Sanchez M, Jullié D, Hockemeyer D, and Dillin A
- Subjects
- Cells, Cultured, Fibroblasts physiology, Humans, Induced Pluripotent Stem Cells physiology, Proteome analysis, Signal Transduction, Cellular Reprogramming, Endoplasmic Reticulum physiology, Endoplasmic Reticulum Stress, Fibroblasts cytology, Heat-Shock Response, Induced Pluripotent Stem Cells cytology, Unfolded Protein Response
- Abstract
Somatic cells can be reprogrammed into pluripotent stem cells using the Yamanaka transcription factors. Reprogramming requires both epigenetic landscape reshaping and global remodeling of cell identity, structure, basic metabolic processes, and organelle form and function. We hypothesize that variable regulation of the proteostasis network and its influence upon the protein-folding environment within cells and their organelles is responsible for the low efficiency and stochasticity of reprogramming. We find that the unfolded protein response of the endoplasmic reticulum (UPR
ER ), the mitochondrial UPR, and the heat shock response, which ensure proteome quality during stress, are activated during reprogramming. The UPRER is particularly crucial, and its ectopic, transient activation, genetically or pharmacologically, enhances reprogramming. Last, stochastic activation of the UPRER predicts reprogramming efficiency in naïve cells. Thus, the low efficiency and stochasticity of cellular reprogramming are due partly to the inability to properly initiate the UPRER to remodel the ER and its proteome.- Published
- 2019
- Full Text
- View/download PDF
24. 4D cell biology: big data image analytics and lattice light-sheet imaging reveal dynamics of clathrin-mediated endocytosis in stem cell-derived intestinal organoids.
- Author
-
Schöneberg J, Dambournet D, Liu TL, Forster R, Hockemeyer D, Betzig E, and Drubin DG
- Subjects
- Animals, Big Data, Cell Culture Techniques methods, Cell Differentiation physiology, Dynamin II metabolism, Endocytosis physiology, Human Embryonic Stem Cells metabolism, Humans, Intestinal Mucosa cytology, Intestinal Mucosa metabolism, Mice, Organoids cytology, Organoids diagnostic imaging, Organoids metabolism, Clathrin metabolism, Clathrin-Coated Vesicles metabolism, Human Embryonic Stem Cells cytology, Image Processing, Computer-Assisted methods, Intestines cytology
- Abstract
New methods in stem cell 3D organoid tissue culture, advanced imaging, and big data image analytics now allow tissue-scale 4D cell biology, but currently available analytical pipelines are inadequate for handing and analyzing the resulting gigabytes and terabytes of high-content imaging data. We expressed fluorescent protein fusions of clathrin and dynamin2 at endogenous levels in genome-edited human embryonic stem cells, which were differentiated into hESC-derived intestinal epithelial organoids. Lattice light-sheet imaging with adaptive optics (AO-LLSM) allowed us to image large volumes of these organoids (70 × 60 × 40 µm xyz) at 5.7 s/frame. We developed an open-source data analysis package termed pyLattice to process the resulting large (∼60 Gb) movie data sets and to track clathrin-mediated endocytosis (CME) events. CME tracks could be recorded from ∼35 cells at a time, resulting in ∼4000 processed tracks per movie. On the basis of their localization in the organoid, we classified CME tracks into apical, lateral, and basal events and found that CME dynamics is similar for all three classes, despite reported differences in membrane tension. pyLattice coupled with AO-LLSM makes possible quantitative high temporal and spatial resolution analysis of subcellular events within tissues.
- Published
- 2018
- Full Text
- View/download PDF
25. Genetically engineered human cortical spheroid models of tuberous sclerosis.
- Author
-
Blair JD, Hockemeyer D, and Bateup HS
- Subjects
- CRISPR-Cas Systems genetics, Cell Line, Cerebral Cortex metabolism, Cerebral Cortex pathology, Humans, Neuroglia metabolism, Neuroglia pathology, Neurons metabolism, Neurons pathology, Pluripotent Stem Cells transplantation, Spheroids, Cellular metabolism, Tuberous Sclerosis physiopathology, Genetic Engineering, Tuberous Sclerosis genetics, Tuberous Sclerosis Complex 1 Protein genetics, Tuberous Sclerosis Complex 2 Protein genetics
- Abstract
Tuberous sclerosis complex (TSC) is a multisystem developmental disorder caused by mutations in the TSC1 or TSC2 genes, whose protein products are negative regulators of mechanistic target of rapamycin complex 1 signaling. Hallmark pathologies of TSC are cortical tubers-regions of dysmorphic, disorganized neurons and glia in the cortex that are linked to epileptogenesis. To determine the developmental origin of tuber cells, we established human cellular models of TSC by CRISPR-Cas9-mediated gene editing of TSC1 or TSC2 in human pluripotent stem cells (hPSCs). Using heterozygous TSC2 hPSCs with a conditional mutation in the functional allele, we show that mosaic biallelic inactivation during neural progenitor expansion is necessary for the formation of dysplastic cells and increased glia production in three-dimensional cortical spheroids. Our findings provide support for the second-hit model of cortical tuber formation and suggest that variable developmental timing of somatic mutations could contribute to the heterogeneity in the neurological presentation of TSC.
- Published
- 2018
- Full Text
- View/download PDF
26. Genome-edited human stem cells expressing fluorescently labeled endocytic markers allow quantitative analysis of clathrin-mediated endocytosis during differentiation.
- Author
-
Dambournet D, Sochacki KA, Cheng AT, Akamatsu M, Taraska JW, Hockemeyer D, and Drubin DG
- Subjects
- Animals, Cell Line, Clathrin metabolism, Embryonic Stem Cells metabolism, Fibroblasts metabolism, Gene Editing, Gene Expression Regulation, Developmental, Humans, Mice, Microscopy, Electron, Transmission methods, Neural Stem Cells metabolism, Phosphatidylinositol 3-Kinase metabolism, Adaptor Proteins, Vesicular Transport biosynthesis, Cell Differentiation physiology, Embryonic Stem Cells cytology, Endocytosis physiology, Fibroblasts cytology, Neural Stem Cells cytology
- Abstract
We developed a general approach for investigation of how cellular processes become adapted for specific cell types during differentiation. Previous studies reported substantial differences in the morphology and dynamics of clathrin-mediated endocytosis (CME) sites. However, associating specific CME properties with distinct differentiated cell types and determining how these properties are developmentally specified during differentiation have been elusive. Using genome-edited human embryonic stem cells, and isogenic fibroblasts and neuronal progenitor cells derived from them, we established by live-cell imaging and platinum replica transmission electron microscopy that CME site dynamics and ultrastructure on the plasma membrane are precisely reprogrammed during differentiation. Expression levels for the endocytic adaptor protein AP2μ2 were found to underlie dramatic changes in CME dynamics and structure. Additionally, CME dependency on actin assembly and phosphoinositide-3 kinase activity are distinct for each cell type. Collectively, our results demonstrate that key CME properties are reprogrammed during differentiation at least in part through AP2μ2 expression regulation., (© 2018 Dambournet et al.)
- Published
- 2018
- Full Text
- View/download PDF
27. Observing the cell in its native state: Imaging subcellular dynamics in multicellular organisms.
- Author
-
Liu TL, Upadhyayula S, Milkie DE, Singh V, Wang K, Swinburne IA, Mosaliganti KR, Collins ZM, Hiscock TW, Shea J, Kohrman AQ, Medwig TN, Dambournet D, Forster R, Cunniff B, Ruan Y, Yashiro H, Scholpp S, Meyerowitz EM, Hockemeyer D, Drubin DG, Martin BL, Matus DQ, Koyama M, Megason SG, Kirchhausen T, and Betzig E
- Subjects
- Animals, Cell Movement, Endocytosis, Eye ultrastructure, Humans, Mitosis, Organelles, Single-Cell Analysis, Zebrafish, Imaging, Three-Dimensional methods, Microscopy methods
- Abstract
True physiological imaging of subcellular dynamics requires studying cells within their parent organisms, where all the environmental cues that drive gene expression, and hence the phenotypes that we actually observe, are present. A complete understanding also requires volumetric imaging of the cell and its surroundings at high spatiotemporal resolution, without inducing undue stress on either. We combined lattice light-sheet microscopy with adaptive optics to achieve, across large multicellular volumes, noninvasive aberration-free imaging of subcellular processes, including endocytosis, organelle remodeling during mitosis, and the migration of axons, immune cells, and metastatic cancer cells in vivo. The technology reveals the phenotypic diversity within cells across different organisms and developmental stages and may offer insights into how cells harness their intrinsic variability to adapt to different physiological environments., (Copyright © 2018 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2018
- Full Text
- View/download PDF
28. Single Molecule Imaging in Live Embryos Using Lattice Light-Sheet Microscopy.
- Author
-
Mir M, Reimer A, Stadler M, Tangara A, Hansen AS, Hockemeyer D, Eisen MB, Garcia H, and Darzacq X
- Subjects
- Animals, Data Analysis, Embryo, Mammalian cytology, Embryo, Mammalian diagnostic imaging, Embryo, Nonmammalian cytology, Mice, Reproducibility of Results, Drosophila melanogaster embryology, Embryo, Nonmammalian diagnostic imaging, Microscopy, Fluorescence methods, Single Molecule Imaging methods
- Abstract
In the past decade, live-cell single molecule imaging studies have provided unique insights on how DNA-binding molecules such as transcription factors explore the nuclear environment to search for and bind to their targets. However, due to technological limitations, single molecule experiments in living specimens have largely been limited to monolayer cell cultures. Lattice light-sheet microscopy overcomes these limitations and has now enabled single molecule imaging within thicker specimens such as embryos. Here we describe a general procedure to perform single molecule imaging in living Drosophila melanogaster embryos using lattice light-sheet microscopy. This protocol allows direct observation of both transcription factor diffusion and binding dynamics. Finally, we illustrate how this Drosophila protocol can be extended to other thick samples using single molecule imaging in live mouse embryos as an example.
- Published
- 2018
- Full Text
- View/download PDF
29. Widespread Translational Remodeling during Human Neuronal Differentiation.
- Author
-
Blair JD, Hockemeyer D, Doudna JA, Bateup HS, and Floor SN
- Subjects
- 3' Untranslated Regions, Cell Line, Cells, Cultured, Humans, Neural Stem Cells cytology, Prosencephalon cytology, Prosencephalon metabolism, Protein Biosynthesis, Proteome genetics, Proteome metabolism, TOR Serine-Threonine Kinases genetics, TOR Serine-Threonine Kinases metabolism, Transcriptome, Gene Expression Regulation, Developmental, Neural Stem Cells metabolism, Neurogenesis
- Abstract
Faithful cellular differentiation requires temporally precise activation of gene expression programs, which are coordinated at the transcriptional and translational levels. Neurons express the most complex set of mRNAs of any human tissue, but translational changes during neuronal differentiation remain incompletely understood. Here, we induced forebrain neuronal differentiation of human embryonic stem cells (hESCs) and measured genome-wide RNA and translation levels with transcript-isoform resolution. We found that thousands of genes change translation status during differentiation without a corresponding change in RNA level. Specifically, we identified mTOR signaling as a key driver for elevated translation of translation-related genes in hESCs. In contrast, translational repression in active neurons is mediated by regulatory sequences in 3' UTRs. Together, our findings identify extensive translational control changes during human neuronal differentiation and a crucial role of 3' UTRs in driving cell-type-specific translation., (Copyright © 2017 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
30. Mutations in the promoter of the telomerase gene TERT contribute to tumorigenesis by a two-step mechanism.
- Author
-
Chiba K, Lorbeer FK, Shain AH, McSwiggen DT, Schruf E, Oh A, Ryu J, Darzacq X, Bastian BC, and Hockemeyer D
- Subjects
- Cells, Cultured, Humans, Mutation, Telomere, Telomere Shortening, Carcinogenesis genetics, Genomic Instability genetics, Melanoma genetics, Promoter Regions, Genetic genetics, Skin Neoplasms genetics, Telomerase genetics
- Abstract
TERT promoter mutations (TPMs) are the most common noncoding mutations in cancer. The timing and consequences of TPMs have not been fully established. Here, we show that TPMs acquired at the transition from benign nevus to malignant melanoma do not support telomere maintenance. In vitro experiments revealed that TPMs do not prevent telomere attrition, resulting in cells with critically short and unprotected telomeres. Immortalization by TPMs requires a gradual up-regulation of telomerase, coinciding with telomere fusions. These data suggest that TPMs contribute to tumorigenesis by promoting immortalization and genomic instability in two phases. In an initial phase, TPMs do not prevent bulk telomere shortening but extend cellular life span by healing the shortest telomeres. In the second phase, the critically short telomeres lead to genome instability and telomerase is further up-regulated to sustain cell proliferation., (Copyright © 2017 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2017
- Full Text
- View/download PDF
31. Defined and Scalable Differentiation of Human Oligodendrocyte Precursors from Pluripotent Stem Cells in a 3D Culture System.
- Author
-
Rodrigues GMC, Gaj T, Adil MM, Wahba J, Rao AT, Lorbeer FK, Kulkarni RU, Diogo MM, Cabral JMS, Miller EW, Hockemeyer D, and Schaffer DV
- Subjects
- Animals, Biocompatible Materials chemistry, Brain metabolism, CRISPR-Cas Systems genetics, Cell Culture Techniques, Cell Line, Cellular Reprogramming, Genes, Reporter, Homeobox Protein Nkx-2.2, Homeodomain Proteins genetics, Homeodomain Proteins metabolism, Humans, Mice, Mice, Inbred NOD, Mice, SCID, Nuclear Proteins, Oligodendrocyte Precursor Cells metabolism, Oligodendrocyte Precursor Cells transplantation, Oligodendrocyte Transcription Factor 2 genetics, Oligodendrocyte Transcription Factor 2 metabolism, Pluripotent Stem Cells metabolism, Tissue Scaffolds chemistry, Transcription Factors genetics, Transcription Factors metabolism, Transplantation, Heterologous, Zebrafish Proteins, Cell Differentiation, Oligodendrocyte Precursor Cells cytology, Pluripotent Stem Cells cytology
- Abstract
Oligodendrocyte precursor cells (OPCs) offer considerable potential for the treatment of demyelinating diseases and injuries of the CNS. However, generating large quantities of high-quality OPCs remains a substantial challenge that impedes their therapeutic application. Here, we show that OPCs can be generated from human pluripotent stem cells (hPSCs) in a three-dimensional (3D), scalable, and fully defined thermoresponsive biomaterial system. We used CRISPR/Cas9 to create a NKX2.2-EGFP human embryonic stem cell reporter line that enabled fine-tuning of early OPC specification and identification of conditions that markedly increased the number of OLIG2
+ and NKX2.2+ cells generated from hPSCs. Transplantation of 50-day-old OPCs into the brains of NOD/SCID mice revealed that progenitors generated in 3D without cell selection or purification subsequently engrafted, migrated, and matured into myelinating oligodendrocytes in vivo. These results demonstrate the potential of harnessing lineage reporter lines to develop 3D platforms for rapid and large-scale production of OPCs., (Copyright © 2017 The Author(s). Published by Elsevier Inc. All rights reserved.)- Published
- 2017
- Full Text
- View/download PDF
32. Endogenous Telomerase Reverse Transcriptase N-Terminal Tagging Affects Human Telomerase Function at Telomeres In Vivo.
- Author
-
Chiba K, Vogan JM, Wu RA, Gill MS, Zhang X, Collins K, and Hockemeyer D
- Subjects
- Blotting, Southern, Gene Editing, Genotype, Human Embryonic Stem Cells metabolism, Humans, Phenotype, Repetitive Sequences, Nucleic Acid genetics, Telomere Homeostasis, Epitopes metabolism, Telomerase metabolism, Telomere metabolism
- Abstract
Telomerase action at telomeres is essential for the immortal phenotype of stem cells and the aberrant proliferative potential of cancer cells. Insufficient telomere maintenance can cause stem cell and tissue failure syndromes, while increased telomerase levels are associated with tumorigenesis. Both pathologies can arise from only small perturbation of telomerase function. To analyze telomerase at its low endogenous expression level, we genetically engineered human pluripotent stem cells (hPSCs) to express various N-terminal fusion proteins of the telomerase reverse transcriptase from its endogenous locus. Using this approach, we found that these modifications can perturb telomerase function in hPSCs and cancer cells, resulting in telomere length defects. Biochemical analysis suggests that this defect is multileveled, including changes in expression and activity. These findings highlight the unknown complexity of telomerase structural requirements for expression and function in vivo., (Copyright © 2017 American Society for Microbiology.)
- Published
- 2017
- Full Text
- View/download PDF
33. Minimized human telomerase maintains telomeres and resolves endogenous roles of H/ACA proteins, TCAB1, and Cajal bodies.
- Author
-
Vogan JM, Zhang X, Youmans DT, Regalado SG, Johnson JZ, Hockemeyer D, and Collins K
- Subjects
- Humans, Molecular Chaperones, Coiled Bodies metabolism, Ribonucleoproteins, Small Nuclear metabolism, Telomerase metabolism, Telomere metabolism
- Abstract
We dissected the importance of human telomerase biogenesis and trafficking pathways for telomere maintenance. Biological stability of human telomerase RNA (hTR) relies on H/ACA proteins, but other eukaryotes use other RNP assembly pathways. To investigate additional rationale for human telomerase assembly as H/ACA RNP, we developed a minimized cellular hTR. Remarkably, with only binding sites for telomerase reverse transcriptase (TERT), minimized hTR assembled biologically active enzyme. TERT overexpression was required for cellular interaction with minimized hTR, indicating that H/ACA RNP assembly enhances endogenous hTR-TERT interaction. Telomere maintenance by minimized telomerase was unaffected by the elimination of the telomerase holoenzyme Cajal body chaperone TCAB1 or the Cajal body scaffold protein Coilin. Surprisingly, wild-type hTR also maintained and elongated telomeres in TCAB1 or Coilin knockout cells, with distinct changes in telomerase action. Overall, we elucidate trafficking requirements for telomerase biogenesis and function and expand mechanisms by which altered telomere maintenance engenders human disease., Competing Interests: KC: Reviewing editor, eLife. The other authors declare that no competing interests exist.
- Published
- 2016
- Full Text
- View/download PDF
34. Induced Pluripotent Stem Cells Meet Genome Editing.
- Author
-
Hockemeyer D and Jaenisch R
- Subjects
- CRISPR-Cas Systems genetics, Cellular Reprogramming genetics, Epigenesis, Genetic, Humans, Gene Editing, Genome, Human, Induced Pluripotent Stem Cells cytology, Induced Pluripotent Stem Cells metabolism
- Abstract
It is extremely rare for a single experiment to be so impactful and timely that it shapes and forecasts the experiments of the next decade. Here, we review how two such experiments-the generation of human induced pluripotent stem cells (iPSCs) and the development of CRISPR/Cas9 technology-have fundamentally reshaped our approach to biomedical research, stem cell biology, and human genetics. We will also highlight the previous knowledge that iPSC and CRISPR/Cas9 technologies were built on as this groundwork demonstrated the need for solutions and the benefits that these technologies provided and set the stage for their success., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
35. Human stem cell-based disease modeling: prospects and challenges.
- Author
-
Johnson JZ and Hockemeyer D
- Subjects
- Animals, Cell Differentiation, Genome, Human, Humans, Intestinal Mucosa metabolism, Intestines cytology, Models, Biological, Organoids, Stem Cells cytology, Stem Cells metabolism
- Abstract
Human stem cell-based disease models have great promise to advance our understanding of human disease. These models can be derived from patients with genetic disorders and manipulated with genome editing and myriad differentiation protocols to model pathologies in vitro. However, several challenges have impeded the full potential of stem cell-based in vitro disease modeling. Many genetically predisposed diseases take time to manifest and occur in specific tissue microenvironments, and these parameters are often not adequately modeled using conventional shorter-term monolayer cultures. These challenges must be overcome especially for cases where animal models also incompletely recapitulate the complex pathologies found in humans. As prominent ways to tackle these challenges we discuss here how advanced genome editing tools in human stem cells and human organoid cultures, specifically the example of intestinal organoids, contribute genetically defined models that recapitulate phenotypes of disease., (Copyright © 2015. Published by Elsevier Ltd.)
- Published
- 2015
- Full Text
- View/download PDF
36. Control of telomerase action at human telomeres.
- Author
-
Hockemeyer D and Collins K
- Subjects
- Humans, Models, Biological, Protein Binding, Shelterin Complex, DNA metabolism, Telomerase metabolism, Telomere Homeostasis, Telomere-Binding Proteins metabolism
- Abstract
Recent progress has greatly increased the understanding of telomere-bound shelterin proteins and the telomerase holoenzyme, predominantly as separate complexes. Pioneering studies have begun to investigate the requirements for shelterin-telomerase interaction. From this vantage point, focusing on human cells, we review and discuss models for how telomerase and shelterin subunits coordinate to achieve balanced telomere-length homeostasis.
- Published
- 2015
- Full Text
- View/download PDF
37. Cancer-associated TERT promoter mutations abrogate telomerase silencing.
- Author
-
Chiba K, Johnson JZ, Vogan JM, Wagner T, Boyle JM, and Hockemeyer D
- Subjects
- Cell Differentiation, Cells, Cultured, Humans, Pluripotent Stem Cells physiology, Telomerase genetics, Cell Proliferation, Mutation, Promoter Regions, Genetic, Telomerase biosynthesis
- Abstract
Mutations in the human telomerase reverse transcriptase (TERT) promoter are the most frequent non-coding mutations in cancer, but their molecular mechanism in tumorigenesis has not been established. We used genome editing of human pluripotent stem cells with physiological telomerase expression to elucidate the mechanism by which these mutations contribute to human disease. Surprisingly, telomerase-expressing embryonic stem cells engineered to carry any of the three most frequent TERT promoter mutations showed only a modest increase in TERT transcription with no impact on telomerase activity. However, upon differentiation into somatic cells, which normally silence telomerase, cells with TERT promoter mutations failed to silence TERT expression, resulting in increased telomerase activity and aberrantly long telomeres. Thus, TERT promoter mutations are sufficient to overcome the proliferative barrier imposed by telomere shortening without additional tumor-selected mutations. These data establish that TERT promoter mutations can promote immortalization and tumorigenesis of incipient cancer cells.
- Published
- 2015
- Full Text
- View/download PDF
38. Genome editing in human pluripotent stem cells using site-specific nucleases.
- Author
-
Chiba K and Hockemeyer D
- Subjects
- Cloning, Molecular methods, DNA End-Joining Repair, Electroporation, Embryonic Stem Cells metabolism, Flow Cytometry methods, Gene Expression, Genetic Vectors genetics, Genotyping Techniques, Humans, Induced Pluripotent Stem Cells metabolism, Single-Cell Analysis methods, DNA Restriction Enzymes genetics, DNA Restriction Enzymes metabolism, Gene Targeting, Homologous Recombination, Pluripotent Stem Cells metabolism
- Abstract
Human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) (Thomson, Science 282:1145-1147, 1998; Takahashi et al. Cell 131:861-872, 2007), collectively referred to as pluripotent stem cells (hPSCs), are currently used in disease modeling to address questions specific to humans and to complement our insight gained from model organisms (Soldner et al. Cell 146:318-331, 2011; Soldner and Jaenisch, Science 338:1155-1156, 2012). Recently, genetic engineering using site-specific nucleases has been established in hPSCs (Hockemeyer et al. Nat Biotechnol 27:851-857, 2009; Hockemeyer et al., Nat Biotechnol 29:731-734, 2011; Zou et al., Cell Stem Cell 5:97-110, 2011; Yusa et al., Nature 478:391-394, 2011; DeKelver et al., Genome Res 20:1133-1142, 2010), allowing a level of genetic control previously limited to model systems. Thus, we can now perform targeted gene knockouts, generate tissue-specific cell lineage reporters, overexpress genes from a defined locus, and introduce and repair single point mutations in hPSCs. This ability to genetically engineer pluripotent stem cells will significantly facilitate the study of human disease in a defined genetic context. Here we outline protocols for efficient gene targeting in hPSCs.
- Published
- 2015
- Full Text
- View/download PDF
39. TALEN gene knockouts reveal no requirement for the conserved human shelterin protein Rap1 in telomere protection and length regulation.
- Author
-
Kabir S, Hockemeyer D, and de Lange T
- Subjects
- Animals, Cell Proliferation, Chromatin metabolism, Gene Expression Regulation, Humans, Mice, Shelterin Complex, Telomere-Binding Proteins deficiency, Telomere-Binding Proteins metabolism, Transcription, Genetic, Conserved Sequence, Endonucleases metabolism, Gene Knockout Techniques, Telomere metabolism, Telomere Homeostasis genetics, Telomere-Binding Proteins genetics, Trans-Activators metabolism
- Abstract
The conserved protein Rap1 functions at telomeres in fungi, protozoa, and vertebrates. Like yeast Rap1, human Rap1 has been implicated in telomere length regulation and repression of nonhomologous end-joining (NHEJ) at telomeres. However, mouse telomeres lacking Rap1 do not succumb to NHEJ. To determine the functions of human Rap1, we generated several transcription activator-like effector nuclease (TALEN)-mediated human cell lines lacking Rap1. Loss of Rap1 did not affect the other components of shelterin, the modification of telomeric histones, the subnuclear position of telomeres, or the 3' telomeric overhang. Telomeres lacking Rap1 did not show a DNA damage response, NHEJ, or consistent changes in their length, indicating that Rap1 does not have an important function in protection or length regulation of human telomeres. As human Rap1, like its mouse and unicellular orthologs, affects gene expression, we propose that the conservation of Rap1 reflects its role in transcriptional regulation rather than a function at telomeres., (Copyright © 2014 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
40. Genetic and molecular identification of three human TPP1 functions in telomerase action: recruitment, activation, and homeostasis set point regulation.
- Author
-
Sexton AN, Regalado SG, Lai CS, Cost GJ, O'Neil CM, Urnov FD, Gregory PD, Jaenisch R, Collins K, and Hockemeyer D
- Subjects
- Embryonic Stem Cells, Enzyme Activation genetics, Gene Knockout Techniques, Genetic Complementation Test, Humans, Proto-Oncogene Proteins c-ets genetics, Proto-Oncogene Proteins c-ets metabolism, Repressor Proteins genetics, Repressor Proteins metabolism, Shelterin Complex, Telomerase genetics, Telomere genetics, Telomere-Binding Proteins genetics, Telomere-Binding Proteins metabolism, ETS Translocation Variant 6 Protein, Telomerase metabolism, Telomere enzymology, Telomere Homeostasis genetics
- Abstract
Telomere length homeostasis is essential for the long-term survival of stem cells, and its set point determines the proliferative capacity of differentiated cell lineages by restricting the reservoir of telomeric repeats. Knockdown and overexpression studies in human tumor cells showed that the shelterin subunit TPP1 recruits telomerase to telomeres through a region termed the TEL patch. However, these studies do not resolve whether the TPP1 TEL patch is the only mechanism for telomerase recruitment and whether telomerase regulation studied in tumor cells is representative of nontransformed cells such as stem cells. Using genome engineering of human embryonic stem cells, which have physiological telomere length homeostasis, we establish that the TPP1 TEL patch is genetically essential for telomere elongation and thus long-term cell viability. Furthermore, genetic bypass, protein fusion, and intragenic complementation assays define two distinct additional mechanisms of TPP1 involvement in telomerase action at telomeres. We demonstrate that TPP1 provides an essential step of telomerase activation as well as feedback regulation of telomerase by telomere length, which is necessary to determine the appropriate telomere length set point in human embryonic stem cells. These studies reveal and resolve multiple TPP1 roles in telomere elongation and stem cell telomere length homeostasis., (© 2014 Sexton et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2014
- Full Text
- View/download PDF
41. Human intestinal tissue with adult stem cell properties derived from pluripotent stem cells.
- Author
-
Forster R, Chiba K, Schaeffer L, Regalado SG, Lai CS, Gao Q, Kiani S, Farin HF, Clevers H, Cost GJ, Chan A, Rebar EJ, Urnov FD, Gregory PD, Pachter L, Jaenisch R, and Hockemeyer D
- Subjects
- Cells, Cultured, Humans, Receptors, G-Protein-Coupled metabolism, Adult Stem Cells cytology, Adult Stem Cells metabolism, Gene Expression Profiling methods, Intestines cytology, Pluripotent Stem Cells cytology, Pluripotent Stem Cells metabolism
- Abstract
Genetically engineered human pluripotent stem cells (hPSCs) have been proposed as a source for transplantation therapies and are rapidly becoming valuable tools for human disease modeling. However, many applications are limited due to the lack of robust differentiation paradigms that allow for the isolation of defined functional tissues. Here, using an endogenous LGR5-GFP reporter, we derived adult stem cells from hPSCs that gave rise to functional human intestinal tissue comprising all major cell types of the intestine. Histological and functional analyses revealed that such human organoid cultures could be derived with high purity and with a composition and morphology similar to those of cultures obtained from human biopsies. Importantly, hPSC-derived organoids responded to the canonical signaling pathways that control self-renewal and differentiation in the adult human intestinal stem cell compartment. This adult stem cell system provides a platform for studying human intestinal disease in vitro using genetically engineered hPSCs.
- Published
- 2014
- Full Text
- View/download PDF
42. Genome editing 101: let's go digital.
- Author
-
Forster R and Hockemeyer D
- Subjects
- Humans, Cytological Techniques methods, Genome, Human, Induced Pluripotent Stem Cells cytology
- Published
- 2014
- Full Text
- View/download PDF
43. Global transcriptional and translational repression in human-embryonic-stem-cell-derived Rett syndrome neurons.
- Author
-
Li Y, Wang H, Muffat J, Cheng AW, Orlando DA, Lovén J, Kwok SM, Feldman DA, Bateup HS, Gao Q, Hockemeyer D, Mitalipova M, Lewis CA, Vander Heiden MG, Sur M, Young RA, and Jaenisch R
- Subjects
- Cells, Cultured, Embryonic Stem Cells metabolism, Humans, Mutation, Neurons metabolism, Embryonic Stem Cells pathology, Methyl-CpG-Binding Protein 2 metabolism, Neurons pathology, Protein Biosynthesis genetics, Rett Syndrome genetics, Rett Syndrome pathology, Transcription, Genetic genetics
- Abstract
Rett syndrome (RTT) is caused by mutations of MECP2, a methyl CpG binding protein thought to act as a global transcriptional repressor. Here we show, using an isogenic human embryonic stem cell model of RTT, that MECP2 mutant neurons display key molecular and cellular features of this disorder. Unbiased global gene expression analyses demonstrate that MECP2 functions as a global activator in neurons but not in neural precursors. Decreased transcription in neurons was coupled with a significant reduction in nascent protein synthesis and lack of MECP2 was manifested as a severe defect in the activity of the AKT/mTOR pathway. Lack of MECP2 also leads to impaired mitochondrial function in mutant neurons. Activation of AKT/mTOR signaling by exogenous growth factors or by depletion of PTEN boosted protein synthesis and ameliorated disease phenotypes in mutant neurons. Our findings indicate a vital function for MECP2 in maintaining active gene transcription in human neuronal cells., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
44. Deletion of the de novo DNA methyltransferase Dnmt3a promotes lung tumor progression.
- Author
-
Gao Q, Steine EJ, Barrasa MI, Hockemeyer D, Pawlak M, Fu D, Reddy S, Bell GW, and Jaenisch R
- Subjects
- Animals, Base Sequence, Cell Division, DNA Methylation, DNA Methyltransferase 3A, DNA Primers, Disease Models, Animal, Disease Progression, Gene Silencing, Lung Neoplasms enzymology, Lung Neoplasms genetics, Mice, Mice, Knockout, Oligonucleotide Array Sequence Analysis, Real-Time Polymerase Chain Reaction, DNA (Cytosine-5-)-Methyltransferases genetics, Lung Neoplasms pathology
- Abstract
Alterations in DNA methylation have been associated with genome-wide hypomethylation and regional de novo methylation in numerous cancers. De novo methylation is mediated by the de novo methyltransferases Dnmt3a and 3b, but only Dnmt3b has been implicated in promoting cancer by silencing of tumor-suppressor genes. In this study, we have analyzed the role of Dnmt3a in lung cancer by using a conditional mouse tumor model. We show that Dnmt3a deficiency significantly promotes tumor growth and progression but not initiation. Changes in gene expression show that Dnmt3a deficiency affects key steps in cancer progression, such as angiogenesis, cell adhesion, and cell motion, consistent with accelerated and more malignant growth. Our results suggest that Dnmt3a may act like a tumor-suppressor gene in lung tumor progression and may be a critical determinant of lung cancer malignancy.
- Published
- 2011
- Full Text
- View/download PDF
45. Generation of isogenic pluripotent stem cells differing exclusively at two early onset Parkinson point mutations.
- Author
-
Soldner F, Laganière J, Cheng AW, Hockemeyer D, Gao Q, Alagappan R, Khurana V, Golbe LI, Myers RH, Lindquist S, Zhang L, Guschin D, Fong LK, Vu BJ, Meng X, Urnov FD, Rebar EJ, Gregory PD, Zhang HS, and Jaenisch R
- Subjects
- Cell Line, Embryonic Stem Cells, Genetic Engineering, Genome-Wide Association Study, Humans, Mutagenesis, Oligonucleotides metabolism, alpha-Synuclein genetics, Parkinson Disease pathology, Pluripotent Stem Cells, Point Mutation
- Abstract
Patient-specific induced pluripotent stem cells (iPSCs) derived from somatic cells provide a unique tool for the study of human disease, as well as a promising source for cell replacement therapies. One crucial limitation has been the inability to perform experiments under genetically defined conditions. This is particularly relevant for late age onset disorders in which in vitro phenotypes are predicted to be subtle and susceptible to significant effects of genetic background variations. By combining zinc finger nuclease (ZFN)-mediated genome editing and iPSC technology, we provide a generally applicable solution to this problem, generating sets of isogenic disease and control human pluripotent stem cells that differ exclusively at either of two susceptibility variants for Parkinson's disease by modifying the underlying point mutations in the α-synuclein gene. The robust capability to genetically correct disease-causing point mutations in patient-derived hiPSCs represents significant progress for basic biomedical research and an advance toward hiPSC-based cell replacement therapies., (Copyright © 2011 Elsevier Inc. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
46. Genetic engineering of human pluripotent cells using TALE nucleases.
- Author
-
Hockemeyer D, Wang H, Kiani S, Lai CS, Gao Q, Cassady JP, Cost GJ, Zhang L, Santiago Y, Miller JC, Zeitler B, Cherone JM, Meng X, Hinkley SJ, Rebar EJ, Gregory PD, Urnov FD, and Jaenisch R
- Subjects
- Base Sequence, Endonucleases genetics, Homeodomain Proteins genetics, Humans, Molecular Sequence Data, Myosin-Light-Chain Phosphatase genetics, Octamer Transcription Factor-3 genetics, Transcription Factors genetics, Zinc Fingers, Embryonic Stem Cells physiology, Endonucleases metabolism, Gene Targeting methods, Genetic Engineering methods, Induced Pluripotent Stem Cells physiology, Transcription Factors metabolism
- Abstract
Targeted genetic engineering of human pluripotent cells is a prerequisite for exploiting their full potential. Such genetic manipulations can be achieved using site-specific nucleases. Here we engineered transcription activator-like effector nucleases (TALENs) for five distinct genomic loci. At all loci tested we obtained human embryonic stem cell (ESC) and induced pluripotent stem cell (iPSC) clones carrying transgenic cassettes solely at the TALEN-specified location. Our data suggest that TALENs employing the specific architectures described here mediate site-specific genome modification in human pluripotent cells with similar efficiency and precision as do zinc-finger nucleases (ZFNs).
- Published
- 2011
- Full Text
- View/download PDF
47. Transgene excision has no impact on in vivo integration of human iPS derived neural precursors.
- Author
-
Major T, Menon J, Auyeung G, Soldner F, Hockemeyer D, Jaenisch R, and Tabar V
- Subjects
- Animals, Astrocytes cytology, Astrocytes metabolism, Cell Movement, Cells, Cultured, Corpus Striatum surgery, Doublecortin Domain Proteins, Female, Glial Fibrillary Acidic Protein analysis, Humans, Immunohistochemistry, Induced Pluripotent Stem Cells metabolism, Microtubule-Associated Proteins analysis, Neural Stem Cells metabolism, Neurons cytology, Neurons metabolism, Neuropeptides analysis, Oligodendroglia cytology, Oligodendroglia metabolism, Rats, Rats, Sprague-Dawley, Transgenes genetics, Transplantation, Heterologous, Tubulin analysis, Cell Differentiation, Induced Pluripotent Stem Cells cytology, Neural Stem Cells cytology, Stem Cell Transplantation methods
- Abstract
The derivation of induced human pluripotent stem cells (hiPS) has generated significant enthusiasm particularly for the prospects of cell-based therapy. But there are concerns about the suitability of iPS cells for in vivo applications due in part to the introduction of potentially oncogenic transcription factors via viral vectors. Recently developed lentiviral vectors allow the excision of viral reprogramming factors and the development of transgene-free iPS lines. However it is unclear if reprogramming strategy has an impact on the differentiation potential and the in vivo behavior of hiPS progeny. Here we subject viral factor-free, c-myc-free and conventionally reprogrammed four-factor human iPS lines to a further challenge, by analyzing their differentiation potential along the 3 neural lineages and over extended periods of time in vitro, as well as by interrogating their ability to respond to local environmental cues by grafting into the striatum. We demonstrate similar and efficient differentiation into neurons, astrocytes and oligodendrocytes among all hiPS and human ES line controls. Upon intracranial grafting in the normal rat (Sprague Dawley), precursors derived from all hiPS lines exhibited good survival and response to environmental cues by integrating into the subventricular zone, acquiring phenotypes typical of type A, B or C cells and migrating along the rostral migratory stream into the olfactory bulb. There was no teratoma or other tumor formation 12 weeks after grafting in any of the 26 animals used in the study. Thus neither factor excision nor persistence of c-myc impact the behavior of hiPS lines in vivo.
- Published
- 2011
- Full Text
- View/download PDF
48. Differentiated Parkinson patient-derived induced pluripotent stem cells grow in the adult rodent brain and reduce motor asymmetry in Parkinsonian rats.
- Author
-
Hargus G, Cooper O, Deleidi M, Levy A, Lee K, Marlow E, Yow A, Soldner F, Hockemeyer D, Hallett PJ, Osborn T, Jaenisch R, and Isacson O
- Subjects
- Animals, Humans, Parkinson Disease pathology, Rats, Parkinson Disease surgery, Pluripotent Stem Cells cytology
- Abstract
Recent advances in deriving induced pluripotent stem (iPS) cells from patients offer new possibilities for biomedical research and clinical applications, as these cells could be used for autologous transplantation. We differentiated iPS cells from patients with Parkinson's disease (PD) into dopaminergic (DA) neurons and show that these DA neurons can be transplanted without signs of neurodegeneration into the adult rodent striatum. The PD patient iPS (PDiPS) cell-derived DA neurons survived at high numbers, showed arborization, and mediated functional effects in an animal model of PD as determined by reduction of amphetamine- and apomorphine-induced rotational asymmetry, but only a few DA neurons projected into the host striatum at 16 wk after transplantation. We next applied FACS for the neural cell adhesion molecule NCAM on differentiated PDiPS cells before transplantation, which resulted in surviving DA neurons with functional effects on amphetamine-induced rotational asymmetry in a 6-OHDA animal model of PD. Morphologically, we found that PDiPS cell-derived non-DA neurons send axons along white matter tracts into specific close and remote gray matter target areas in the adult brain. Such findings establish the transplantation of human PDiPS cell-derived neurons as a long-term in vivo method to analyze potential disease-related changes in a physiological context. Our data also demonstrate proof of principle of survival and functional effects of PDiPS cell-derived DA neurons in an animal model of PD and encourage further development of differentiation protocols to enhance growth and function of implanted PDiPS cell-derived DA neurons in regard to potential therapeutic applications.
- Published
- 2010
- Full Text
- View/download PDF
49. Chromatin structure and gene expression programs of human embryonic and induced pluripotent stem cells.
- Author
-
Guenther MG, Frampton GM, Soldner F, Hockemeyer D, Mitalipova M, Jaenisch R, and Young RA
- Subjects
- Cell Line, Chromatin Assembly and Disassembly genetics, Cluster Analysis, Genome, Human genetics, Histones metabolism, Humans, Chromatin chemistry, Chromatin genetics, Embryonic Stem Cells metabolism, Gene Expression Regulation, Induced Pluripotent Stem Cells metabolism
- Abstract
Knowledge of both the global chromatin structure and the gene expression programs of human embryonic stem cells (ESCs) and induced pluripotent stem cells (iPSCs) should provide a robust means to assess whether the genomes of these cells have similar pluripotent states. Recent studies have suggested that ESCs and iPSCs represent different pluripotent states with substantially different gene expression profiles. We describe here a comparison of global chromatin structure and gene expression data for a panel of human ESCs and iPSCs. Genome-wide maps of nucleosomes with histone H3K4me3 and H3K27me3 modifications indicate that there is little difference between ESCs and iPSCs with respect to these marks. Gene expression profiles confirm that the transcriptional programs of ESCs and iPSCs show very few consistent differences. Although some variation in chromatin structure and gene expression was observed in these cell lines, these variations did not serve to distinguish ESCs from iPSCs., (Copyright 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
50. Functional genomics, proteomics, and regulatory DNA analysis in isogenic settings using zinc finger nuclease-driven transgenesis into a safe harbor locus in the human genome.
- Author
-
DeKelver RC, Choi VM, Moehle EA, Paschon DE, Hockemeyer D, Meijsing SH, Sancak Y, Cui X, Steine EJ, Miller JC, Tam P, Bartsevich VV, Meng X, Rupniewski I, Gopalan SM, Sun HC, Pitz KJ, Rock JM, Zhang L, Davis GD, Rebar EJ, Cheeseman IM, Yamamoto KR, Sabatini DM, Jaenisch R, Gregory PD, and Urnov FD
- Subjects
- Base Sequence, Cell Line, Endonucleases genetics, Genetic Loci, Humans, Molecular Sequence Data, Proteomics methods, Gene Transfer Techniques, Genome, Human, Genomics methods, Regulatory Sequences, Nucleic Acid genetics, Zinc Fingers genetics
- Abstract
Isogenic settings are routine in model organisms, yet remain elusive for genetic experiments on human cells. We describe the use of designed zinc finger nucleases (ZFNs) for efficient transgenesis without drug selection into the PPP1R12C gene, a "safe harbor" locus known as AAVS1. ZFNs enable targeted transgenesis at a frequency of up to 15% following transient transfection of both transformed and primary human cells, including fibroblasts and hES cells. When added to this locus, transgenes such as expression cassettes for shRNAs, small-molecule-responsive cDNA expression cassettes, and reporter constructs, exhibit consistent expression and sustained function over 50 cell generations. By avoiding random integration and drug selection, this method allows bona fide isogenic settings for high-throughput functional genomics, proteomics, and regulatory DNA analysis in essentially any transformed human cell type and in primary cells.
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.