360 results on '"Conklin Bruce R"'
Search Results
2. Transcription factor protein interactomes reveal genetic determinants in heart disease
- Author
-
Gonzalez-Teran, Barbara, Pittman, Maureen, Felix, Franco, Thomas, Reuben, Richmond-Buccola, Desmond, Hüttenhain, Ruth, Choudhary, Krishna, Moroni, Elisabetta, Costa, Mauro W, Huang, Yu, Padmanabhan, Arun, Alexanian, Michael, Lee, Clara Youngna, Maven, Bonnie EJ, Samse-Knapp, Kaitlen, Morton, Sarah U, McGregor, Michael, Gifford, Casey A, Seidman, JG, Seidman, Christine E, Gelb, Bruce D, Colombo, Giorgio, Conklin, Bruce R, Black, Brian L, Bruneau, Benoit G, Krogan, Nevan J, Pollard, Katherine S, and Srivastava, Deepak
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Biomedical and Clinical Sciences ,Genetics ,Cardiovascular Medicine and Haematology ,Clinical Sciences ,Stem Cell Research ,Biotechnology ,Heart Disease - Coronary Heart Disease ,Pediatric ,Cardiovascular ,Heart Disease ,Congenital Structural Anomalies ,Stem Cell Research - Embryonic - Human ,Aetiology ,2.1 Biological and endogenous factors ,Animals ,GATA4 Transcription Factor ,Heart Defects ,Congenital ,Mice ,Mutation ,Nuclear Proteins ,Oxidoreductases ,Proteomics ,T-Box Domain Proteins ,Transcription Factors ,GATA4 ,GLYR1 ,NPAC ,TBX5 ,congenital heart disease ,de novo variants ,disease variants ,genetics ,protein interactome networks ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
Congenital heart disease (CHD) is present in 1% of live births, yet identification of causal mutations remains challenging. We hypothesized that genetic determinants for CHDs may lie in the protein interactomes of transcription factors whose mutations cause CHDs. Defining the interactomes of two transcription factors haplo-insufficient in CHD, GATA4 and TBX5, within human cardiac progenitors, and integrating the results with nearly 9,000 exomes from proband-parent trios revealed an enrichment of de novo missense variants associated with CHD within the interactomes. Scoring variants of interactome members based on residue, gene, and proband features identified likely CHD-causing genes, including the epigenetic reader GLYR1. GLYR1 and GATA4 widely co-occupied and co-activated cardiac developmental genes, and the identified GLYR1 missense variant disrupted interaction with GATA4, impairing in vitro and in vivo function in mice. This integrative proteomic and genetic approach provides a framework for prioritizing and interrogating genetic variants in heart disease.
- Published
- 2022
3. Functional analysis of a common BAG3 allele associated with protection from heart failure
- Author
-
Perez-Bermejo, Juan A., Judge, Luke M., Jensen, Christina L., Wu, Kenneth, Watry, Hannah L., Truong, Annie, Ho, Jaclyn J., Carter, Matthew, Runyon, Wendy V., Kaake, Robyn M., Pulido, Ernst H., Mandegar, Mohammad A., Swaney, Danielle L., So, Po-Lin, Krogan, Nevan J., and Conklin, Bruce R.
- Published
- 2023
- Full Text
- View/download PDF
4. Give heart cells a beat: An interactive museum exhibit that synchronizes stem cell–derived cardiomyocytes to visitors’ heartbeat
- Author
-
Perez-Bermejo, Juan A., Reisman, Samuel J., Ma, Joyce, Carrison-Stone, Dana, Cerrito, Chris, Ribeiro, Alexandre J.S., Conklin, Bruce R., and Yu, Kristina
- Published
- 2024
- Full Text
- View/download PDF
5. BRD2 inhibition blocks SARS-CoV-2 infection by reducing transcription of the host cell receptor ACE2
- Author
-
Samelson, Avi J, Tran, Quang Dinh, Robinot, Rémy, Carrau, Lucia, Rezelj, Veronica V, Kain, Alice Mac, Chen, Merissa, Ramadoss, Gokul N, Guo, Xiaoyan, Lim, Shion A, Lui, Irene, Nuñez, James K, Rockwood, Sarah J, Wang, Jianhui, Liu, Na, Carlson-Stevermer, Jared, Oki, Jennifer, Maures, Travis, Holden, Kevin, Weissman, Jonathan S, Wells, James A, Conklin, Bruce R, TenOever, Benjamin R, Chakrabarti, Lisa A, Vignuzzi, Marco, Tian, Ruilin, and Kampmann, Martin
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Pneumonia ,Emerging Infectious Diseases ,Cancer ,Biodefense ,Vaccine Related ,Infectious Diseases ,Prevention ,Lung ,2.1 Biological and endogenous factors ,5.1 Pharmaceuticals ,Development of treatments and therapeutic interventions ,Aetiology ,Infection ,Good Health and Well Being ,Angiotensin-Converting Enzyme 2 ,Antiviral Agents ,COVID-19 ,Cell Line ,Epithelial Cells ,Humans ,Membrane Glycoproteins ,SARS-CoV-2 ,Transcription Factors ,COVID-19 Drug Treatment ,Medical and Health Sciences ,Developmental Biology ,Biochemistry and cell biology - Abstract
SARS-CoV-2 infection of human cells is initiated by the binding of the viral Spike protein to its cell-surface receptor ACE2. We conducted a targeted CRISPRi screen to uncover druggable pathways controlling Spike protein binding to human cells. Here we show that the protein BRD2 is required for ACE2 transcription in human lung epithelial cells and cardiomyocytes, and BRD2 inhibitors currently evaluated in clinical trials potently block endogenous ACE2 expression and SARS-CoV-2 infection of human cells, including those of human nasal epithelia. Moreover, pharmacological BRD2 inhibition with the drug ABBV-744 inhibited SARS-CoV-2 replication in Syrian hamsters. We also found that BRD2 controls transcription of several other genes induced upon SARS-CoV-2 infection, including the interferon response, which in turn regulates the antiviral response. Together, our results pinpoint BRD2 as a potent and essential regulator of the host response to SARS-CoV-2 infection and highlight the potential of BRD2 as a therapeutic target for COVID-19.
- Published
- 2022
6. SARS-CoV-2 infection of human iPSC-derived cardiac cells reflects cytopathic features in hearts of patients with COVID-19.
- Author
-
Perez-Bermejo, Juan A, Kang, Serah, Rockwood, Sarah J, Simoneau, Camille R, Joy, David A, Silva, Ana C, Ramadoss, Gokul N, Flanigan, Will R, Fozouni, Parinaz, Li, Huihui, Chen, Pei-Yi, Nakamura, Ken, Whitman, Jeffrey D, Hanson, Paul J, McManus, Bruce M, Ott, Melanie, Conklin, Bruce R, and McDevitt, Todd C
- Subjects
Myocardium ,Heart ,Cells ,Cultured ,Myocytes ,Cardiac ,Humans ,Autopsy ,Induced Pluripotent Stem Cells ,Transcriptome ,COVID-19 ,SARS-CoV-2 ,Cardiovascular ,Pneumonia ,Infectious Diseases ,Stem Cell Research ,Heart Disease ,Emerging Infectious Diseases ,Lung ,Clinical Research ,Stem Cell Research - Induced Pluripotent Stem Cell ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,2.1 Biological and endogenous factors ,Biological Sciences ,Medical and Health Sciences - Abstract
Although coronavirus disease 2019 (COVID-19) causes cardiac dysfunction in up to 25% of patients, its pathogenesis remains unclear. Exposure of human induced pluripotent stem cell (iPSC)-derived heart cells to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) revealed productive infection and robust transcriptomic and morphological signatures of damage, particularly in cardiomyocytes. Transcriptomic disruption of structural genes corroborates adverse morphologic features, which included a distinct pattern of myofibrillar fragmentation and nuclear disruption. Human autopsy specimens from patients with COVID-19 reflected similar alterations, particularly sarcomeric fragmentation. These notable cytopathic features in cardiomyocytes provide insights into SARS-CoV-2-induced cardiac damage, offer a platform for discovery of potential therapeutics, and raise concerns about the long-term consequences of COVID-19 in asymptomatic and severe cases.
- Published
- 2021
7. Gain-of-function cardiomyopathic mutations in RBM20 rewire splicing regulation and re-distribute ribonucleoprotein granules within processing bodies
- Author
-
Fenix, Aidan M, Miyaoka, Yuichiro, Bertero, Alessandro, Blue, Steven M, Spindler, Matthew J, Tan, Kenneth KB, Perez-Bermejo, Juan A, Chan, Amanda H, Mayerl, Steven J, Nguyen, Trieu D, Russell, Caitlin R, Lizarraga, Paweena P, Truong, Annie, So, Po-Lin, Kulkarni, Aishwarya, Chetal, Kashish, Sathe, Shashank, Sniadecki, Nathan J, Yeo, Gene W, Murry, Charles E, Conklin, Bruce R, and Salomonis, Nathan
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Rare Diseases ,Heart Disease ,Genetics ,Cardiovascular ,Aetiology ,2.1 Biological and endogenous factors ,Cardiomyopathies ,Cardiomyopathy ,Dilated ,DEAD-box RNA Helicases ,DNA Helicases ,Gain of Function Mutation ,Gene Knockdown Techniques ,Humans ,Induced Pluripotent Stem Cells ,Male ,Mutation ,Mutation ,Missense ,Poly-ADP-Ribose Binding Proteins ,Proto-Oncogene Proteins ,RNA Helicases ,RNA Recognition Motif Proteins ,RNA Splicing ,RNA-Binding Proteins ,Ribonucleoproteins - Abstract
Mutations in the cardiac splicing factor RBM20 lead to malignant dilated cardiomyopathy (DCM). To understand the mechanism of RBM20-associated DCM, we engineered isogenic iPSCs with DCM-associated missense mutations in RBM20 as well as RBM20 knockout (KO) iPSCs. iPSC-derived engineered heart tissues made from these cell lines recapitulate contractile dysfunction of RBM20-associated DCM and reveal greater dysfunction with missense mutations than KO. Analysis of RBM20 RNA binding by eCLIP reveals a gain-of-function preference of mutant RBM20 for 3' UTR sequences that are shared with amyotrophic lateral sclerosis (ALS) and processing-body associated RNA binding proteins (FUS, DDX6). Deep RNA sequencing reveals that the RBM20 R636S mutant has unique gene, splicing, polyadenylation and circular RNA defects that differ from RBM20 KO. Super-resolution microscopy verifies that mutant RBM20 maintains very limited nuclear localization potential; rather, the mutant protein associates with cytoplasmic processing bodies (DDX6) under basal conditions, and with stress granules (G3BP1) following acute stress. Taken together, our results highlight a pathogenic mechanism in cardiac disease through splicing-dependent and -independent pathways.
- Published
- 2021
8. SARS-CoV-2 infection of human iPSC-derived cardiac cells predicts novel cytopathic features in hearts of COVID-19 patients.
- Author
-
Pérez-Bermejo, Juan A, Kang, Serah, Rockwood, Sarah J, Simoneau, Camille R, Joy, David A, Ramadoss, Gokul N, Silva, Ana C, Flanigan, Will R, Li, Huihui, Nakamura, Ken, Whitman, Jeffrey D, Ott, Melanie, Conklin, Bruce R, and McDevitt, Todd C
- Subjects
Cardiovascular ,Stem Cell Research ,Emerging Infectious Diseases ,Infectious Diseases ,Clinical Research ,Heart Disease ,2.1 Biological and endogenous factors ,Aetiology - Abstract
Although COVID-19 causes cardiac dysfunction in up to 25% of patients, its pathogenesis remains unclear. Exposure of human iPSC-derived heart cells to SARS-CoV-2 revealed productive infection and robust transcriptomic and morphological signatures of damage, particularly in cardiomyocytes. Transcriptomic disruption of structural proteins corroborated adverse morphologic features, which included a distinct pattern of myofibrillar fragmentation and numerous iPSC-cardiomyocytes lacking nuclear DNA. Human autopsy specimens from COVID-19 patients displayed similar sarcomeric disruption, as well as cardiomyocytes without DNA staining. These striking cytopathic features provide new insights into SARS-CoV-2 induced cardiac damage, offer a platform for discovery of potential therapeutics, and raise serious concerns about the long-term consequences of COVID-19.
- Published
- 2020
9. Rapid, precise quantification of large DNA excisions and inversions by ddPCR.
- Author
-
Watry, Hannah L, Feliciano, Carissa M, Gjoni, Ketrin, Takahashi, Gou, Miyaoka, Yuichiro, Conklin, Bruce R, and Judge, Luke M
- Abstract
The excision of genomic sequences using paired CRISPR-Cas nucleases is a powerful tool to study gene function, create disease models and holds promise for therapeutic gene editing. However, our understanding of the factors that favor efficient excision is limited by the lack of a rapid, accurate measurement of DNA excision outcomes that is free of amplification bias. Here, we introduce ddXR (droplet digital PCR eXcision Reporter), a method that enables the accurate and sensitive detection of excisions and inversions independent of length. The method can be completed in a few hours without the need for next-generation sequencing. The ddXR method uncovered unexpectedly high rates of large (> 20 kb) excisions and inversions, while also revealing a surprisingly low dependence on linear distance, up to 170 kb. We further modified the method to measure precise repair of excision junctions and allele-specific excision, with important implications for disease modeling and therapeutic gene editing.
- Published
- 2020
10. Critical Roles of Translation Initiation and RNA Uridylation in Endogenous Retroviral Expression and Neural Differentiation in Pluripotent Stem Cells
- Author
-
Takahashi, Kazutoshi, Jeong, Daeun, Wang, Songnan, Narita, Megumi, Jin, Xuemei, Iwasaki, Mio, Perli, Samuel D, Conklin, Bruce R, and Yamanaka, Shinya
- Subjects
Regenerative Medicine ,Stem Cell Research - Embryonic - Human ,Genetics ,Stem Cell Research ,Underpinning research ,1.1 Normal biological development and functioning ,Generic health relevance ,Animals ,Arylamine N-Acetyltransferase ,Cell Differentiation ,Cell Line ,Cell Lineage ,Cell Self Renewal ,Endogenous Retroviruses ,Gene Editing ,Humans ,Isoenzymes ,Mice ,Neurons ,Peptide Chain Initiation ,Translational ,Pluripotent Stem Cells ,RNA Interference ,RNA Nucleotidyltransferases ,RNA ,Small Interfering ,RNA ,Viral ,Transcription Factors ,endogenous retrovirus ,neural differentiation ,pluripotency ,translation ,Biochemistry and Cell Biology ,Medical Physiology - Abstract
Previous studies have suggested that the loss of the translation initiation factor eIF4G1 homolog NAT1 induces excessive self-renewability of naive pluripotent stem cells (PSCs); yet the role of NAT1 in the self-renewal and differentiation of primed PSCs is still unclear. Here, we generate a conditional knockout of NAT1 in primed PSCs and use the cells for the functional analyses of NAT1. Our results show that NAT1 is required for the self-renewal and neural differentiation of primed PSCs. In contrast, NAT1 deficiency in naive pluripotency attenuates the differentiation to all cell types. We also find that NAT1 is involved in efficient protein expression of an RNA uridyltransferase, TUT7. TUT7 is involved in the neural differentiation of primed PSCs via the regulation of human endogenous retrovirus accumulation. These data demonstrate the essential roles of NAT1 and TUT7 in the precise transition of stem cell fate.
- Published
- 2020
11. CRISPR off-target detection with DISCOVER-seq
- Author
-
Wienert, Beeke, Wyman, Stacia K, Yeh, Charles D, Conklin, Bruce R, and Corn, Jacob E
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Genetics ,Cancer Genomics ,Cancer ,Biotechnology ,Human Genome ,Animals ,CRISPR-Cas Systems ,Clustered Regularly Interspaced Short Palindromic Repeats ,DNA Breaks ,Double-Stranded ,DNA Repair ,Gene Editing ,Humans ,K562 Cells ,Mice ,Sequence Analysis ,DNA ,Chemical Sciences ,Medical and Health Sciences ,Bioinformatics - Abstract
DISCOVER-seq (discovery of in situ Cas off-targets and verification by sequencing) is a broadly applicable approach for unbiased CRISPR-Cas off-target identification in cells and tissues. It leverages the recruitment of DNA repair factors to double-strand breaks (DSBs) after genome editing with CRISPR nucleases. Here, we describe a detailed experimental protocol and analysis pipeline with which to perform DISCOVER-seq. The principle of this method is to track the precise recruitment of MRE11 to DSBs by chromatin immunoprecipitation followed by next-generation sequencing. A customized open-source bioinformatics pipeline, BLENDER (blunt end finder), then identifies off-target sequences genome wide. DISCOVER-seq is capable of finding and measuring off-targets in primary cells and in situ. The two main advantages of DISCOVER-seq are (i) low false-positive rates because DNA repair enzyme binding is required for genome edits to occur and (ii) its applicability to a wide variety of systems, including patient-derived cells and animal models. The whole protocol, including the analysis, can be completed within 2 weeks.
- Published
- 2020
12. Timed inhibition of CDC7 increases CRISPR-Cas9 mediated templated repair.
- Author
-
Wienert, Beeke, Nguyen, David N, Guenther, Alexis, Feng, Sharon J, Locke, Melissa N, Wyman, Stacia K, Shin, Jiyung, Kazane, Katelynn R, Gregory, Georgia L, Carter, Matthew AM, Wright, Francis, Conklin, Bruce R, Marson, Alex, Richardson, Chris D, and Corn, Jacob E
- Subjects
HCT116 Cells ,Hela Cells ,K562 Cells ,Humans ,Protein-Serine-Threonine Kinases ,Cell Cycle Proteins ,RNA ,Guide ,Genetic Engineering ,S Phase ,Phenotype ,DNA Breaks ,Double-Stranded ,HEK293 Cells ,Homologous Recombination ,Recombinational DNA Repair ,CRISPR-Cas Systems ,Gene Editing ,HeLa Cells ,Biotechnology ,Genetics ,2.1 Biological and endogenous factors - Abstract
Repair of double strand DNA breaks (DSBs) can result in gene disruption or gene modification via homology directed repair (HDR) from donor DNA. Altering cellular responses to DSBs may rebalance editing outcomes towards HDR and away from other repair outcomes. Here, we utilize a pooled CRISPR screen to define host cell involvement in HDR between a Cas9 DSB and a plasmid double stranded donor DNA (dsDonor). We find that the Fanconi Anemia (FA) pathway is required for dsDonor HDR and that other genes act to repress HDR. Small molecule inhibition of one of these repressors, CDC7, by XL413 and other inhibitors increases the efficiency of HDR by up to 3.5 fold in many contexts, including primary T cells. XL413 stimulates HDR during a reversible slowing of S-phase that is unexplored for Cas9-induced HDR. We anticipate that XL413 and other such rationally developed inhibitors will be useful tools for gene modification.
- Published
- 2020
13. Maladaptive Contractility of 3D Human Cardiac Microtissues to Mechanical Nonuniformity
- Author
-
Wang, Chenyan, Koo, Sangmo, Park, Minok, Vangelatos, Zacharias, Hoang, Plansky, Conklin, Bruce R, Grigoropoulos, Costas P, Healy, Kevin E, and Ma, Zhen
- Subjects
Engineering ,Biomedical Engineering ,Bioengineering ,Heart Disease ,Cardiovascular ,Heart ,Humans ,Mechanical Phenomena ,Muscle Contraction ,Tissue Engineering ,3D cardiac tissue models ,3D-printed microtissues ,cardiac tissue models ,hybrid biomaterial scaffolds ,tissue mechanical environments ,3D-printed microtissues ,cardiac tissue models ,Medicinal and Biomolecular Chemistry ,Medical Biotechnology ,Medical biotechnology ,Biomedical engineering - Abstract
Cardiac tissues are able to adjust their contractile behavior to adapt to the local mechanical environment. Nonuniformity of the native tissue mechanical properties contributes to the development of heart dysfunctions, yet the current in vitro cardiac tissue models often fail to recapitulate the mechanical nonuniformity. To address this issue, a 3D cardiac microtissue model is developed with engineered mechanical nonuniformity, enabled by 3D-printed hybrid matrices composed of fibers with different diameters. When escalating the complexity of tissue mechanical environments, cardiac microtissues start to develop maladaptive hypercontractile phenotypes, demonstrated in both contractile motion analysis and force-power analysis. This novel hybrid system could potentially facilitate the establishment of "pathologically-inspired" cardiac microtissue models for deeper understanding of heart pathology due to nonuniformity of the tissue mechanical environment.
- Published
- 2020
14. AlleleAnalyzer: a tool for personalized and allele-specific sgRNA design
- Author
-
Keough, Kathleen C, Lyalina, Svetlana, Olvera, Michael P, Whalen, Sean, Conklin, Bruce R, and Pollard, Katherine S
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Genetics ,Human Genome ,Generic health relevance ,Good Health and Well Being ,Alleles ,Base Sequence ,CRISPR-Associated Proteins ,Humans ,Polymorphism ,Genetic ,RNA ,Guide ,Kinetoplastida ,Software ,CRISPR ,sgRNA design ,Genomics ,Genome surgery ,Genome editing ,Computational biology ,Environmental Sciences ,Information and Computing Sciences ,Bioinformatics - Abstract
The CRISPR/Cas system is a highly specific genome editing tool capable of distinguishing alleles differing by even a single base pair. Target sites might carry genetic variations that are not distinguishable by sgRNA designing tools based on one reference genome. AlleleAnalyzer is an open-source software that incorporates single-nucleotide variants and short insertions and deletions to design sgRNAs for precisely editing 1 or multiple haplotypes of a sequenced genome, currently supporting 11 Cas proteins. It also leverages patterns of shared genetic variation to optimize sgRNA design for different human populations. AlleleAnalyzer is available at https://github.com/keoughkath/AlleleAnalyzer .
- Published
- 2019
15. Automated Design of Pluripotent Stem Cell Self-Organization
- Author
-
Libby, Ashley RG, Briers, Demarcus, Haghighi, Iman, Joy, David A, Conklin, Bruce R, Belta, Calin, and McDevitt, Todd C
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Regenerative Medicine ,Bioengineering ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Stem Cell Research - Embryonic - Human ,Networking and Information Technology R&D (NITRD) ,Stem Cell Research ,Stem Cell Research - Induced Pluripotent Stem Cell ,Antigens ,CD ,Cadherins ,Cell Differentiation ,Cell Line ,Computational Biology ,Computer Simulation ,Humans ,Machine Learning ,Pluripotent Stem Cells ,rho-Associated Kinases ,bioengineering ,control theory ,machine learning ,mathematical optimization ,multicellular patterning ,stem cell biology ,Biochemistry and cell biology - Abstract
Human pluripotent stem cells (hPSCs) have the intrinsic ability to self-organize into complex multicellular organoids that recapitulate many aspects of tissue development. However, robustly directing morphogenesis of hPSC-derived organoids requires novel approaches to accurately control self-directed pattern formation. Here, we combined genetic engineering with computational modeling, machine learning, and mathematical pattern optimization to create a data-driven approach to control hPSC self-organization by knock down of genes previously shown to affect stem cell colony organization, CDH1 and ROCK1. Computational replication of the in vitro system in silico using an extended cellular Potts model enabled machine learning-driven optimization of parameters that yielded emergence of desired patterns. Furthermore, in vitro the predicted experimental parameters quantitatively recapitulated the in silico patterns. These results demonstrate that morphogenic dynamics can be accurately predicted through model-driven exploration of hPSC behaviors via machine learning, thereby enabling spatial control of multicellular patterning to engineer human organoids and tissues. A record of this paper's Transparent Peer Review process is included in the Supplemental Information.
- Published
- 2019
16. Phenotype-Based High-Throughput Classification of Long QT Syndrome Subtypes Using Human Induced Pluripotent Stem Cells
- Author
-
Yoshinaga, Daisuke, Baba, Shiro, Makiyama, Takeru, Shibata, Hirofumi, Hirata, Takuya, Akagi, Kentaro, Matsuda, Koichi, Kohjitani, Hirohiko, Wuriyanghai, Yimin, Umeda, Katsutsugu, Yamamoto, Yuta, Conklin, Bruce R, Horie, Minoru, Takita, Junko, and Heike, Toshio
- Subjects
Biological Sciences ,Genetics ,Human Genome ,Orphan Drug ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Rare Diseases ,Stem Cell Research - Induced Pluripotent Stem Cell ,Cardiovascular ,Heart Disease ,Clinical Research ,Stem Cell Research ,Aetiology ,2.1 Biological and endogenous factors ,Action Potentials ,Adolescent ,Adult ,Case-Control Studies ,Cell Differentiation ,Child ,Female ,High-Throughput Screening Assays ,Humans ,Induced Pluripotent Stem Cells ,Long QT Syndrome ,Male ,Middle Aged ,Myocytes ,Cardiac ,Patch-Clamp Techniques ,Phenotype ,Potassium Channel Blockers ,Sodium Channel Blockers ,Tetrodotoxin ,Young Adult ,genome editing ,induced pluripotent stem cell ,long QT syndrome ,multi-electrode array ,phenotype-based diagnosis ,Biochemistry and Cell Biology ,Clinical Sciences ,Biochemistry and cell biology - Abstract
For long QT syndrome (LQTS), recent progress in genome-sequencing technologies enabled the identification of rare genomic variants with diagnostic, prognostic, and therapeutic implications. However, pathogenic stratification of the identified variants remains challenging, especially in variants of uncertain significance. This study aimed to propose a phenotypic cell-based diagnostic assay for identifying LQTS to recognize pathogenic variants in a high-throughput manner suitable for screening. We investigated the response of LQT2-induced pluripotent stem cell (iPSC)-derived cardiomyocytes (iPSC-CMs) following IKr blockade using a multi-electrode array, finding that the response to IKr blockade was significantly smaller than in Control-iPSC-CMs. Furthermore, we found that LQT1-iPSC-CMs and LQT3-iPSC-CMs could be distinguished from Control-iPSC-CMs by IKs blockade and INa blockade, respectively. This strategy might be helpful in compensating for the shortcomings of genetic testing of LQTS patients.
- Published
- 2019
17. Mapping cis-regulatory chromatin contacts in neural cells links neuropsychiatric disorder risk variants to target genes
- Author
-
Song, Michael, Yang, Xiaoyu, Ren, Xingjie, Maliskova, Lenka, Li, Bingkun, Jones, Ian R, Wang, Chao, Jacob, Fadi, Wu, Kenneth, Traglia, Michela, Tam, Tsz Wai, Jamieson, Kirsty, Lu, Si-Yao, Ming, Guo-Li, Li, Yun, Yao, Jun, Weiss, Lauren A, Dixon, Jesse R, Judge, Luke M, Conklin, Bruce R, Song, Hongjun, Gan, Li, and Shen, Yin
- Subjects
Biochemistry and Cell Biology ,Bioinformatics and Computational Biology ,Genetics ,Biological Sciences ,Human Genome ,Stem Cell Research ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Stem Cell Research - Induced Pluripotent Stem Cell ,Brain Disorders ,Mental Health ,Neurosciences ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Neurological ,Cell Lineage ,Chromatin ,Chromosome Mapping ,Clustered Regularly Interspaced Short Palindromic Repeats ,Enhancer Elements ,Genetic ,Gene Editing ,Gene Expression Regulation ,Genetic Markers ,Genome ,Human ,Genome-Wide Association Study ,Humans ,Induced Pluripotent Stem Cells ,Infant ,Male ,Mental Disorders ,Neurons ,Polymorphism ,Single Nucleotide ,Promoter Regions ,Genetic ,Medical and Health Sciences ,Developmental Biology ,Agricultural biotechnology ,Bioinformatics and computational biology - Abstract
Mutations in gene regulatory elements have been associated with a wide range of complex neuropsychiatric disorders. However, due to their cell-type specificity and difficulties in characterizing their regulatory targets, the ability to identify causal genetic variants has remained limited. To address these constraints, we perform an integrative analysis of chromatin interactions, open chromatin regions and transcriptomes using promoter capture Hi-C, assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq) and RNA sequencing, respectively, in four functionally distinct neural cell types: induced pluripotent stem cell (iPSC)-induced excitatory neurons and lower motor neurons, iPSC-derived hippocampal dentate gyrus-like neurons and primary astrocytes. We identify hundreds of thousands of long-range cis-interactions between promoters and distal promoter-interacting regions, enabling us to link regulatory elements to their target genes and reveal putative processes that are dysregulated in disease. Finally, we validate several promoter-interacting regions by using clustered regularly interspaced short palindromic repeats (CRISPR) techniques in human excitatory neurons, demonstrating that CDK5RAP3, STRAP and DRD2 are transcriptionally regulated by physically linked enhancers.
- Published
- 2019
18. Unbiased detection of CRISPR off-targets in vivo using DISCOVER-Seq
- Author
-
Wienert, Beeke, Wyman, Stacia K, Richardson, Christopher D, Yeh, Charles D, Akcakaya, Pinar, Porritt, Michelle J, Morlock, Michaela, Vu, Jonathan T, Kazane, Katelynn R, Watry, Hannah L, Judge, Luke M, Conklin, Bruce R, Maresca, Marcello, and Corn, Jacob E
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Stem Cell Research - Induced Pluripotent Stem Cell ,Human Genome ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Biotechnology ,Stem Cell Research ,Regenerative Medicine ,Genetics ,Aetiology ,2.2 Factors relating to the physical environment ,Adenoviridae ,Animals ,CRISPR-Associated Protein 9 ,CRISPR-Cas Systems ,Cell Line ,Chromatin Immunoprecipitation ,Clustered Regularly Interspaced Short Palindromic Repeats ,DNA ,DNA Breaks ,Double-Stranded ,DNA Repair ,DNA Repair Enzymes ,Gene Editing ,Humans ,Induced Pluripotent Stem Cells ,K562 Cells ,MRE11 Homologue Protein ,RNA ,Guide ,Kinetoplastida ,Sequence Analysis ,DNA ,General Science & Technology - Abstract
CRISPR-Cas genome editing induces targeted DNA damage but can also affect off-target sites. Current off-target discovery methods work using purified DNA or specific cellular models but are incapable of direct detection in vivo. We developed DISCOVER-Seq (discovery of in situ Cas off-targets and verification by sequencing), a universally applicable approach for unbiased off-target identification that leverages the recruitment of DNA repair factors in cells and organisms. Tracking the precise recruitment of MRE11 uncovers the molecular nature of Cas activity in cells with single-base resolution. DISCOVER-Seq works with multiple guide RNA formats and types of Cas enzymes, allowing characterization of new editing tools. Off-targets can be identified in cell lines and patient-derived induced pluripotent stem cells and during adenoviral editing of mice, paving the way for in situ off-target discovery within individual patient genotypes during therapeutic genome editing.
- Published
- 2019
19. MESP1 knock-down in human iPSC attenuates early vascular progenitor cell differentiation after completed primitive streak specification
- Author
-
Eskildsen, Tilde V, Ayoubi, Sohrab, Thomassen, Mads, Burton, Mark, Mandegar, Mohammed A, Conklin, Bruce R, Jensen, Charlotte H, Andersen, Ditte C, and Sheikh, Søren P
- Subjects
Biological Sciences ,Cardiovascular ,Stem Cell Research ,Genetics ,1.1 Normal biological development and functioning ,Underpinning research ,Basic Helix-Loop-Helix Transcription Factors ,Cell Differentiation ,Cell Lineage ,Embryonic Stem Cells ,Endothelial Progenitor Cells ,Fetal Proteins ,Gene Expression Regulation ,Developmental ,Helix-Loop-Helix Motifs ,Homeodomain Proteins ,Humans ,Induced Pluripotent Stem Cells ,Mesoderm ,Myocardium ,Myocytes ,Cardiac ,Primitive Streak ,T-Box Domain Proteins ,Transcription Factors ,MESP1 ,IPSC ,Vascular progenitor ,CRISPR ,Differentiation ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
MESP1 is a key transcription factor in development of early cardiovascular tissue and it is required for induction of the cardiomyocyte (CM) gene expression program, but its role in vascular development is unclear. Here, we used inducible CRISPRi knock-down of MESP1 to analyze the molecular processes of the early differentiation stages of human induced pluripotent stem cells into mesoderm and subsequently vascular progenitor cells. We found that expression of the mesodermal marker, BRACHYURY (encoded by T) was unaffected in MESP1 knock-down cells as compared to wild type cells suggesting timely movement through the primitive streak whereas another mesodermal marker MIXL1 was slightly, but significantly decreased. In contrast, the expression of the vascular cell surface marker KDR was decreased and CD31 and CD34 expression were substantially reduced in MESP1 knock-down cells supporting inhibition or delay of vascular specification. In addition, mRNA microarray data revealed several other altered gene expressions including the EMT regulating transcription factors SNAI1 and TWIST1, which were both significantly decreased indicating that MESP1 knock-down cells are less likely to undergo EMT during vascular progenitor differentiation. Our study demonstrates that while leaving primitive streak markers unaffected, MESP1 expression is required for timely vascular progenitor specification. Thus, MESP1 expression is essential for the molecular features of early CM, EC and VSMC lineage specification.
- Published
- 2019
20. Contractile deficits in engineered cardiac microtissues as a result of MYBPC3 deficiency and mechanical overload.
- Author
-
Ma, Zhen, Huebsch, Nathaniel, Koo, Sangmo, Mandegar, Mohammad A, Siemons, Brian, Boggess, Steven, Conklin, Bruce R, Grigoropoulos, Costas P, and Healy, Kevin E
- Subjects
Sarcomeres ,Myocardium ,Cells ,Cultured ,Myocytes ,Cardiac ,Humans ,Cardiomyopathy ,Dilated ,Calcium ,Carrier Proteins ,Tissue Engineering ,Myocardial Contraction ,Stress ,Mechanical ,E1A-Associated p300 Protein ,GATA4 Transcription Factor ,Gene Knockout Techniques ,Induced Pluripotent Stem Cells ,Cells ,Cultured ,Myocytes ,Cardiac ,Cardiomyopathy ,Dilated ,Stress ,Mechanical - Abstract
The integration of in vitro cardiac tissue models, human induced pluripotent stem cells (hiPSCs) and genome-editing tools allows for the enhanced interrogation of physiological phenotypes and recapitulation of disease pathologies. Here, using a cardiac tissue model consisting of filamentous three-dimensional matrices populated with cardiomyocytes derived from healthy wild-type (WT) hiPSCs (WT hiPSC-CMs) or isogenic hiPSCs deficient in the sarcomere protein cardiac myosin-binding protein C (MYBPC3-/- hiPSC-CMs), we show that the WT microtissues adapted to the mechanical environment with increased contraction force commensurate to matrix stiffness, whereas the MYBPC3-/- microtissues exhibited impaired force development kinetics regardless of matrix stiffness and deficient contraction force only when grown on matrices with high fibre stiffness. Under mechanical overload, the MYBPC3-/- microtissues had a higher degree of calcium transient abnormalities, and exhibited an accelerated decay of calcium dynamics as well as calcium desensitization, which accelerated when contracting against stiffer fibres. Our findings suggest that MYBPC3 deficiency and the presence of environmental stresses synergistically lead to contractile deficits in cardiac tissues.
- Published
- 2018
21. Spatiotemporal mosaic self-patterning of pluripotent stem cells using CRISPR interference.
- Author
-
Libby, Ashley Rg, Joy, David A, So, Po-Lin, Mandegar, Mohammad A, Muncie, Jonathon M, Mendoza-Camacho, Federico N, Weaver, Valerie M, Conklin, Bruce R, and McDevitt, Todd C
- Subjects
Humans ,Cadherins ,Antigens ,CD ,Cell Communication ,Cell Differentiation ,Cell Lineage ,Morphogenesis ,rho-Associated Kinases ,Gene Knockdown Techniques ,Induced Pluripotent Stem Cells ,CRISPR-Cas Systems ,bio-engineering ,developmental biology ,human ,morphogenesis ,pluripotent stem cells ,regenerative medicine ,stem cells ,Stem Cell Research - Induced Pluripotent Stem Cell ,Stem Cell Research ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Genetics ,1.1 Normal biological development and functioning ,Generic health relevance ,Biochemistry and Cell Biology - Abstract
Morphogenesis involves interactions of asymmetric cell populations to form complex multicellular patterns and structures comprised of distinct cell types. However, current methods to model morphogenic events lack control over cell-type co-emergence and offer little capability to selectively perturb specific cell subpopulations. Our in vitro system interrogates cell-cell interactions and multicellular organization within human induced pluripotent stem cell (hiPSC) colonies. We examined effects of induced mosaic knockdown of molecular regulators of cortical tension (ROCK1) and cell-cell adhesion (CDH1) with CRISPR interference. Mosaic knockdown of ROCK1 or CDH1 resulted in differential patterning within hiPSC colonies due to cellular self-organization, while retaining an epithelial pluripotent phenotype. Knockdown induction stimulates a transient wave of differential gene expression within the mixed populations that stabilized in coordination with observed self-organization. Mosaic patterning enables genetic interrogation of emergent multicellular properties, which can facilitate better understanding of the molecular pathways that regulate symmetry-breaking during morphogenesis.
- Published
- 2018
22. Quantitatively characterizing drug‐induced arrhythmic contractile motions of human stem cell‐derived cardiomyocytes
- Author
-
Hoang, Plansky, Huebsch, Nathaniel, Bang, Shin Hyuk, Siemons, Brian A, Conklin, Bruce R, Healy, Kevin E, Ma, Zhen, and Jacquir, Sabir
- Subjects
Regenerative Medicine ,Cardiovascular ,Heart Disease ,Networking and Information Technology R&D (NITRD) ,Bioengineering ,Stem Cell Research ,Stem Cell Research - Induced Pluripotent Stem Cell ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Arrhythmias ,Cardiac ,Cytological Techniques ,Humans ,Image Processing ,Computer-Assisted ,Induced Pluripotent Stem Cells ,Motion ,Myocardial Contraction ,Myocytes ,Cardiac ,Optical Imaging ,Software ,arrhythmia ,biosignal processing ,cardiac motion ,phase space reconstruction ,optical flow ,Biotechnology - Abstract
Quantification of abnormal contractile motions of cardiac tissue has been a noteworthy challenge and significant limitation in assessing and classifying the drug-induced arrhythmias (i.e., Torsades de pointes). To overcome these challenges, researchers have taken advantage of computational image processing tools to measure contractile motion from cardiomyocytes derived from human induced pluripotent stem cells (hiPSC-CMs). However, the amplitude and frequency analysis of contractile motion waveforms does not produce sufficient information to objectively classify the degree of variations between two or more sets of cardiac contractile motions. In this paper, we generated contractile motion data from beating hiPSC-CMs using motion tracking software based on optical flow analysis, and then implemented a computational algorithm, phase space reconstruction (PSR), to derive parameters (embedding, regularity, and fractal dimensions) to further characterize the dynamic nature of the cardiac contractile motions. Application of drugs known to cause cardiac arrhythmia induced significant changes to these resultant dimensional parameters calculated from PSR analysis. Integrating this new computational algorithm with the existing analytical toolbox of cardiac contractile motions will allow us to expand current assessments of cardiac tissue physiology into an automated, high-throughput, and quantifiable manner which will allow more objective assessments of drug-induced proarrhythmias.
- Published
- 2018
23. Generation of spatial-patterned early-developing cardiac organoids using human pluripotent stem cells
- Author
-
Hoang, Plansky, Wang, Jason, Conklin, Bruce R, Healy, Kevin E, and Ma, Zhen
- Subjects
Medical Biotechnology ,Biomedical and Clinical Sciences ,Stem Cell Research - Induced Pluripotent Stem Cell ,Stem Cell Research ,Regenerative Medicine ,Stem Cell Research - Embryonic - Human ,Clinical Research ,Stem Cell Research - Nonembryonic - Human ,Cardiovascular ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Heart Disease ,Heart ,Humans ,Induced Pluripotent Stem Cells ,Models ,Biological ,Organ Culture Techniques ,Organogenesis ,Organoids ,Chemical Sciences ,Biological Sciences ,Medical and Health Sciences ,Bioinformatics - Abstract
The creation of human induced pluripotent stem cells (hiPSCs) has provided an unprecedented opportunity to study tissue morphogenesis and organ development through 'organogenesis-in-a-dish'. Current approaches to cardiac organoid engineering rely on either direct cardiac differentiation from embryoid bodies (EBs) or generation of aligned cardiac tissues from predifferentiated cardiomyocytes from monolayer hiPSCs. To experimentally model early cardiac organogenesis in vitro, our protocol combines biomaterials-based cell patterning with stem cell organoid engineering. 3D cardiac microchambers are created from 2D hiPSC colonies; these microchambers approximate an early-development heart with distinct spatial organization and self-assembly. With proper training in photolithography microfabrication, maintenance of human pluripotent stem cells, and cardiac differentiation, a graduate student with guidance will likely be able to carry out this experimental protocol, which requires ∼3 weeks. We envisage that this in vitro model of human early heart development could serve as an embryotoxicity screening assay in drug discovery, regulation, and prescription for healthy fetal development. We anticipate that, when applied to hiPSC lines derived from patients with inherited diseases, this protocol can be used to study the disease mechanisms of cardiac malformations at an early stage of embryogenesis.
- Published
- 2018
24. Detection and Quantification of HDR and NHEJ Induced by Genome Editing at Endogenous Gene Loci Using Droplet Digital PCR
- Author
-
Miyaoka, Yuichiro, Mayerl, Steven J, Chan, Amanda H, and Conklin, Bruce R
- Subjects
Biological Sciences ,Genetics ,Human Genome ,Biotechnology ,DNA ,DNA End-Joining Repair ,Gene Editing ,Genetic Loci ,HEK293 Cells ,Humans ,Induced Pluripotent Stem Cells ,Polymerase Chain Reaction ,Recombinational DNA Repair ,Genome editing ,TALEN ,CRISPR/Cas9 ,HDR ,NHEJ ,ddPCR ,Other Chemical Sciences ,Biochemistry and Cell Biology ,Developmental Biology ,Biochemistry and cell biology ,Medicinal and biomolecular chemistry - Abstract
Genome editing holds great promise for experimental biology and potential clinical use. To successfully utilize genome editing, it is critical to sensitively detect and quantify its outcomes: homology-directed repair (HDR) and nonhomologous end joining (NHEJ). This has been difficult at endogenous gene loci and instead is frequently done using artificial reporter systems. Here, we describe a droplet digital PCR (ddPCR)-based method to simultaneously measure HDR and NHEJ at endogenous gene loci. This highly sensitive and quantitative method may significantly contribute to a better understanding of DNA repair mechanisms underlying genome editing and to the improvement of genome editing technology by allowing for efficient and systematic testing of many genome editing conditions in parallel.
- Published
- 2018
25. Reversal of C9orf72 mutation-induced transcriptional dysregulation and pathology in cultured human neurons by allele-specific excision
- Author
-
Sachdev, Aradhana, primary, Gill, Kamaljot, additional, Sckaff, Maria, additional, Birk, Alisha M., additional, Aladesuyi Arogundade, Olubankole, additional, Brown, Katherine A., additional, Chouhan, Runvir S., additional, Issagholian-Lewin, Patrick Oliver, additional, Patel, Esha, additional, Watry, Hannah L., additional, Bernardi, Mylinh T., additional, Keough, Kathleen C., additional, Tsai, Yu-Chih, additional, Smith, Alec Simon Tulloch, additional, Conklin, Bruce R., additional, and Clelland, Claire Dudley, additional
- Published
- 2024
- Full Text
- View/download PDF
26. Bioengineered optogenetic model of human neuromuscular junction
- Author
-
Vila, Olaia F., Chavez, Miguel, Ma, Stephen P., Yeager, Keith, Zholudeva, Lyandysha V., Colón-Mercado, Jennifer M., Qu, Yihuai, Nash, Trevor R., Lai, Carmen, Feliciano, Carissa M., Carter, Matthew, Kamm, Roger D., Judge, Luke M., Conklin, Bruce R., Ward, Michael E., McDevitt, Todd C., and Vunjak-Novakovic, Gordana
- Published
- 2021
- Full Text
- View/download PDF
27. Transcription factor overexpression drives reliable differentiation of retinal pigment epithelium from human induced pluripotent stem cells
- Author
-
Dewell, Tessa E., Gjoni, Ketrin, Liu, Angela Z., Libby, Ashley R.G., Moore, Anthony T., So, Po-Lin, and Conklin, Bruce R.
- Published
- 2021
- Full Text
- View/download PDF
28. A BAG3 chaperone complex maintains cardiomyocyte function during proteotoxic stress.
- Author
-
Judge, Luke M, Perez-Bermejo, Juan A, Truong, Annie, Ribeiro, Alexandre Js, Yoo, Jennie C, Jensen, Christina L, Mandegar, Mohammad A, Huebsch, Nathaniel, Kaake, Robyn M, So, Po-Lin, Srivastava, Deepak, Pruitt, Beth L, Krogan, Nevan J, and Conklin, Bruce R
- Subjects
Cardiology ,Cell Biology - Abstract
Molecular chaperones regulate quality control in the human proteome, pathways that have been implicated in many diseases, including heart failure. Mutations in the BAG3 gene, which encodes a co-chaperone protein, have been associated with heart failure due to both inherited and sporadic dilated cardiomyopathy. Familial BAG3 mutations are autosomal dominant and frequently cause truncation of the coding sequence, suggesting a heterozygous loss-of-function mechanism. However, heterozygous knockout of the murine BAG3 gene did not cause a detectable phenotype. To model BAG3 cardiomyopathy in a human system, we generated an isogenic series of human induced pluripotent stem cells (iPSCs) with loss-of-function mutations in BAG3. Heterozygous BAG3 mutations reduced protein expression, disrupted myofibril structure, and compromised contractile function in iPSC-derived cardiomyocytes (iPS-CMs). BAG3-deficient iPS-CMs were particularly sensitive to further myofibril disruption and contractile dysfunction upon exposure to proteasome inhibitors known to cause cardiotoxicity. We performed affinity tagging of the endogenous BAG3 protein and mass spectrometry proteomics to further define the cardioprotective chaperone complex that BAG3 coordinates in the human heart. Our results establish a model for evaluating protein quality control pathways in human cardiomyocytes and their potential as therapeutic targets and susceptibility factors for cardiac drug toxicity.
- Published
- 2017
29. Multi-Imaging Method to Assay the Contractile Mechanical Output of Micropatterned Human iPSC-Derived Cardiac Myocytes
- Author
-
Ribeiro, Alexandre JS, Schwab, Olivier, Mandegar, Mohammad A, Ang, Yen-Sin, Conklin, Bruce R, Srivastava, Deepak, and Pruitt, Beth L
- Subjects
Bioengineering ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Cardiovascular ,Stem Cell Research - Induced Pluripotent Stem Cell ,Stem Cell Research ,Heart Disease ,Cells ,Cultured ,Humans ,Induced Pluripotent Stem Cells ,Multimodal Imaging ,Myocardial Contraction ,Myocytes ,Cardiac ,cardiac myocyte ,contractility ,sarcomere length ,single cell ,stem cell ,Cardiorespiratory Medicine and Haematology ,Clinical Sciences ,Cardiovascular System & Hematology - Abstract
RationaleDuring each beat, cardiac myocytes (CMs) generate the mechanical output necessary for heart function through contractile mechanisms that involve shortening of sarcomeres along myofibrils. Human-induced pluripotent stem cells (hiPSCs) can be differentiated into CMs (hiPSC-CMs) that model cardiac contractile mechanical output more robustly when micropatterned into physiological shapes. Quantifying the mechanical output of these cells enables us to assay cardiac activity in a dish.ObjectiveWe sought to develop a computational platform that integrates analytic approaches to quantify the mechanical output of single micropatterned hiPSC-CMs from microscopy videos.Methods and resultsWe micropatterned single hiPSC-CMs on deformable polyacrylamide substrates containing fluorescent microbeads. We acquired videos of single beating cells, of microbead displacement during contractions, and of fluorescently labeled myofibrils. These videos were independently analyzed to obtain parameters that capture the mechanical output of the imaged single cells. We also developed novel methods to quantify sarcomere length from videos of moving myofibrils and to analyze loss of synchronicity of beating in cells with contractile defects. We tested this computational platform by detecting variations in mechanical output induced by drugs and in cells expressing low levels of myosin-binding protein C.ConclusionsOur method can measure the cardiac function of single micropatterned hiPSC-CMs and determine contractile parameters that can be used to elucidate mechanisms that underlie variations in CM function. This platform will be amenable to future studies of the effects of mutations and drugs on cardiac function.
- Published
- 2017
30. CRISPRi-based genome-scale identification of functional long noncoding RNA loci in human cells
- Author
-
Liu, S John, Horlbeck, Max A, Cho, Seung Woo, Birk, Harjus S, Malatesta, Martina, He, Daniel, Attenello, Frank J, Villalta, Jacqueline E, Cho, Min Y, Chen, Yuwen, Mandegar, Mohammad A, Olvera, Michael P, Gilbert, Luke A, Conklin, Bruce R, Chang, Howard Y, Weissman, Jonathan S, and Lim, Daniel A
- Subjects
Stem Cell Research ,Regenerative Medicine ,Stem Cell Research - Induced Pluripotent Stem Cell ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Stem Cell Research - Embryonic - Human ,Biotechnology ,Human Genome ,Genetics ,Aetiology ,Underpinning research ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Generic health relevance ,Cell Growth Processes ,Cell Line ,Clustered Regularly Interspaced Short Palindromic Repeats ,Gene Knockdown Techniques ,Gene Regulatory Networks ,Genetic Loci ,Genetic Testing ,Genome ,Human ,Humans ,Induced Pluripotent Stem Cells ,Machine Learning ,RNA Interference ,RNA ,Long Noncoding ,Transcription ,Genetic ,Transcriptome ,General Science & Technology - Abstract
The human genome produces thousands of long noncoding RNAs (lncRNAs)-transcripts >200 nucleotides long that do not encode proteins. Although critical roles in normal biology and disease have been revealed for a subset of lncRNAs, the function of the vast majority remains untested. We developed a CRISPR interference (CRISPRi) platform targeting 16,401 lncRNA loci in seven diverse cell lines, including six transformed cell lines and human induced pluripotent stem cells (iPSCs). Large-scale screening identified 499 lncRNA loci required for robust cellular growth, of which 89% showed growth-modifying function exclusively in one cell type. We further found that lncRNA knockdown can perturb complex transcriptional networks in a cell type-specific manner. These data underscore the functional importance and cell type specificity of many lncRNAs.
- Published
- 2017
31. Engineered human pluripotent-stem-cell-derived intestinal tissues with a functional enteric nervous system
- Author
-
Workman, Michael J, Mahe, Maxime M, Trisno, Stephen, Poling, Holly M, Watson, Carey L, Sundaram, Nambirajan, Chang, Ching-Fang, Schiesser, Jacqueline, Aubert, Philippe, Stanley, Edouard G, Elefanty, Andrew G, Miyaoka, Yuichiro, Mandegar, Mohammad A, Conklin, Bruce R, Neunlist, Michel, Brugmann, Samantha A, Helmrath, Michael A, and Wells, James M
- Subjects
Neurosciences ,Stem Cell Research - Induced Pluripotent Stem Cell ,Stem Cell Research - Nonembryonic - Human ,Regenerative Medicine ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Stem Cell Research - Embryonic - Human ,Digestive Diseases ,Congenital Structural Anomalies ,Stem Cell Research ,Pediatric ,1.1 Normal biological development and functioning ,5.2 Cellular and gene therapies ,Development of treatments and therapeutic interventions ,Underpinning research ,Generic health relevance ,Animals ,Calcium ,Cell Line ,Chick Embryo ,Enteric Nervous System ,Gastrointestinal Motility ,Hirschsprung Disease ,Homeodomain Proteins ,Humans ,Immunohistochemistry ,In Vitro Techniques ,Induced Pluripotent Stem Cells ,Interstitial Cells of Cajal ,Intestines ,Mice ,Mice ,SCID ,Microscopy ,Confocal ,Models ,Biological ,Mutation ,Myenteric Plexus ,Neural Crest ,Neurogenesis ,Neuroglia ,Neurons ,Organoids ,Permeability ,Real-Time Polymerase Chain Reaction ,Submucous Plexus ,Tissue Engineering ,Transcription Factors ,Medical and Health Sciences ,Immunology - Abstract
The enteric nervous system (ENS) of the gastrointestinal tract controls many diverse functions, including motility and epithelial permeability. Perturbations in ENS development or function are common, yet there is no human model for studying ENS-intestinal biology and disease. We used a tissue-engineering approach with embryonic and induced pluripotent stem cells (PSCs) to generate human intestinal tissue containing a functional ENS. We recapitulated normal intestinal ENS development by combining human-PSC-derived neural crest cells (NCCs) and developing human intestinal organoids (HIOs). NCCs recombined with HIOs in vitro migrated into the mesenchyme, differentiated into neurons and glial cells and showed neuronal activity, as measured by rhythmic waves of calcium transients. ENS-containing HIOs grown in vivo formed neuroglial structures similar to a myenteric and submucosal plexus, had functional interstitial cells of Cajal and had an electromechanical coupling that regulated waves of propagating contraction. Finally, we used this system to investigate the cellular and molecular basis for Hirschsprung's disease caused by a mutation in the gene PHOX2B. This is, to the best of our knowledge, the first demonstration of human-PSC-derived intestinal tissue with a functional ENS and how this system can be used to study motility disorders of the human gastrointestinal tract.
- Published
- 2017
32. BMP-SMAD-ID promotes reprogramming to pluripotency by inhibiting p16/INK4A-dependent senescence
- Author
-
Hayashi, Yohei, Hsiao, Edward C, Sami, Salma, Lancero, Mariselle, Schlieve, Christopher R, Nguyen, Trieu, Yano, Koyori, Nagahashi, Ayako, Ikeya, Makoto, Matsumoto, Yoshihisa, Nishimura, Ken, Fukuda, Aya, Hisatake, Koji, Tomoda, Kiichiro, Asaka, Isao, Toguchida, Junya, Conklin, Bruce R, and Yamanaka, Shinya
- Subjects
Stem Cell Research ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Stem Cell Research - Induced Pluripotent Stem Cell ,2.1 Biological and endogenous factors ,Aetiology ,Underpinning research ,1.1 Normal biological development and functioning ,Activin Receptors ,Type I ,Adolescent ,Adult ,Animals ,Bone Morphogenetic Proteins ,Cell Line ,Cellular Reprogramming ,Cellular Senescence ,Child ,Cyclin-Dependent Kinase Inhibitor p16 ,Female ,Fibroblasts ,Humans ,Induced Pluripotent Stem Cells ,Male ,Mice ,Transgenic ,Middle Aged ,Mutation ,Myositis Ossificans ,Signal Transduction ,Smad Proteins ,reprogramming ,pluripotency ,BMP ,senescence ,FOP - Abstract
Fibrodysplasia ossificans progressiva (FOP) patients carry a missense mutation in ACVR1 [617G > A (R206H)] that leads to hyperactivation of BMP-SMAD signaling. Contrary to a previous study, here we show that FOP fibroblasts showed an increased efficiency of induced pluripotent stem cell (iPSC) generation. This positive effect was attenuated by inhibitors of BMP-SMAD signaling (Dorsomorphin or LDN1931890) or transducing inhibitory SMADs (SMAD6 or SMAD7). In normal fibroblasts, the efficiency of iPSC generation was enhanced by transducing mutant ACVR1 (617G > A) or SMAD1 or adding BMP4 protein at early times during the reprogramming. In contrast, adding BMP4 at later times decreased iPSC generation. ID genes, transcriptional targets of BMP-SMAD signaling, were critical for iPSC generation. The BMP-SMAD-ID signaling axis suppressed p16/INK4A-mediated cell senescence, a major barrier to reprogramming. These results using patient cells carrying the ACVR1 R206H mutation reveal how cellular signaling and gene expression change during the reprogramming processes.
- Published
- 2016
33. Transcription Factor GATA4 Regulates Cell Type–Specific Splicing Through Direct Interaction With RNA in Human Induced Pluripotent Stem Cell–Derived Cardiac Progenitors
- Author
-
Zhu, Lili, Choudhary, Krishna, Gonzalez-Teran, Barbara, Ang, Yen-Sin, Thomas, Reuben, Stone, Nicole R., Liu, Lei, Zhou, Ping, Zhu, Chenchen, Ruan, Hongmei, Huang, Yu, Jin, Shibo, Pelonero, Angelo, Koback, Frances, Padmanabhan, Arun, Sadagopan, Nandhini, Hsu, Austin, Costa, Mauro W., Gifford, Casey A., van Bemmel, Joke G., Hüttenhain, Ruth, Vedantham, Vasanth, Conklin, Bruce R., Black, Brian L., Bruneau, Benoit G., Steinmetz, Lars, Krogan, Nevan J., Pollard, Katherine S., and Srivastava, Deepak
- Published
- 2022
- Full Text
- View/download PDF
34. Ligand-binding domains of nuclear receptors facilitate tight control of split CRISPR activity
- Author
-
Nguyen, Duy P, Miyaoka, Yuichiro, Gilbert, Luke A, Mayerl, Steven J, Lee, Brian H, Weissman, Jonathan S, Conklin, Bruce R, and Wells, James A
- Published
- 2016
35. Miniaturized iPS-Cell-Derived Cardiac Muscles for Physiologically Relevant Drug Response Analyses.
- Author
-
Huebsch, Nathaniel, Loskill, Peter, Deveshwar, Nikhil, Spencer, C Ian, Judge, Luke M, Mandegar, Mohammad A, Fox, Cade B, Mohamed, Tamer MA, Ma, Zhen, Mathur, Anurag, Sheehan, Alice M, Truong, Annie, Saxton, Mike, Yoo, Jennie, Srivastava, Deepak, Desai, Tejal A, So, Po-Lin, Healy, Kevin E, and Conklin, Bruce R
- Subjects
Sarcomeres ,Cells ,Cultured ,Stromal Cells ,Myocytes ,Cardiac ,Humans ,Fluorescent Antibody Technique ,Cell Differentiation ,Induced Pluripotent Stem Cells ,Cells ,Cultured ,Myocytes ,Cardiac ,Cardiovascular ,Heart Disease ,Stem Cell Research ,Biochemistry and Cell Biology ,Other Physical Sciences - Abstract
Tissue engineering approaches have the potential to increase the physiologic relevance of human iPS-derived cells, such as cardiomyocytes (iPS-CM). However, forming Engineered Heart Muscle (EHM) typically requires >1 million cells per tissue. Existing miniaturization strategies involve complex approaches not amenable to mass production, limiting the ability to use EHM for iPS-based disease modeling and drug screening. Micro-scale cardiospheres are easily produced, but do not facilitate assembly of elongated muscle or direct force measurements. Here we describe an approach that combines features of EHM and cardiospheres: Micro-Heart Muscle (μHM) arrays, in which elongated muscle fibers are formed in an easily fabricated template, with as few as 2,000 iPS-CM per individual tissue. Within μHM, iPS-CM exhibit uniaxial contractility and alignment, robust sarcomere assembly, and reduced variability and hypersensitivity in drug responsiveness, compared to monolayers with the same cellular composition. μHM mounted onto standard force measurement apparatus exhibited a robust Frank-Starling response to external stretch, and a dose-dependent inotropic response to the β-adrenergic agonist isoproterenol. Based on the ease of fabrication, the potential for mass production and the small number of cells required to form μHM, this system provides a potentially powerful tool to study cardiomyocyte maturation, disease and cardiotoxicology in vitro.
- Published
- 2016
36. CRISPR Interference Efficiently Induces Specific and Reversible Gene Silencing in Human iPSCs
- Author
-
Mandegar, Mohammad A, Huebsch, Nathaniel, Frolov, Ekaterina B, Shin, Edward, Truong, Annie, Olvera, Michael P, Chan, Amanda H, Miyaoka, Yuichiro, Holmes, Kristin, Spencer, C Ian, Judge, Luke M, Gordon, David E, Eskildsen, Tilde V, Villalta, Jacqueline E, Horlbeck, Max A, Gilbert, Luke A, Krogan, Nevan J, Sheikh, Søren P, Weissman, Jonathan S, Qi, Lei S, So, Po-Lin, and Conklin, Bruce R
- Subjects
Biological Sciences ,Genetics ,Stem Cell Research ,Human Genome ,Stem Cell Research - Induced Pluripotent Stem Cell ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Biotechnology ,Regenerative Medicine ,Stem Cell Research - Induced Pluripotent Stem Cell - Non-Human ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Generic health relevance ,Clustered Regularly Interspaced Short Palindromic Repeats ,Gene Silencing ,Humans ,Induced Pluripotent Stem Cells ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
Developing technologies for efficient and scalable disruption of gene expression will provide powerful tools for studying gene function, developmental pathways, and disease mechanisms. Here, we develop clustered regularly interspaced short palindromic repeat interference (CRISPRi) to repress gene expression in human induced pluripotent stem cells (iPSCs). CRISPRi, in which a doxycycline-inducible deactivated Cas9 is fused to a KRAB repression domain, can specifically and reversibly inhibit gene expression in iPSCs and iPSC-derived cardiac progenitors, cardiomyocytes, and T lymphocytes. This gene repression system is tunable and has the potential to silence single alleles. Compared with CRISPR nuclease (CRISPRn), CRISPRi gene repression is more efficient and homogenous across cell populations. The CRISPRi system in iPSCs provides a powerful platform to perform genome-scale screens in a wide range of iPSC-derived cell types, dissect developmental pathways, and model disease.
- Published
- 2016
37. Systematic quantification of HDR and NHEJ reveals effects of locus, nuclease, and cell type on genome-editing.
- Author
-
Miyaoka, Yuichiro, Berman, Jennifer R, Cooper, Samantha B, Mayerl, Steven J, Chan, Amanda H, Zhang, Bin, Karlin-Neumann, George A, and Conklin, Bruce R
- Subjects
Cell Line ,Hela Cells ,Humans ,Biological Assay ,Transfection ,Polymerase Chain Reaction ,Genome ,Human ,Plasmids ,DNA Breaks ,Double-Stranded ,DNA Breaks ,Single-Stranded ,Genetic Loci ,Induced Pluripotent Stem Cells ,HEK293 Cells ,DNA End-Joining Repair ,Recombinational DNA Repair ,CRISPR-Cas Systems ,Transcription Activator-Like Effector Nucleases ,Gene Editing ,HeLa Cells ,Genetics ,Stem Cell Research ,Human Genome ,Biotechnology ,Generic Health Relevance ,Genome ,Human ,DNA Breaks ,Double-Stranded ,Single-Stranded ,Biochemistry and Cell Biology ,Other Physical Sciences - Abstract
Precise genome-editing relies on the repair of sequence-specific nuclease-induced DNA nicking or double-strand breaks (DSBs) by homology-directed repair (HDR). However, nonhomologous end-joining (NHEJ), an error-prone repair, acts concurrently, reducing the rate of high-fidelity edits. The identification of genome-editing conditions that favor HDR over NHEJ has been hindered by the lack of a simple method to measure HDR and NHEJ directly and simultaneously at endogenous loci. To overcome this challenge, we developed a novel, rapid, digital PCR-based assay that can simultaneously detect one HDR or NHEJ event out of 1,000 copies of the genome. Using this assay, we systematically monitored genome-editing outcomes of CRISPR-associated protein 9 (Cas9), Cas9 nickases, catalytically dead Cas9 fused to FokI, and transcription activator-like effector nuclease at three disease-associated endogenous gene loci in HEK293T cells, HeLa cells, and human induced pluripotent stem cells. Although it is widely thought that NHEJ generally occurs more often than HDR, we found that more HDR than NHEJ was induced under multiple conditions. Surprisingly, the HDR/NHEJ ratios were highly dependent on gene locus, nuclease platform, and cell type. The new assay system, and our findings based on it, will enable mechanistic studies of genome-editing and help improve genome-editing technology.
- Published
- 2016
38. Generating trunk neural crest from human pluripotent stem cells.
- Author
-
Huang, Miller, Miller, Matthew L, McHenry, Lauren K, Zheng, Tina, Zhen, Qiqi, Ilkhanizadeh, Shirin, Conklin, Bruce R, Bronner, Marianne E, and Weiss, William A
- Subjects
Cells ,Cultured ,Pluripotent Stem Cells ,Neural Crest ,Humans ,Tretinoin ,Homeodomain Proteins ,Bone Morphogenetic Proteins ,Signal Transduction ,Cell Differentiation ,Gene Expression Regulation ,Developmental ,Biomarkers ,Cells ,Cultured ,Gene Expression Regulation ,Developmental ,Stem Cell Research ,Clinical Research ,Rare Diseases ,Regenerative Medicine ,Dental/Oral and Craniofacial Disease ,Neurosciences ,Pediatric ,Stem Cell Research - Nonembryonic - Human ,Stem Cell Research - Embryonic - Human ,1.1 Normal biological development and functioning ,Generic Health Relevance ,Biochemistry and Cell Biology ,Other Physical Sciences - Abstract
Neural crest cells (NCC) are stem cells that generate different lineages, including neuroendocrine, melanocytic, cartilage, and bone. The differentiation potential of NCC varies according to the level from which cells emerge along the neural tube. For example, only anterior "cranial" NCC form craniofacial bone, whereas solely posterior "trunk" NCC contribute to sympathoadrenal cells. Importantly, the isolation of human fetal NCC carries ethical and scientific challenges, as NCC induction typically occur before pregnancy is detectable. As a result, current knowledge of NCC biology derives primarily from non-human organisms. Important differences between human and non-human NCC, such as expression of HNK1 in human but not mouse NCC, suggest a need to study human NCC directly. Here, we demonstrate that current protocols to differentiate human pluripotent stem cells (PSC) to NCC are biased toward cranial NCC. Addition of retinoic acid drove trunk-related markers and HOX genes characteristic of a posterior identity. Subsequent treatment with bone morphogenetic proteins (BMPs) enhanced differentiation to sympathoadrenal cells. Our approach provides methodology for detailed studies of human NCC, and clarifies roles for retinoids and BMPs in the differentiation of human PSC to trunk NCC and to sympathoadrenal lineages.
- Published
- 2016
39. Efficient CRISPR/Cas9‐Based Genome Engineering in Human Pluripotent Stem Cells
- Author
-
Kime, Cody, Mandegar, Mohammad A, Srivastava, Deepak, Yamanaka, Shinya, Conklin, Bruce R, and Rand, Tim A
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Stem Cell Research - Induced Pluripotent Stem Cell ,Stem Cell Research ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Human Genome ,Biotechnology ,Stem Cell Research - Embryonic - Human ,Regenerative Medicine ,Genetics ,Stem Cell Research - Nonembryonic - Human ,Generic health relevance ,CRISPR-Cas Systems ,Cell Culture Techniques ,Cells ,Cultured ,Genetic Engineering ,Genome ,Human ,Humans ,INDEL Mutation ,Pluripotent Stem Cells ,Reproducibility of Results ,CRISPR ,Cas9 ,genomic engineering ,human pluripotent stem cells ,Genetics & Heredity - Abstract
Human pluripotent stem cells (hPS cells) are rapidly emerging as a powerful tool for biomedical discovery. The advent of human induced pluripotent stem cells (hiPS cells) with human embryonic stem (hES)-cell-like properties has led to hPS cells with disease-specific genetic backgrounds for in vitro disease modeling and drug discovery as well as mechanistic and developmental studies. To fully realize this potential, it will be necessary to modify the genome of hPS cells with precision and flexibility. Pioneering experiments utilizing site-specific double-strand break (DSB)-mediated genome engineering tools, including zinc finger nucleases (ZFNs) and transcription activator-like effector nucleases (TALENs), have paved the way to genome engineering in previously recalcitrant systems such as hPS cells. However, these methods are technically cumbersome and require significant expertise, which has limited adoption. A major recent advance involving the clustered regularly interspaced short palindromic repeats (CRISPR) endonuclease has dramatically simplified the effort required for genome engineering and will likely be adopted widely as the most rapid and flexible system for genome editing in hPS cells. In this unit, we describe commonly practiced methods for CRISPR endonuclease genomic editing of hPS cells into cell lines containing genomes altered by insertion/deletion (indel) mutagenesis or insertion of recombinant genomic DNA.
- Published
- 2016
40. Self-organizing human cardiac microchambers mediated by geometric confinement.
- Author
-
Ma, Zhen, Wang, Jason, Loskill, Peter, Huebsch, Nathaniel, Koo, Sangmo, Svedlund, Felicia L, Marks, Natalie C, Hua, Ethan W, Grigoropoulos, Costas P, Conklin, Bruce R, and Healy, Kevin E
- Subjects
Myocardium ,Heart ,Myocytes ,Cardiac ,Humans ,Cadherins ,Cell Count ,Cues ,Cell Differentiation ,Cell Proliferation ,Cell Movement ,Cell Lineage ,Morphogenesis ,Body Patterning ,Stress ,Mechanical ,Models ,Cardiovascular ,Induced Pluripotent Stem Cells ,Myofibroblasts ,Epithelial-Mesenchymal Transition ,Wnt Signaling Pathway ,In Vitro Techniques ,Heart Disease ,Cardiovascular ,Regenerative Medicine ,Stem Cell Research - Embryonic - Non-Human ,Stem Cell Research ,1.1 Normal biological development and functioning ,Myocytes ,Cardiac ,Stress ,Mechanical ,Models - Abstract
Tissue morphogenesis and organ formation are the consequences of biochemical and biophysical cues that lead to cellular spatial patterning in development. To model such events in vitro, we use PEG-patterned substrates to geometrically confine human pluripotent stem cell colonies and spatially present mechanical stress. Modulation of the WNT/β-catenin pathway promotes spatial patterning via geometric confinement of the cell condensation process during epithelial-mesenchymal transition, forcing cells at the perimeter to express an OCT4+ annulus, which is coincident with a region of higher cell density and E-cadherin expression. The biochemical and biophysical cues synergistically induce self-organizing lineage specification and creation of a beating human cardiac microchamber confined by the pattern geometry. These highly defined human cardiac microchambers can be used to study aspects of embryonic spatial patterning, early cardiac development and drug-induced developmental toxicity.
- Published
- 2015
41. Automated Video-Based Analysis of Contractility and Calcium Flux in Human-Induced Pluripotent Stem Cell-Derived Cardiomyocytes Cultured over Different Spatial Scales
- Author
-
Huebsch, Nathaniel, Loskill, Peter, Mandegar, Mohammad A, Marks, Natalie C, Sheehan, Alice S, Ma, Zhen, Mathur, Anurag, Nguyen, Trieu N, Yoo, Jennie C, Judge, Luke M, Spencer, C Ian, Chukka, Anand C, Russell, Caitlin R, So, Po-Lin, Conklin, Bruce R, and Healy, Kevin E
- Subjects
Engineering ,Biomedical Engineering ,Stem Cell Research - Induced Pluripotent Stem Cell ,Stem Cell Research ,Bioengineering ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Regenerative Medicine ,Algorithms ,Calcium ,Cell Differentiation ,Cells ,Cultured ,Humans ,Image Processing ,Computer-Assisted ,Induced Pluripotent Stem Cells ,Microscopy ,Video ,Myocardial Contraction ,Myocytes ,Cardiac ,Patch-Clamp Techniques ,Pattern Recognition ,Automated ,Signal Transduction ,Signal-To-Noise Ratio ,Software ,Biochemistry and Cell Biology ,Biomedical engineering - Abstract
Contractile motion is the simplest metric of cardiomyocyte health in vitro, but unbiased quantification is challenging. We describe a rapid automated method, requiring only standard video microscopy, to analyze the contractility of human-induced pluripotent stem cell-derived cardiomyocytes (iPS-CM). New algorithms for generating and filtering motion vectors combined with a newly developed isogenic iPSC line harboring genetically encoded calcium indicator, GCaMP6f, allow simultaneous user-independent measurement and analysis of the coupling between calcium flux and contractility. The relative performance of these algorithms, in terms of improving signal to noise, was tested. Applying these algorithms allowed analysis of contractility in iPS-CM cultured over multiple spatial scales from single cells to three-dimensional constructs. This open source software was validated with analysis of isoproterenol response in these cells, and can be applied in future studies comparing the drug responsiveness of iPS-CM cultured in different microenvironments in the context of tissue engineering.
- Published
- 2015
42. Structure-based discovery of NANOG variant with enhanced properties to promote self-renewal and reprogramming of pluripotent stem cells
- Author
-
Hayashi, Yohei, Caboni, Laura, Das, Debanu, Yumoto, Fumiaki, Clayton, Thomas, Deller, Marc C, Nguyen, Phuong, Farr, Carol L, Chiu, Hsiu-Ju, Miller, Mitchell D, Elsliger, Marc-André, Deacon, Ashley M, Godzik, Adam, Lesley, Scott A, Tomoda, Kiichiro, Conklin, Bruce R, Wilson, Ian A, Yamanaka, Shinya, and Fletterick, Robert J
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Stem Cell Research ,Stem Cell Research - Embryonic - Non-Human ,Genetics ,Stem Cell Research - Induced Pluripotent Stem Cell ,Regenerative Medicine ,1.1 Normal biological development and functioning ,Generic health relevance ,Amino Acid Sequence ,Animals ,Base Sequence ,Cell Line ,Cell Proliferation ,Cells ,Cultured ,Cellular Reprogramming ,Crystallography ,X-Ray ,DNA ,Embryonic Stem Cells ,Germ Layers ,Homeodomain Proteins ,Humans ,Induced Pluripotent Stem Cells ,Mice ,Inbred C57BL ,Models ,Molecular ,Molecular Sequence Data ,Mutation ,Nanog Homeobox Protein ,Nucleic Acid Conformation ,Pluripotent Stem Cells ,Promoter Regions ,Genetic ,Protein Binding ,Protein Structure ,Tertiary ,Transfection ,NANOG ,crystal structure ,pluripotent stem cells ,DNA-binding ,reprogramming - Abstract
NANOG (from Irish mythology Tír na nÓg) transcription factor plays a central role in maintaining pluripotency, cooperating with OCT4 (also known as POU5F1 or OCT3/4), SOX2, and other pluripotency factors. Although the physiological roles of the NANOG protein have been extensively explored, biochemical and biophysical properties in relation to its structural analysis are poorly understood. Here we determined the crystal structure of the human NANOG homeodomain (hNANOG HD) bound to an OCT4 promoter DNA, which revealed amino acid residues involved in DNA recognition that are likely to be functionally important. We generated a series of hNANOG HD alanine substitution mutants based on the protein-DNA interaction and evolutionary conservation and determined their biological activities. Some mutant proteins were less stable, resulting in loss or decreased affinity for DNA binding. Overexpression of the orthologous mouse NANOG (mNANOG) mutants failed to maintain self-renewal of mouse embryonic stem cells without leukemia inhibitory factor. These results suggest that these residues are critical for NANOG transcriptional activity. Interestingly, one mutant, hNANOG L122A, conversely enhanced protein stability and DNA-binding affinity. The mNANOG L122A, when overexpressed in mouse embryonic stem cells, maintained their expression of self-renewal markers even when retinoic acid was added to forcibly drive differentiation. When overexpressed in epiblast stem cells or human induced pluripotent stem cells, the L122A mutants enhanced reprogramming into ground-state pluripotency. These findings demonstrate that structural and biophysical information on key transcriptional factors provides insights into the manipulation of stem cell behaviors and a framework for rational protein engineering.
- Published
- 2015
43. A Non-invasive Platform for Functional Characterization of Stem-Cell-Derived Cardiomyocytes with Applications in Cardiotoxicity Testing
- Author
-
Maddah, Mahnaz, Heidmann, Julia D, Mandegar, Mohammad A, Walker, Chase D, Bolouki, Sara, Conklin, Bruce R, and Loewke, Kevin E
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Heart Disease ,Regenerative Medicine ,Stem Cell Research ,Stem Cell Research - Induced Pluripotent Stem Cell ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Cardiovascular ,Calcium ,Calcium Signaling ,Cardiotoxicity ,Cell Culture Techniques ,Cell Differentiation ,Drug Evaluation ,Preclinical ,High-Throughput Screening Assays ,Humans ,Microscopy ,Video ,Myocytes ,Cardiac ,Patch-Clamp Techniques ,Stem Cells ,Clinical Sciences ,Biochemistry and cell biology - Abstract
We present a non-invasive method to characterize the function of pluripotent stem-cell-derived cardiomyocytes based on video microscopy and image analysis. The platform, called Pulse, generates automated measurements of beating frequency, beat duration, amplitude, and beat-to-beat variation based on motion analysis of phase-contrast images captured at a fast frame rate. Using Pulse, we demonstrate recapitulation of drug effects in stem-cell-derived cardiomyocytes without the use of exogenous labels and show that our platform can be used for high-throughput cardiotoxicity drug screening and studying physiologically relevant phenotypes.
- Published
- 2015
44. Human iPSC-based cardiac microphysiological system for drug screening applications.
- Author
-
Mathur, Anurag, Loskill, Peter, Shao, Kaifeng, Huebsch, Nathaniel, Hong, SoonGweon, Marcus, Sivan G, Marks, Natalie, Mandegar, Mohammad, Conklin, Bruce R, Lee, Luke P, and Healy, Kevin E
- Subjects
Cells ,Cultured ,Myocytes ,Cardiac ,Humans ,Cardiovascular Agents ,Biological Assay ,Flow Injection Analysis ,Tissue Array Analysis ,Equipment Design ,Equipment Failure Analysis ,Drug Evaluation ,Preclinical ,Cell Differentiation ,Lab-On-A-Chip Devices ,Induced Pluripotent Stem Cells ,Cells ,Cultured ,Myocytes ,Cardiac ,Drug Evaluation ,Preclinical ,Regenerative Medicine ,Cardiovascular ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Stem Cell Research - Induced Pluripotent Stem Cell ,Heart Disease ,Stem Cell Research ,Biochemistry and Cell Biology ,Other Physical Sciences - Abstract
Drug discovery and development are hampered by high failure rates attributed to the reliance on non-human animal models employed during safety and efficacy testing. A fundamental problem in this inefficient process is that non-human animal models cannot adequately represent human biology. Thus, there is an urgent need for high-content in vitro systems that can better predict drug-induced toxicity. Systems that predict cardiotoxicity are of uppermost significance, as approximately one third of safety-based pharmaceutical withdrawals are due to cardiotoxicty. Here, we present a cardiac microphysiological system (MPS) with the attributes required for an ideal in vitro system to predict cardiotoxicity: i) cells with a human genetic background; ii) physiologically relevant tissue structure (e.g. aligned cells); iii) computationally predictable perfusion mimicking human vasculature; and, iv) multiple modes of analysis (e.g. biological, electrophysiological, and physiological). Our MPS is able to keep human induced pluripotent stem cell derived cardiac tissue viable and functional over multiple weeks. Pharmacological studies using the cardiac MPS show half maximal inhibitory/effective concentration values (IC₅₀/EC₅₀) that are more consistent with the data on tissue scale references compared to cellular scale studies. We anticipate the widespread adoption of MPSs for drug screening and disease modeling.
- Published
- 2015
45. Astrocytic adenosine receptor A2A and Gs-coupled signaling regulate memory.
- Author
-
Orr, Anna G, Hsiao, Edward C, Wang, Max M, Ho, Kaitlyn, Kim, Daniel H, Wang, Xin, Guo, Weikun, Kang, Jing, Yu, Gui-Qiu, Adame, Anthony, Devidze, Nino, Dubal, Dena B, Masliah, Eliezer, Conklin, Bruce R, and Mucke, Lennart
- Subjects
Astrocytes ,Animals ,Mice ,Inbred C57BL ,Animals ,Newborn ,Mice ,Transgenic ,Humans ,Mice ,Alzheimer Disease ,Sulfonamides ,Indoles ,Glial Fibrillary Acidic Protein ,Cytoskeletal Proteins ,Receptor ,Adenosine A2A ,Receptors ,Serotonin ,5-HT4 ,Nerve Tissue Proteins ,Serotonin Antagonists ,Exploratory Behavior ,Maze Learning ,Signal Transduction ,Gene Expression Regulation ,Memory ,Long-Term ,Recognition ,Psychology ,Alzheimer's Disease including Alzheimer's Disease Related Dementias (AD/ADRD) ,Aging ,Neurodegenerative ,Dementia ,Brain Disorders ,Neurosciences ,Acquired Cognitive Impairment ,Alzheimer's Disease ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Neurological ,Neurology & Neurosurgery ,Psychology ,Cognitive Sciences - Abstract
Astrocytes express a variety of G protein-coupled receptors and might influence cognitive functions, such as learning and memory. However, the roles of astrocytic Gs-coupled receptors in cognitive function are not known. We found that humans with Alzheimer's disease (AD) had increased levels of the Gs-coupled adenosine receptor A2A in astrocytes. Conditional genetic removal of these receptors enhanced long-term memory in young and aging mice and increased the levels of Arc (also known as Arg3.1), an immediate-early gene that is required for long-term memory. Chemogenetic activation of astrocytic Gs-coupled signaling reduced long-term memory in mice without affecting learning. Like humans with AD, aging mice expressing human amyloid precursor protein (hAPP) showed increased levels of astrocytic A2A receptors. Conditional genetic removal of these receptors enhanced memory in aging hAPP mice. Together, these findings establish a regulatory role for astrocytic Gs-coupled receptors in memory and suggest that AD-linked increases in astrocytic A2A receptor levels contribute to memory loss.
- Published
- 2015
46. Classifying the Electrophysiological Effects of Chronotropic Drugs on Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes using Voltage Sensitive Dyes and Supervised Machine Learning
- Author
-
Heylman, Christopher M, Datta, Rupsa, Conklin, Bruce R, George, Steven C, and Gratton, Enrico
- Subjects
Biophysics ,Physical Sciences ,Chemical Sciences ,Biological Sciences - Published
- 2015
47. Dual α-globin and truncated erythropoietin receptor knock-in restores hemoglobin production in α-thalassemia major-derived hematopoietic stem and progenitor cells
- Author
-
Chu, Simon N, primary, Soupene, Eric, additional, Wienert, Beeke, additional, Yin, Han, additional, Sharma, Devesh, additional, Jia, Kun, additional, Homma, Shota, additional, Hampton, Jessica P, additional, Gardner, James M, additional, Conklin, Bruce R, additional, MacKenzie, Tippi C, additional, Porteus, Matthew H, additional, and Cromer, M. Kyle, additional
- Published
- 2023
- Full Text
- View/download PDF
48. On the road to a gene drive in mammals
- Author
-
Conklin, Bruce R.
- Published
- 2019
- Full Text
- View/download PDF
49. Detection and Quantification of HDR and NHEJ Induced by Genome Editing at Endogenous Gene Loci Using Droplet Digital PCR
- Author
-
Miyaoka, Yuichiro, primary, Mayerl, Steven J., additional, Chan, Amanda H., additional, and Conklin, Bruce R., additional
- Published
- 2018
- Full Text
- View/download PDF
50. Evaluation of CRISPR/Cas9 exon‐skipping vector for choroideremia using human induced pluripotent stem cell‐derived RPE
- Author
-
Iwagawa, Toshiro, primary, Masumoto, Hiroki, additional, Tabuchi, Hitoshi, additional, Tani, Kenzaburo, additional, Conklin, Bruce R., additional, and Watanabe, Sumiko, additional
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.