Genome Editing in Cardiovascular Biology

生物 基因组编辑 计算生物学 基因组 进化生物学 遗传学 基因
作者
Timon Seeger,Matthew H. Porteus,Joseph C. Wu
出处
期刊:Circulation Research [Ovid Technologies (Wolters Kluwer)]
卷期号:120 (5): 778-780 被引量:46
标识
DOI:10.1161/circresaha.116.310197
摘要

HomeCirculation ResearchVol. 120, No. 5Genome Editing in Cardiovascular Biology Free AccessArticle CommentaryPDF/EPUBAboutView PDFView EPUBSections ToolsAdd to favoritesDownload citationsTrack citationsPermissions ShareShare onFacebookTwitterLinked InMendeleyReddit Jump toFree AccessArticle CommentaryPDF/EPUBGenome Editing in Cardiovascular Biology Timon Seeger, Matthew Porteus and Joseph C. Wu Timon SeegerTimon Seeger From the Stanford Cardiovascular Institute, CA (T.S., J.C.W.); Department of Medicine, Division of Cardiology (T.S., J.S.W.); Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine (M.P.); and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (M.P., J.C.W.). , Matthew PorteusMatthew Porteus From the Stanford Cardiovascular Institute, CA (T.S., J.C.W.); Department of Medicine, Division of Cardiology (T.S., J.S.W.); Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine (M.P.); and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (M.P., J.C.W.). and Joseph C. WuJoseph C. Wu From the Stanford Cardiovascular Institute, CA (T.S., J.C.W.); Department of Medicine, Division of Cardiology (T.S., J.S.W.); Department of Pediatrics, Division of Stem Cell Transplantation and Regenerative Medicine (M.P.); and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, CA (M.P., J.C.W.). Originally published3 Mar 2017https://doi.org/10.1161/CIRCRESAHA.116.310197Circulation Research. 2017;120:778–780Genome editing has emerged as a powerful tool in research and is entering the stage of therapeutic applications. In the cardiovascular field, its role in basic and translational research is well established. However, biological and technical barriers currently hamper the therapeutic potential of genome editing for cardiovascular diseases. This viewpoint discusses possible routes for promoting therapeutic use of genome editing in the cardiovascular system.Genome editing has rapidly emerged as a powerful tool in basic and translational research. Zinc finger nucleases and TALENs (transcription activator-like effector nucleases) catalyzed the field initially. With the development of the CRISPR (clustered regularly interspaced short palindromic repeats)/Cas9 (CRISPR-associated protein 9) system, the field is expanding even more rapidly because of its efficacy, specificity, and ease of use (Figure [A]). Generally, genome editing tools induce site-specific DNA double-strand breaks at a specific genomic site, resulting in the activation of the nonhomologous end-joining (NHEJ) and homologous recombination (HR) cellular endogenous double-strand break repair machinery (Figure [B]). Recent advances of the CRISPR technology also allow for RNA recognition, making it possible to cleave RNA, enhance or inhibit translation, support isolation of specific RNA:protein complexes, or induce specific post-transcriptional modifications.1 Genome editing tools can also be used to control gene expression on the transcriptional level. Deactivation of the catalytic site of CRISPR/Cas9 (dCas9) results in specific binding to the DNA without inducing double-strand breaks. Fusion of dCas9 to DNA-binding domains, such as activator or repressor domains, results in transcriptional activation or inhibition of a specific gene, respectively.2 Despite the major impact genome editing already has on basic and translational research, regulatory processes have delayed therapeutic applications from reaching the clinic. Nevertheless, effective new therapies are anticipated in the near future for various disease conditions, especially if major issues around safety and toxicity are resolved.Download figureDownload PowerPointFigure. Genome editing in cardiovascular development, biology, and therapy. A, The main programmable nucleases are ZFN (zinc finger nucleases), TALEN (transcription activator-like effector nucleases), and CRISPR (clustered regularly interspaced short palindromic repeats). B, They induce double-strand breaks (DSBs) at specific DNA loci leading to nonhomologous end joining (NHEJ), as well as homologous directed repair (HDR). Modified nucleases can be used for transcription interference without causing DSB. Recent advances in CRISPR technology also allow for targeting RNA. C, Genome editing already facilitates the generation of genetically modified animals to study cardiovascular development and biology and plays a major role in disease modeling and drug screening, especially in combination with iPSC technology. However, the role of genome editing in therapeutic approaches for cardiovascular diseases is less evident. Ex vivo and in vivo approaches are possible routes for therapeutic applications. Autologous induced pluripotent stem cells (iPSCs) can be modified ex vivo and differentiated into the desired cell type for cell therapy. Furthermore, mutations might be directly targeted in vivo or excised by exon skipping. c-iPSC indicates corrected iPSC; Cas9, CRISPR-associated protein 9; CM, cardiomyocyte; EC, endothelial cell; HNH, HNH nuclease domain; PAM, protospacer adjacent motif; and RuvC, RuvC nuclease domain.Genome Editing in Preclinical ResearchThe use of genome editing has facilitated basic and translational research across multiple disciplines, including the cardiovascular field. Genome editing already plays a key role in the generation of new genetically modified animal models. In addition, the combination of genome editing and induced pluripotent stem cell (iPSC) technology now makes it possible to elucidate complex pathophysiological mechanisms directly in human cellular models (Figure [C]). Using genome editing to derive isogenic iPSC lines may become the standard approach to characterize disease-causing mechanisms of pathogenic mutations underlying cardiomyopathies, to investigate variants of unknown significance, or to establish disease-modifying variants.3 In addition, high-throughput CRISPR-mediated large-scale functional screens with gene knockout, loss-of-function, and gain-of-function approaches will help to further elucidate molecular mechanisms of cardiovascular diseases. However, the ability to translate insights gained from iPSC-based cellular models into therapeutic strategies still needs to be proven. Further improvements in maturation of iPSC-derived cardiomyocytes (iPSC-CMs) and other derivatives, as well as the generation of organoids or 3-dimensional structures, are needed to fully realize the utility of this technique.4Therapeutic Potential of Genome Editing in Cardiovascular DiseasesGenome editing comprises great potential for innovating therapies. Although its important role in basic and translational research is evident, the barriers to therapeutic applications remain significant. In principle, there will be 2 main routes for therapeutic applications of genome editing in cardiovascular diseases: ex vivo and in vivo approaches (Figure [C]).Ex Vivo Approaches for Genome EditingBecause culturing primary human cardiomyocytes is extremely difficult, the major ex vivo approach for therapeutically applied genome editing in cardiac diseases is to correct iPSCs derived from patients with disease-causing mutations. Studies have shown that iPSC lines generated by CRISPR and TALEN rarely have identifiable off-target mutations.5 These corrected iPSCs can then be differentiated into iPSC-cardiac progenitors or iPSC-CMs and delivered to the patient's heart. Extensive preclinical studies have already been performed to evaluate ways of delivering embryonic stem cell–derived cardiomyocytes (ESC-CM) or iPSC-CMs to the diseased heart, by injecting cells via the intracoronary or intramyocardial route, or as tissue-engineered constructs.6 However, only modest functional improvements have been achieved to date. In addition, several safety concerns exist using ESC-CM or iPSC-CMs in terms of immunogenicity, tumorigenicity, and the risk of arrhythmia because of insufficient electric coupling.7,8 Here, genome editing might be also applied to introduce inducible suicide genes into the genome of the iPSCs or ESC. In the rare case of tumor formation arising from the injected cells, these cells could be targeted directly for elimination.9 A potential downside of the suicide gene approach is that it would also eliminate any cells that might be contributing to positive effects. Moreover, genome editing using ex vivo therapeutic approaches can only be tested once the major obstacles for ESC or iPSC-CM therapy are resolved.In Vivo Approaches for Genome EditingIn vivo genome editing entails correcting disease-causing variants in primary cells in situ. Recently, a smaller Cas9 protein from Staphylococcus aureus has been described that makes it feasible to deliver CRISPR/Cas9 using adeno-associated virus vectors.10 Adeno-associated virus vectors are already approved for cardiac gene therapy and have been tested in clinical trials. Genome editing has safety advantages over integrating viral-based gene therapy methods because it avoids generating large number of random insertions into the genome.Proof-of-concept preclinical mouse studies have also been performed with in vivo genome editing. Using CRISPR/Cas9-mediated NHEJ, the exon in the dystrophin gene carrying a mutation leading to Duchenne muscular dystrophy could be excised, resulting in a significant restoration of muscular function.11 Furthermore, in vivo CRISPR/Cas9-directed HR was sufficient to treat mice with an urea cycle disorder in the enzyme ornithine transcarbamylase, proving the feasibility of an in vivo genome editing approach that uses HR.12However, in vivo therapeutic genome editing within the cardiovascular system faces several major hurdles. First, despite improvements in algorithms predicting the highest specificity, off-target effects can still occur on rare occasions. Subsequently, NHEJ repair mechanisms may lead to cellular dysfunction or give rise to tumor formations. Second, induction of double-strand breaks in the healthy allele in the setting of a heterozygous mutation may result in worsening of the phenotype. Third, the cellular repair mechanisms depend on cell cycle phase. While NHEJ can lead to DNA repair during the G1, S, and G2 phases, natural HR is limited to the late S and G2 phases when the sister chromatid is present.13 Because the vast majority of cardiomyocytes in the postnatal heart are postmitotic, it will take new discoveries to apply HR-mediated editing to postmitotic cells. Nevertheless, NHEJ-mediated exon skipping may be technically applicable especially for mutations in TTN (titin) and MYBPC3 (myosin-binding protein C3).14,15 Fourth, unlike skeletal muscle satellite cells that have huge regenerative potential, the inherent regenerative capacity of an adult heart is highly limited. Thus, the rate of cardiomyocytes successfully being edited has to be relatively high to mediate cardiac improvements. Taken together, these physiological and technical restrictions currently pose significant challenges for in vivo genome editing as a treatment for cardiomyopathies.Beside cardiomyocytes, endothelial cells and smooth muscle cells might become targetable by in vivo genome editing in cardiovascular diseases. One possible scenario is the correction of validated genomic variants in endothelial cells, as identified in genome-wide association studies.16,17 Additional disease-causing or disease-modifying variants will be discovered with increasing availability of whole exome and whole genome sequencing. However, not all of these variants will be in genes specifically expressed in endothelial or smooth muscle cells, so cell-specific delivery of CRISPR/Cas9 will be a major hurdle for clinical use.Cardiac fibroblasts are playing a key role in cardiac homeostasis and most disease conditions in the heart, but the characterization and the therapeutic approaches are limited by the heterogeneity of the cardiac fibroblast population. Ongoing research will further elucidate the contributions of subpopulations and specific gene dysregulations in cardiac diseases, making these cells a valuable source also for gene editing. Again, if cell type–specific delivery can be performed, genome editing can come into play to target distinct subtypes of cardiac fibroblasts to either enhance regeneration or slow down disease progression.Although conceptually genome editing might be applied to germ cells or embryos to correct disease-causing variants, the ethical issues surrounding such an approach remain unresolved, clouding the prospects of its implementation in the near future. Moreover, preimplantation genetic diagnosis with selective embryo implantation, a currently accepted practice (although not covered by insurance), would achieve many of the same goals as those under editing of germ cells or embryos.ConclusionsGenome editing has already established itself as a powerful tool for the generation of new cellular and animal models to investigate pathophysiological mechanisms in various diseases. The therapeutic potential of genome editing for cardiovascular disease is currently hampered by biological (notably the postmitotic nature of cardiomyocytes) and technical barriers. Cardiac fibroblasts, endothelial, and smooth muscle cells might become potential sources of targeted in vivo genome editing in cardiovascular diseases. However, major scientific and technical advances are needed before genome editing can be applied clinically in cardiovascular diseases.AcknowledgmentsWe thank Amy Thomas for her help with preparation of the figure. Because of space limitations, we are unable to include all the important papers relevant to cardiovascular genome editing; we apologize to those investigators whose significant contributions are not mentioned here.Sources of FundingThis publication was supported, in part, by research grants from the German Research Foundation (T.S.), American Heart Association 13EIA14420025, National Institutes of Health (NIH) HL130020, NIH R01 HL128170, and NIH R01 HL126527 (J.C.W.) and the Laurie Kraus Lacob Faculty Scholar Award in Pediatric Translational Research (M.P.).DisclosuresM.P. is a consultant for and has equity in CRISPR Therapeutics.FootnotesThe opinions expressed in this article are not necessarily those of the editors or of the American Heart Association.Correspondence to Joseph C. Wu, MD, PhD, 265 Campus Dr, Room G1120, Stanford, CA 94305. E-mail [email protected]References1. O'Connell MR, Oakes BL, Sternberg SH, East-Seletsky A, Kaplan M, Doudna JA. Programmable RNA recognition and cleavage by CRISPR/Cas9.Nature. 2014; 516:263–266. doi: 10.1038/nature13769.CrossrefMedlineGoogle Scholar2. Gilbert LA, Horlbeck MA, Adamson B, Villalta JE, Chen Y, Whitehead EH, Guimaraes C, Panning B, Ploegh HL, Bassik MC, Qi LS, Kampmann M, Weissman JS. Genome-scale CRISPR-mediated control of gene repression and activation.Cell. 2014; 159:647–661. doi: 10.1016/j.cell.2014.09.029.CrossrefMedlineGoogle Scholar3. Matsa E, Ahrens JH, Wu JC. Human induced pluripotent stem cells as a platform for personalized and precision cardiovascular medicine.Physiol Rev. 2016; 96:1093–1126. doi: 10.1152/physrev.00036.2015.CrossrefMedlineGoogle Scholar4. Tzatzalos E, Abilez OJ, Shukla P, Wu JC. Engineered heart tissues and induced pluripotent stem cells: Macro- and microstructures for disease modeling, drug screening, and translational studies.Adv Drug Deliv Rev. 2016; 96:234–244. doi: 10.1016/j.addr.2015.09.010.CrossrefMedlineGoogle Scholar5. Veres A, Gosis BS, Ding Q, Collins R, Ragavendran A, Brand H, Erdin S, Cowan CA, Talkowski ME, Musunuru K. Low incidence of off-target mutations in individual CRISPR-Cas9 and TALEN targeted human stem cell clones detected by whole-genome sequencing.Cell Stem Cell. 2014; 15:27–30. doi: 10.1016/j.stem.2014.04.020.CrossrefMedlineGoogle Scholar6. Menasché P, Vanneaux V, Hagège A, et al. Human embryonic stem cell-derived cardiac progenitors for severe heart failure treatment: first clinical case report.Eur Heart J. 2015; 36:2011–2017. doi: 10.1093/eurheartj/ehv189.CrossrefMedlineGoogle Scholar7. Neofytou E, O'Brien CG, Couture LA, Wu JC. Hurdles to clinical translation of human induced pluripotent stem cells.J Clin Invest. 2015; 125:2551–2557. doi: 10.1172/JCI80575.CrossrefMedlineGoogle Scholar8. Nguyen PK, Neofytou E, Rhee J-W, Wu JC. Potential strategies to address the major clinical barriers facing stem cell regenerative therapy for cardiovascular disease: areview.JAMA Cardiol. 2016; 1:953–962. doi: 10.1001/jamacardio.2016.2750.CrossrefMedlineGoogle Scholar9. Wang Y, Zhang WY, Hu S, et al. Genome editing of human embryonic stem cells and induced pluripotent stem cells with zinc finger nucleases for cellular imaging.Circ Res. 2012; 111:1494–1503. doi: 10.1161/CIRCRESAHA.112.274969.LinkGoogle Scholar10. Ran FA, Cong L, Yan WX, Scott DA, Gootenberg JS, Kriz AJ, Zetsche B, Shalem O, Wu X, Makarova KS, Koonin EV, Sharp PA, Zhang F. In vivo genome editing using Staphylococcus aureus Cas9.Nature. 2015; 520:186–191. doi: 10.1038/nature14299.CrossrefMedlineGoogle Scholar11. Mendell JR, Rodino-Klapac LR. Duchenne muscular dystrophy: CRISPR/Cas9 treatment.Cell Res. 2016; 26:513–514. doi: 10.1038/cr.2016.28.CrossrefMedlineGoogle Scholar12. Yang Y, Wang L, Bell P, McMenamin D, He Z, White J, Yu H, Xu C, Morizono H, Musunuru K, Batshaw ML, Wilson JM. A dual AAV system enables the Cas9-mediated correction of a metabolic liver disease in newborn mice.Nat Biotechnol. 2016; 34:334–338. doi: 10.1038/nbt.3469.CrossrefMedlineGoogle Scholar13. Heyer WD, Ehmsen KT, Liu J. Regulation of homologous recombination in eukaryotes.Annu Rev Genet. 2010; 44:113–139. doi: 10.1146/annurev-genet-051710-150955.CrossrefMedlineGoogle Scholar14. Gramlich M, Pane LS, Zhou Q, et al. Antisense-mediated exon skipping: a therapeutic strategy for titin-based dilated cardiomyopathy.EMBO Mol Med. 2015; 7:562–576. doi: 10.15252/emmm.201505047.CrossrefMedlineGoogle Scholar15. Gedicke-Hornung C, Behrens-Gawlik V, Reischmann S, et al. Rescue of cardiomyopathy through U7snRNA-mediated exon skipping in Mybpc3-targeted knock-in mice.EMBO Mol Med. 2013; 5:1128–1145. doi: 10.1002/emmm.201202168.CrossrefMedlineGoogle Scholar16. Ehret GB, Ferreira T, Chasman DI, et al; CHARGE-EchoGen Consortium; CHARGE-HF Consortium; Wellcome Trust Case Control Consortium. The genetics of blood pressure regulation and its target organs from association studies in 342,415 individuals.Nat Genet. 2016; 48:1171–1184. doi: 10.1038/ng.3667.CrossrefMedlineGoogle Scholar17. Nikpay M, Goel A, Won HH, et al; CARDIoGRAMplusC4D Consortium. A comprehensive 1,000 Genomes-based genome-wide association meta-analysis of coronary artery disease.Nat Genet. 2015; 47:1121–1130. doi: 10.1038/ng.3396.CrossrefMedlineGoogle Scholar Previous Back to top Next FiguresReferencesRelatedDetailsCited By Fraley C, Milgrom S, Kondapalli L, Taylor M, Mestroni L and Miyamoto S (2021) Mechanisms and Insights for the Development of Heart Failure Associated with Cancer Therapy, Children, 10.3390/children8090829, 8:9, (829) Libby P (2021) The changing landscape of atherosclerosis, Nature, 10.1038/s41586-021-03392-8, 592:7855, (524-533), Online publication date: 22-Apr-2021. Huang J, Feng Q, Wang L and Zhou B (2021) Human Pluripotent Stem Cell-Derived Cardiac Cells: Application in Disease Modeling, Cell Therapy, and Drug Discovery, Frontiers in Cell and Developmental Biology, 10.3389/fcell.2021.655161, 9 Tu C, Cunningham N, Zhang M and Wu J (2021) Human Induced Pluripotent Stem Cells as a Screening Platform for Drug-Induced Vascular Toxicity, Frontiers in Pharmacology, 10.3389/fphar.2021.613837, 12 Lam C and Wu J (2021) Clinical Trial in a Dish, Arteriosclerosis, Thrombosis, and Vascular Biology, 41:3, (1019-1031), Online publication date: 1-Mar-2021. Rezaei H, khadempar S, Farahani N, Hosseingholi E, hayat S, Sathyapalan T and Sahebkar A (2020) Harnessing CRISPR/Cas9 technology in cardiovascular disease, Trends in Cardiovascular Medicine, 10.1016/j.tcm.2019.03.005, 30:2, (93-101), Online publication date: 1-Feb-2020. Pourrier M and Fedida D (2020) The Emergence of Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes (hiPSC-CMs) as a Platform to Model Arrhythmogenic Diseases, International Journal of Molecular Sciences, 10.3390/ijms21020657, 21:2, (657) Neofytou E, Chour T and Wu J (2020) Human pluripotent stem cells for cardiac regeneration Emerging Technologies for Heart Diseases, 10.1016/B978-0-12-813706-2.00012-9, (245-257), . Parrotta E, Scalise S, Scaramuzzino L and Cuda G (2019) Stem Cells: The Game Changers of Human Cardiac Disease Modelling and Regenerative Medicine, International Journal of Molecular Sciences, 10.3390/ijms20225760, 20:22, (5760) Gintant G, Burridge P, Gepstein L, Harding S, Herron T, Hong C, Jalife J and Wu J (2019) Use of Human Induced Pluripotent Stem Cell–Derived Cardiomyocytes in Preclinical Cancer Drug Cardiotoxicity Testing: A Scientific Statement From the American Heart Association, Circulation Research, 125:10, (e75-e92), Online publication date: 25-Oct-2019.Lam C, Tian L, Belbachir N, Wnorowski A, Shrestha R, Ma N, Kitani T, Rhee J and Wu J (2019) Identifying the Transcriptome Signatures of Calcium Channel Blockers in Human Induced Pluripotent Stem Cell–Derived Cardiomyocytes, Circulation Research, 125:2, (212-222), Online publication date: 5-Jul-2019. Sayed N, Ameen M and Wu J (2019) Personalized medicine in cardio-oncology: the role of induced pluripotent stem cell, Cardiovascular Research, 10.1093/cvr/cvz024, 115:5, (949-959), Online publication date: 15-Apr-2019. Guo H, Tian L, Zhang J, Kitani T, Paik D, Lee W and Wu J (2019) Single-Cell RNA Sequencing of Human Embryonic Stem Cell Differentiation Delineates Adverse Effects of Nicotine on Embryonic Development, Stem Cell Reports, 10.1016/j.stemcr.2019.01.022, 12:4, (772-786), Online publication date: 1-Apr-2019. Qin X, Han D and Wu J (2019) Molecular imaging of cardiac regenerative medicine, Current Opinion in Biomedical Engineering, 10.1016/j.cobme.2019.04.006, 9, (66-73), Online publication date: 1-Mar-2019. Ma N, Zhang J, Itzhaki I, Zhang S, Chen H, Haddad F, Kitani T, Wilson K, Tian L, Shrestha R, Wu H, Lam C, Sayed N and Wu J (2018) Determining the Pathogenicity of a Genomic Variant of Uncertain Significance Using CRISPR/Cas9 and Human-Induced Pluripotent Stem Cells, Circulation, 138:23, (2666-2681), Online publication date: 4-Dec-2018. Lam C and Wu J (2018) Disease modelling and drug discovery for hypertrophic cardiomyopathy using pluripotent stem cells: how far have we come?, European Heart Journal, 10.1093/eurheartj/ehy388, 39:43, (3893-3895), Online publication date: 14-Nov-2018. Bruyneel A, McKeithan W, Feyen D and Mercola M (2018) Using iPSC Models to Probe Regulation of Cardiac Ion Channel Function, Current Cardiology Reports, 10.1007/s11886-018-1000-0, 20:7, Online publication date: 1-Jul-2018. Rhee J and Wu J (2018) In vivo genome editing of ANGPTL3: a therapy for atherosclerosis?, Nature Reviews Cardiology, 10.1038/nrcardio.2018.38, 15:5, (259-260), Online publication date: 1-May-2018. Trachana K, Bargaje R, Glusman G, Price N, Huang S and Hood L (2018) Taking Systems Medicine to Heart, Circulation Research, 122:9, (1276-1289), Online publication date: 27-Apr-2018. Liu C, Oikonomopoulos A, Sayed N and Wu J (2018) Modeling human diseases with induced pluripotent stem cells: from 2D to 3D and beyond, Development, 10.1242/dev.156166, 145:5, Online publication date: 1-Mar-2018. Takata N and Eiraku M (2017) Stem cells and genome editing: approaches to tissue regeneration and regenerative medicine, Journal of Human Genetics, 10.1038/s10038-017-0348-0, 63:2, (165-178), Online publication date: 1-Feb-2018. Rhee J and Wu J (2018) Cardiac Cell Cycle Activation as a Strategy to Improve iPSC-Derived Cardiomyocyte Therapy, Circulation Research, 122:1, (14-16), Online publication date: 5-Jan-2018.Chadwick A and Musunuru K (2017) CRISPR-Cas9 Genome Editing for Treatment of Atherogenic Dyslipidemia, Arteriosclerosis, Thrombosis, and Vascular Biology, 38:1, (12-18), Online publication date: 1-Jan-2018. Chadwick A and Musunuru K (2017) Treatment of Dyslipidemia Using CRISPR/Cas9 Genome Editing, Current Atherosclerosis Reports, 10.1007/s11883-017-0668-8, 19:7, Online publication date: 1-Jul-2017. Hulot J (2017) Get Your Cell K.O. in the First Round, Circulation Research, 120:10, (1522-1523), Online publication date: 12-May-2017. Motta B, Pramstaller P, Hicks A and Rossini A (2017) The Impact of CRISPR/Cas9 Technology on Cardiac Research: From Disease Modelling to Therapeutic Approaches, Stem Cells International, 10.1155/2017/8960236, 2017, (1-13), . Martinez-Lage M, Torres-Ruiz R and Rodriguez-Perales S (2017) CRISPR/Cas9 Technology: Applications and Human Disease Modeling CRISPR in Animals and Animal Models, 10.1016/bs.pmbts.2017.09.002, (23-48), . March 3, 2017Vol 120, Issue 5 Advertisement Article InformationMetrics © 2017 American Heart Association, Inc.https://doi.org/10.1161/CIRCRESAHA.116.310197PMID: 28254802 Originally publishedMarch 3, 2017 Keywordsinduced pluripotent stem cellscardiovascular diseasesCRISPRgene editingcardiomyopathiesPDF download Advertisement
最长约 10秒,即可获得该文献文件

科研通智能强力驱动
Strongly Powered by AbleSci AI
更新
大幅提高文件上传限制,最高150M (2024-4-1)

科研通是完全免费的文献互助平台,具备全网最快的应助速度,最高的求助完成率。 对每一个文献求助,科研通都将尽心尽力,给求助人一个满意的交代。
实时播报
1秒前
香蕉觅云应助junyang采纳,获得10
1秒前
忧郁的凝竹完成签到 ,获得积分10
2秒前
鲤鱼眼睛关注了科研通微信公众号
3秒前
烟花应助小名余土土采纳,获得10
3秒前
4秒前
烟花应助科研通管家采纳,获得10
4秒前
小二郎应助科研通管家采纳,获得10
4秒前
bkagyin应助科研通管家采纳,获得10
5秒前
KWang应助科研通管家采纳,获得10
5秒前
汉堡包应助科研通管家采纳,获得10
5秒前
wanci应助科研通管家采纳,获得10
5秒前
Jasper应助科研通管家采纳,获得10
5秒前
Judy发布了新的文献求助10
5秒前
彭于晏应助科研通管家采纳,获得10
5秒前
小马甲应助科研通管家采纳,获得10
6秒前
科研通AI2S应助科研通管家采纳,获得10
6秒前
华仔应助科研通管家采纳,获得10
6秒前
脑洞疼应助科研通管家采纳,获得10
6秒前
6秒前
6秒前
6秒前
6秒前
儒雅冰岚发布了新的文献求助10
7秒前
充电宝应助Youth采纳,获得10
7秒前
南瓜猪猪头完成签到 ,获得积分10
7秒前
杨杨发布了新的文献求助10
8秒前
卿总未梦发布了新的文献求助10
9秒前
寻道图强应助xin采纳,获得30
9秒前
夜白发布了新的文献求助10
9秒前
海清完成签到 ,获得积分10
10秒前
大个应助my采纳,获得10
10秒前
11秒前
12秒前
香蕉觅云应助多多采纳,获得10
15秒前
16秒前
16秒前
kangkirk发布了新的文献求助10
17秒前
18秒前
junyang发布了新的文献求助10
20秒前
高分求助中
Sustainability in Tides Chemistry 1500
TM 5-855-1(Fundamentals of protective design for conventional weapons) 1000
CLSI EP47 Evaluation of Reagent Carryover Effects on Test Results, 1st Edition 800
Threaded Harmony: A Sustainable Approach to Fashion 799
Livre et militantisme : La Cité éditeur 1958-1967 500
Retention of title in secured transactions law from a creditor's perspective: A comparative analysis of selected (non-)functional approaches 500
"Sixth plenary session of the Eighth Central Committee of the Communist Party of China" 400
热门求助领域 (近24小时)
化学 医学 生物 材料科学 工程类 有机化学 生物化学 物理 内科学 纳米技术 计算机科学 化学工程 复合材料 基因 遗传学 催化作用 物理化学 免疫学 量子力学 细胞生物学
热门帖子
关注 科研通微信公众号,转发送积分 3055748
求助须知:如何正确求助?哪些是违规求助? 2712398
关于积分的说明 7431409
捐赠科研通 2357400
什么是DOI,文献DOI怎么找? 1248780
科研通“疑难数据库(出版商)”最低求助积分说明 606786
版权声明 596163