Skip to main content
Clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein 9 nuclease (Cas9)-based transcriptional repressors (CRISPRi) have emerged as powerful tools for functional epigenetic silencing.1 They rely on a nuclease-deficient Cas9 fused to a repressor domain (dCas9i) and a target-specific guide RNA. In contrast to gene editing approaches, dCas9-KRAB (Krüppel associated box domain)–mediated CRISPRi is generally reversible and titratable, and circumvents the risks associated with direct modification of the genomic sequence.
Meet the First Author, see p 137
Adeno-associated viruses (AAVs) are promising nonintegrating vectors for gene therapy, but their limited packaging capacity makes them incompatible with large inserts such as the common S. pyogenes dCas9i. Here, we describe a CRISPR-based gene-silencing method in the heart relying on a single compact vector compatible with AAV delivery (AAVi).
We engineered an AAV vector containing both an S. aureus–derived nuclease-deficient Cas9 fused to a KRAB repressor domain2 driven by a cytomegalovirus promoter and a guide RNA cassette under the control of the U6 promoter (Figure [A]). The cytomegalovirus promoter ensures robust expression and can be exchanged if cell type-specific expression is required. Organ and cell type selectivity can further be modulated by the selected capsid serotypes as shown for muscle tissue.3 The insert size of the engineered AAVi construct is 4.7 kb, reaching the upper limit insert size of the AAV genome. Nevertheless, the yields of AAV6 and AAV9 particles were comparable to those produced using an AAV vector with a smaller (3 kb) insert size (Figure [B]).
Figure. Functional epigenetic silencing using adeno-associated virus (AAV)-CRISPRi. A, AAVi (AAV delivery) of CRISPRi (clustered regularly interspaced short palindromic repeats-based repression). B, Quantitative real-time PCR (qPCR) quantification of AAVi and reference (insert size, 3 kb) particle yields. C, AAVi dose-response curve for the Nppa promoter (AAViNppa) and a control construct (AAViCtrl) in mouse HL-1 cells as measured by RT-qPCR (reverse transcription qPCR). The half-maximal inhibitory concentration (IC50±SEM) and the goodness of fit (r2) are shown (left plot). The effect of AAViNppa on chromatin accessibility (ATAC) formation of trimethylated lysine 9 of histone H3 (H3K9me3), and gene expression (RNA) in HL-1 cells is displayed for the Nppa locus (middle plot) and all promoters (±2 kb) for H3K9me3 (right plot). D, RT-qPCR measurement of AAVi-mediated gene silencing in neonatal rat cardiomyocytes (NRVM; left), human cardioids (middle), and human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CM, right) using a single guide RNA (gRNA) except for NPPA where data was merged from two. Immuno-labeling of NPPA (red) in human cardioids and visualization of cardiomyocyte-specific MYL7-GFP (myosin light chain 7-driven green fluorescence protein) fluorescence (green). Western blot analysis and quantification of NPPA protein levels on AAViNppa in human iPSC-CM. E, AAVi in vivo timeline. Phenylephrine (Phe)- and angiotensin II (AngII)-releasing osmotic pumps induced cardiac hypertrophy. RT-qPCR analysis of Nppa expression in ventricular tissue (left). Single nuclear RNA-seq analysis of Nppa and Tnnt2 (troponin T2) expression. F, Original traces of chromatin accessibility (ATAC-seq) in sorted cardiomyocyte nuclei. Shown are mean±SEM and n numbers. P values (Mann-Whitney U test) were calculated for all comparisons. Multiple testing was corrected in D (6 tests) and E (2 tests; q=5%; Benjamini, Krieger, and Yekutieli). Prism10 was used. CMV indicates cytomegalovirus; GAPDH, glyceraldehyde-3-phosphate-dehydrogenase; and KRAB, Krüppel associated box domain.
To show the functionality of AAVi for cardiac research, we silenced the NPPA (natriuretic peptide A) because NPPA is highly and specifically expressed in cardiomyocytes. We designed guide RNAs targeting the accessible Nppa proximal promoter region (AAViNppa; Figure [A]) or without homology to mammalian genomes (AAViCtrl). We packaged them into AAV6 particles to transduce cultured mouse HL-1 cardiomyocytes. AAViNppa reduced Nppa gene expression in HL-1 cells in a dose-dependent manner after 1 week (Figure [C]). Due to cell proliferation–associated AAV dilution, the effect is then progressively lost (data not shown) as AAVi-mediated silencing is not heritable. Epigenome analysis using ATAC-seq (assay for transposase-accessible chromatin using sequencing) and ChIP-seq (chromatin Immunoprecipitation-sequencing) showed local loss of chromatin accessibility and a stretch of trimethylated histone H3 Lys-9 (H3K9me3)±2 kb around the target site, orchestrating loss of transcriptional activity (RNA sequencing) and confirming successful epigenetic silencing (Figure [C]). A genome-wide analysis of gene promoters revealed that de novo deposition of the heterochromatin mark was specific to the target promoter region of Nppa and did not spread to known interacting regions such as the Nppb promoter and the shared superenhancer (Figure [C])4. One H3K9me3 positive off-target site was identified at a nonregulatory site of the genome homologous with the 3′half of the spacer. We then confirmed the broad applicability of AAVi to different cardiomyocyte cell culture systems by silencing several genes in primary neonatal rat ventricular cardiomyocytes, human induced pluripotent stem cells (iPSC)-derived cardioids, and iPSC-cardiomyocytes (Figure [D]). For human iPSC–derived cells, we confirmed the effect of AAVi on NPPA RNA and protein levels, as representatively shown for cardioids by immunofluorescence and quantitatively for iPSC-cardiomyocytes by Western blot analysis. In total, 80% (17 of 21) of tested guide RNAs permitted efficient gene silencing documenting the high efficacy of AAVi.
Nppa transcription is induced during cardiac stress and heart disease. To demonstrate the robustness of AAVi in vivo, we aimed to antagonize this process in an experimental heart disease model. Therefore, we retro-orbitally injected 8-week-old male C57Bl/6 mice with a single dose of AAViNppa or AAViCtrl (n=6–7, 3x10E12 AAV9 VG/mouse). Seven days later, we implanted osmotic minipumps releasing phenylephrine (50 mg/kg per day) and angiotensin II (450 µg/kg per day) over 2 weeks to mimic neurohormonal stress signals and induce cardiac hypertrophy. AAViNppa resulted in >7-fold downregulation of ventricular Nppa expression (Figure [E]). Single-nuclei RNA-seq analysis of cardiac nuclei further revealed that Nppa expression is detectable by snRNA-seq in 3.5% (41/1165) of cardiomyocyte nuclei (1151/1165 Tnnt2+) from mice treated with AAViNppa compared with 95% (864/913) on AAViCtrl (770/913 Tnnt2+), demonstrating homogenous downregulation of Nppa (Figure [E]). Notably, the efficacy of AAVi exceeded a previous study reporting a 32% reduction of Pcsk9 mRNA expression in mouse liver using an alternative single AAV-CRISPRi system.5 To show the effect on the epigenetic layer and the specificity of AAVi, we sorted cardiomyocyte nuclei from diseased hearts and analyzed changes in chromatin accessibility (ATAC-seq) and gene expression (nuclear RNA sequencing) on a genome-wide scale. AAViNppa established closed chromatin at the Nppa promoter (Figure [F]), and Nppa was downregulated, while accessibility and expression of neighboring genes were not affected. Besides Nppa, we found only 25 genes mainly linked to antiviral response to be affected (n=3; q≤0.001).
In summary, we present an efficient AAV-based method for CRISPRi-mediated epigenetic silencing of gene expression in cardiac myocytes in vivo and in vitro. This functional epigenetic approach provides a novel and efficient way to modulate gene expression in the heart and could become a standard method for cardiovascular disease modeling and translational research.

ARTICLE INFORMATION

Data Availability

Data are available on request from the corresponding author.

Acknowledgments

The authors thank Sabine Brummer, Joshua Hartmann, and Núria Díaz i Pedrosa for their technical assistance and Benjamin Meder (Institute for Cardiomyopathies, Heidelberg) for providing access to Novaseq 6000. The authors thank Sasha Mendjan (IMBA, Vienna, Austria; Mercator Fellow of CRC1550) for help with establishing the generation of cardioids in the laboratory of E.E.M. Furlong.

REFERENCES

1.
Qi LS, Larson MH, Gilbert LA, Doudna JA, Weissman JS, Arkin AP, Lim WA. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell. 2013;152:1173–1183. doi: 10.1016/j.cell.2013.02.022
2.
Thakore PI, Kwon JB, Nelson CE, Rouse DC, Gemberling MP, Oliver ML, Gersbach CA. RNA-guided transcriptional silencing in vivo with S. aureus CRISPR-Cas9 repressors. Nat Commun. 2018;9:1674. doi: 10.1038/s41467-018-04048-4
3.
Weinmann J, Weis S, Sippel J, Tulalamba W, Remes A, El Andari J, Herrmann AK, Pham QH, Borowski C, Hille S, et al. Identification of a myotropic AAV by massively parallel in vivo evaluation of barcoded capsid variants. Nat Commun. 2020;11:5432. doi: 10.1038/s41467-020-19230-w
4.
Man JCK, van Duijvenboden K, Krijger PHL, Hooijkaas IB, van der Made I, de Gier-de Vries C, Wakker V, Creemers EE, de Laat W, Boukens BJ, et al. Genetic dissection of a super enhancer controlling the Nppa-Nppb cluster in the heart. Circ Res. 2021;128:115–129. doi: 10.1161/CIRCRESAHA.120.317045
5.
Backstrom JR, Sheng J, Wang MC, Bernardo-Colon A, Rex TS. Optimization of S. aureus dCas9 and CRISPRi elements for a single adeno-associated virus that targets an endogenous gene. Mol Ther Methods Clin Dev. 2020;19:139–148. doi: 10.1016/j.omtm.2020.09.001

eLetters(0)

eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.

Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.

Information & Authors

Information

Published In

Go to Circulation Research
Go to Circulation Research
Circulation Research
Pages: 223 - 225
PubMed: 38131200

Versions

You are viewing the most recent version of this article.

History

Published online: 22 December 2023
Published in print: 19 January 2024

Permissions

Request permissions for this article.

Keywords

  1. epigenetic repression
  2. gene editing
  3. gene silencing
  4. genetic therapy
  5. genomics

Subjects

Authors

Affiliations

Institute of Experimental Cardiology, Heidelberg University Hospital, Germany (P.L., C.C., T.L., S.K., R.G.).
DZHK (German Center for Cardiovascular Research) Partner Site Heidelberg/Mannheim, Germany (P.L., C.C., R.G., E.E.M.F.).
DZHK Partner Site Rhein/Main, Germany (P.L., C.C., L.W., F.R., R.G.).
Institute of Cardiovascular Physiology, Frankfurt University, Germany (P.L., C.C., L.W., F.R., R.G.).
Institute of Experimental Cardiology, Heidelberg University Hospital, Germany (P.L., C.C., T.L., S.K., R.G.).
DZHK (German Center for Cardiovascular Research) Partner Site Heidelberg/Mannheim, Germany (P.L., C.C., R.G., E.E.M.F.).
DZHK Partner Site Rhein/Main, Germany (P.L., C.C., L.W., F.R., R.G.).
Institute of Cardiovascular Physiology, Frankfurt University, Germany (P.L., C.C., L.W., F.R., R.G.).
DZHK Partner Site München, Germany (D.R, S.E.).
Institute of Pharmacology and Toxicology, Technical University of Munich, Germany (D.R., S.E.).
M. Schwaderer
Institute of Clinical and Experimental Pharmacology and Toxicology, University of Freiburg, Germany (M.S.).
T. Lueneburg
Institute of Experimental Cardiology, Heidelberg University Hospital, Germany (P.L., C.C., T.L., S.K., R.G.).
S. Kuss
Institute of Experimental Cardiology, Heidelberg University Hospital, Germany (P.L., C.C., T.L., S.K., R.G.).
DZHK Partner Site Rhein/Main, Germany (P.L., C.C., L.W., F.R., R.G.).
Institute of Cardiovascular Physiology, Frankfurt University, Germany (P.L., C.C., L.W., F.R., R.G.).
European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany (R.D., E.E.M.F.).
DZHK (German Center for Cardiovascular Research) Partner Site Heidelberg/Mannheim, Germany (P.L., C.C., R.G., E.E.M.F.).
European Molecular Biology Laboratory, Genome Biology Unit, Heidelberg, Germany (R.D., E.E.M.F.).
F. Rezende
DZHK Partner Site Rhein/Main, Germany (P.L., C.C., L.W., F.R., R.G.).
Institute of Cardiovascular Physiology, Frankfurt University, Germany (P.L., C.C., L.W., F.R., R.G.).
DZHK Partner Site München, Germany (D.R, S.E.).
Institute of Pharmacology and Toxicology, Technical University of Munich, Germany (D.R., S.E.).
Institute of Experimental Cardiology, Heidelberg University Hospital, Germany (P.L., C.C., T.L., S.K., R.G.).
DZHK (German Center for Cardiovascular Research) Partner Site Heidelberg/Mannheim, Germany (P.L., C.C., R.G., E.E.M.F.).
DZHK Partner Site Rhein/Main, Germany (P.L., C.C., L.W., F.R., R.G.).
Institute of Cardiovascular Physiology, Frankfurt University, Germany (P.L., C.C., L.W., F.R., R.G.).

Notes

For Sources of Funding and Disclosures, see page 225.
Correspondence to: R. Gilsbach, PhD, Institute of Experimental Cardiology, Heidelberg University Hospital, Im Neuenheimer Feld 669, 69120 Heidelberg, Germany. Email [email protected]

Disclosures

Disclosures None.

Funding Information

Deutsche Forschungsgemeinschaft (DE)501100001659
Deutsche Forschungsgemeinschaft (DE)501100001659: SFB1425 to RG
China Scholarship Council (CN)501100004543: 202008410206
Dr. Rolf M. Schwiete Stiftung (DE)501100020027: Brandes 02/2018
Deutsches Zentrum für Herz-Kreislaufforschung (DE)100010447
Deutsche Forschungsgemeinschaft (DE)501100001659: TRR267 to SE
Bundesministerium für Bildung und Forschung (DE)501100002347: CNATM to SE
C. Cao was supported by the China Scholarship Council (202008410206). This study was supported by the German Research Foundation (DFG) Collaborative Research Centers 1425 (project-ID 422681845, projects P02 and S03 to R. Gilsbach) and 1550 (project-ID 464424253, project A02 to R. Gilsbach and project A04 to E.E.M. Furlong), the Rolf M. Schwiete Stiftung (Mannheim, Germany) to R. Gilsbach and the DZHK (German Center for Cardiovasular Research, project 81X450019 to P. Laurette, D. Ramanujam, S. Engelhardt, and R. Gilsbach). S. Engelhardt was supported by the German Research Foundation (DFG, project ID 403584255–TRR267) and the Federal Ministry of Education and Research (BMBF) in the framework of the Cluster4future program (CNATM - Cluster for Nucleic Acid Therapeutics Munich). The authors acknowledge the support of the Freiburg Galaxy Team, Rolf Backofen and Björn Grüning, Bioinformatics, University of Freiburg (Germany), funded by the German Federal Ministry of Education and Research (grant 031 A538A de.NBI-RBC), CRC1425, and CRC992. The authors acknowledge the data storage service SDS@hd supported by the Ministry of Science, Research and Arts Baden-Württemberg and the German Research Foundation (DFG) through grant INST 35/1503-1 FUGG.

Metrics & Citations

Metrics

Citations

Download Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Select your manager software from the list below and click Download.

  1. Catecholamine treatment induces reversible heart injury and cardiomyocyte gene expression, Intensive Care Medicine Experimental, 12, 1, (2024).https://doi.org/10.1186/s40635-024-00632-9
    Crossref
Loading...

View Options

View options

PDF and All Supplements

Download PDF and All Supplements

PDF/EPUB

View PDF/EPUB
Login options

Check if you have access through your login credentials or your institution to get full access on this article.

Personal login Institutional Login
Purchase Options

Purchase this article to access the full text.

Purchase access to this article for 24 hours

In Vivo Silencing of Regulatory Elements Using a Single AAV-CRISPRi Vector
Circulation Research
  • Vol. 134
  • No. 2

Purchase access to this journal for 24 hours

Circulation Research
  • Vol. 134
  • No. 2
Restore your content access

Enter your email address to restore your content access:

Note: This functionality works only for purchases done as a guest. If you already have an account, log in to access the content to which you are entitled.

Figures

Tables

Media

Share

Share

Share article link

Share

Comment Response