• Keine Ergebnisse gefunden

Generation of two iPSC clones (MHHi021-A and MHHi021-B) from a patient with hypertrophic cardiomyopathy with p.Arg723Gly mutation in the MYH7 gene.

N/A
N/A
Protected

Academic year: 2022

Aktie "Generation of two iPSC clones (MHHi021-A and MHHi021-B) from a patient with hypertrophic cardiomyopathy with p.Arg723Gly mutation in the MYH7 gene."

Copied!
5
0
0

Wird geladen.... (Jetzt Volltext ansehen)

Volltext

(1)

Stem Cell Research 52 (2021) 102208

Available online 3 February 2021

1873-5061/© 2021 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license

Lab Resource: Multiple Cell Lines

Generation of two iPSC clones (MHHi021-A and MHHi021-B) from a patient with hypertrophic cardiomyopathy with p.Arg723Gly mutation in the MYH7 gene

S. Merkert

a,*

, S. Wunderlich

a

, J. Beier

a

, A. Franke

a

, K. Schwanke

a

, G. G ¨ ohring

b

, T. Kraft

c

, A. Francino

d

, R. Zweigerdt

a

, U. Martin

a

aLeibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery (HTTG), REBIRTH-Research Center for Translational and Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany

bDepartment of Human Genetics, Hannover Medical School, 30625 Hannover, Germany

cInstitute of Molecular and Cell Physiology, Hannover Medical School, 30625 Hannover, Germany

dHospital Clinic/IDIBAPS, University of Barcelona, Barcelona, Spain

A B S T R A C T

Hypertrophic cardiomyopathy (HCM) is the most common form of genetic heart disease and is characterized by abnormal thickening of the left ventricular wall and interventricular septum. Here we describe the generation of two induced pluripotent stem cell (iPSC) clones from a HCM patient, heterozygous for the p.Arg723Gly (c.2169C >G) mutation in the MYH7 gene. The generated iPSC clones may provide a useful resource for disease modelling to study the mechanisms underlying HCM pathogenesis in iPSC derived progenies, in particular cardiomyocytes.

Resource Table

Unique stem cell lines

identifier MHHi021-A

MHHi021-B Alternative names of stem

cell lines R723G_K1_MOI10 (MHHi021-A) R723G_K2_MOI1 (MHHi021-B)

Institution LEBAO, Hannover Medical School, Germany Contact information of

distributor zweigerdt.robert@mh-hannover.de martin.ulrich@mh-hannover.de Type of cell lines iPSCs

Origin human

Cell Source Skin fibroblasts

Clonality clonal

Method of

reprogramming lentivirus

Multiline rationale isogenic clones from one patient Gene modification no

Type of modification n/a

Associated disease Hypertrophic Cardiomyopathy, p.Arg723Gly Gene/locus MYH7, 14q11.2, c.2169C >G

Method of modification n/a Name of transgene or

resistance n/a

Inducible/constitutive

system n/a

Date archived/stock date April 2013

(continued on next column)

Resource Table (continued)

Cell line repository/bank https://hpscreg.eu/cell-line/MHHi021-A https://hpscreg.eu/cell-line/MHHi021-B Ethical approval The Local Ethics Committee of Hannover Medical

School approved the study (internal No. 409 and No.

1751-2013) and informed consent was obtained from the patient.

1. Resource utility

Our established MYH7_p.Arg723Gly iPSC clones offer a patient- specific disease model of hypertrophic cardiomyopathy (HCM) and differentiated cardiomyocytes derived from these clones may be used to investigate the molecular mechanisms of HCM as well as their phar- macological response to specific modulators.

2. Resource details

Hypertrophic cardiomyopathy (HCM), the most frequent inherited cardiac disease, is a genetically and phenotypically heterogeneous disease. It is caused by mutations in different genes mostly encoding

* Corresponding author.

E-mail address: merkert.sylvia@mh-hannover.de (S. Merkert).

Contents lists available at ScienceDirect

Stem Cell Research

journal homepage: www.elsevier.com/locate/scr

https://doi.org/10.1016/j.scr.2021.102208

Received 5 November 2020; Received in revised form 12 January 2021; Accepted 24 January 2021

(2)

sarcomeric proteins; in particular heterozygous mutations in myosin binding protein C (MYBPC3) and β-myosin heavy chain (MyHC-Beta/

MYH7). The disease is characterized by thickening of the left ventric- ular wall and interventricular septum, histologically marked by myo- cyte hypertrophy and disarray, as well as interstitial fibrosis (Kraft et al., 2016). To form the foundation for a MYH7 mutation induced in vitro model of HCM (Weber et al., 2016), skin fibroblasts were collected by skin biopsy from a 38-year-old male HCM patient. The patient be- longs to a family with a p.Arg723Gly missense mutation in the essential light chain interacting region of the MYH7 gene, which is associated with sudden death and end-stage heart failure (Enjuto et al., 2000). In the original description of this mutation and its clinical characteristics, our patient is named patient III-1 from family 157 (Enjuto et al., 2000).

The fibroblasts were reprogrammed by overexpression of the codon optimized human pluripotency factors OCT4, SOX2, KLF4 and c-MYC in a 4-in-1 lentiviral vector (Warlich et al., 2011) applying an MOI of 10 or 1. Emerging colonies displaying embryonic stem cell-like morphology were further cultivated clonally. One clone was chosen either from MOI10 (MHHi021-A) or MOI1 (MHHi021-B), respectively, for further characterisation (Tables 1 and 2). To estimate the silencing of the lentiviral reprogramming construct, the expression of the transgenic dTomato was determined, which is co-expressed from the 4- in-1 lentiviral vector. Quantitative real-time PCR analysis found that the expression of the reprogramming factors, represented through dTomato, was down regulated in both lines in comparison to the endogenous OCT4 expression level (Suppl. Fig. A). For both clones the presence of the heterozygous c.2169C >G mutation in exon 20 of the MYH7 gene was confirmed by Sanger sequencing (Fig. 1A). The clones displayed the typical colony shape and morphology of pluripotent stem cells, expressing classical pluripotency markers like OCT4 and TRA-1- 60 (Fig. 1B). Flow cytometry assessed the purity of pluripotency marker expression in terms of SSEA-4, TRA-1-60, OCT4 and NANOG (Fig. 1C). Cytogenetic analysis showed a normal male karyotype 46,XY in passage 15 for both clones (Fig. 1D) and contamination with my- coplasma could be excluded (Suppl. Fig. B). Short tandem repeat (STR)

analysis proved the genetic identity of the clones to their parental fi- broblasts (Suppl. Table S1). Three germ layer differentiation potential was confirmed by spontaneous embryoid body formation and positive staining for mesodermal (α-smooth muscle actin/ACTA2 and DESMIN), endodermal (FOXA2 and SOX17) and ectodermal markers (TUBB3 and PAX6) (Fig. 1E). Scale bars in all microscopic pictures represent 100 µm.

3. Materials and methods 3.1. Cell culture and reprogramming

Human fibroblasts were expanded in DMEM supplemented with 10% FCS, 1% nonessential amino acid stock and 1 mM L-glutamine. One day prior to transduction, 2x105 fibroblasts were seeded into a six-well.

The adherent cells were transduced with concentrated 4-in-1 lentiviral vector pRRL.PPT.SF.hOct34.hKlf4.hSox2.hmyc.i2dTomato.pre (War- lich et al., 2011) (MOI 1 or 10) in fibroblast culture medium containing 8 μg/mL polybrene (Sigma-Aldrich). The transduced cells were cultured for 6 days in fibroblast culture medium without splitting. On day 6, the transduced cells were passaged using Trypsin (ThermoFisher) and seeded onto a layer of irradiated murine embryonic fibroblasts (MEFs).

On day 7, the culture medium was changed to human iPSC medium (knockout-DMEM supplemented with 20% knockout serum replace- ment, 1 mM L-glutamine, 0.1 mM β-mercaptoethanol, 1% nonessential amino acid stock and 10 ng/mL b-FGF). Arising iPSC-like colonies were transferred mechanically onto fresh irradiated MEFs. Afterwards, iPSC clones were split every 6–7 days using collagenase IV (ThermoFisher) in a 1:10 split ratio onto fresh MEFs. All cells were incubated at 37 C with 5% CO2.

3.2. Real-Time-PCR and mutation analysis

Total RNA was isolated from TRIzol®-lysed cells via NucleoSpin®

RNA II Kit (Machery-Nagel) followed by c-DNA synthesis via

Table 2

Characterization and validation.

Classification Test Result Data

Morphology Photography normal Fig. 1 panel B

Phenotype Qualitative analysis

(Immunocytochemistry) Positive for OCT4 and TRA-1-60 Fig. 1 panel B

Quantitative analysis (Flow

cytometry) MHHi021-A: SSEA4 (96.9%), TRA-1-60 (98.1%), OCT4 (96.4%), NANOG (95.7%);

MHHi021-B: SSEA4 (97%), TRA-1-60 (92.5%), OCT4 (91%), NANOG (90%)

Fig. 1 panel C Genotype Karyotype (fluorescence R-banding)

and resolution Both 46,XY[10] Resolution min 300 bands Fig. 1 panel D

Identity Microsatellite PCR (mPCR) OR

STR analysis not performed

STR profile for 16 sites tested, all matched submitted in archive with journal Mutation analysis (IF

APPLICABLE) Sequencing Heterozygous for c.2169C >G Fig. 1 panel A

Southern Blot OR WGS N/A

Microbiology and virology Mycoplasma negative Suppl. Fig. panel B

Differentiation potential Embryoid body formation Spontaneous FCS-based in vitro differentiation; positive for mesoderm (ACTA2,

DESMIN), endoderm (FOXA2, SOX17) and ectoderm (TUBB3, PAX6) Fig. 1 panel E

Donor screening HIV 1 +2 Hepatitis B, Hepatitis C Negative not shown but

Table 1

Summary of lines.

iPSC line names Abbreviation in figures Gender Age Ethnicity Genotype of locus Disease

R723G_MOI10_K1 (MHHi012-A) MHHi021-A male 38 n/a MYH7 c.2169C >G Hypertrophic Cardiomyopathy

R723G_MOI1_K2 (MHHi021-B) MHHi021-B male 38 n/a MYH7 c.2169C >G Hypertrophic Cardiomyopathy

(3)

Fig. 1.

(4)

RevertAid™ H Minus First Strand cDNA Synthesis Kit (ThermoFisher).

Real-Time-PCR was performed in duplicates using a Mastercycler® ep realplex2 (Eppendorf) and the Absolute™ QPCR SYBR® Green Mix (ABgene). Size of amplicons and absence of non-specific by-products were controlled by melting curves. Expression levels of target genes were normalized to β-Actin transcript levels using the ΔCt method.

Genomic DNA was isolated using the QIAamp DNA Blood Mini Kit (Qiagen). 100 ng of genomic DNA was amplified by PCR with GoTaq®

(Promega) polymerase following the protocol: initial denaturation at 95 C for 2 min; 35 cycles of denaturation at 95 C for 30 sec, annealing at 60 C for 1 min and extension at 72 C for 1 min. The PCR product was purified and sent for Sanger sequencing to MicroSynth Company. The applied primers are listed in Table 3.

3.3. In vitro differentiation

Human iPSCs were detached from the feeder layer by collagenase IV (ThermoFisher), dispersed into small clumps and cultured in differen- tiation medium (IMDM supplemented with 20% FCS, 1 mM L-glutamine, 0.1 mM β-mercaptoethanol and 1% nonessential amino acid stock) in ultra-low attachment plates (Corning) for 7 days. Subsequently, EBs were plated onto 0.1% gelatin coated tissue culture dishes and cultured for additional 13 days in the same medium before fixation and immunostaining.

3.4. Immunofluorescent staining

Cells were fixed on culture dishes with 4% paraformaldehyde (PFA) for 20 min at room temperature (RT). Cells were permeabilized and blocked with 0.025% Triton-X100 and 0.05% donkey serum for 20 min.

Primary antibodies (see Table 3) were diluted in PBS containing 1% BSA

ZENPro Software 3.0.

3.5. Flow cytometry

Before flow cytometry analysis for pluripotency markers, iPSCs were cultured on Geltrex®-coated plates in iPS Brew XF medium (Miltenyi Biotec) at a seeding density of 4 × 104 cells/cm2. For intracellular markers (OCT4, NANOG), cells were harvested with Accutase® (Ther- moFisher) and fixed with 1% PFA for 20 min at RT. Direct-labeled an- tibodies were diluted in solution B/FIX&PERM® and incubated for 20 min at RT. For the staining of surface markers (SSEA-4, TRA-1-60), cells were harvested with Accutase® and incubated for 20 min at 4 C with direct-labeled antibodies diluted in PBS with 1% BSA. Afterwards cells were resuspended in PBS and analyzed with a MACSQuant Analyzer 10 and FlowJo analysis software. Antibodies are listed in Table 3.

3.6. STR analysis

STR analysis was performed by MicroSynth Company (https://www.

microsynth.ch). STR profiling for 16 specific loci were compared with parental fibroblasts.

3.7. Karyotyping

Karyotyping was performed using standard procedure at the Department of Human Genetics at Hannover Medical School. At least 10 metaphase spreads were analyzed per clone using fluorescence R- banding at a minimum of 300 bands. The chromosomes were identified and described according to the International System for Human Cyto- genetic Nomenclature (ISCN).

Table 3 Reagents details.

Antibodies used for immunocytochemistry/flow-cytometry

Antibody Dilution Company Cat # and RRID

Pluripotency Markers mouse anti-OCT4 (IgG2b) 1:100 Santa Cruz Biotechnology Cat# sc-5729, RRID:AB_628051 mouse anti-TRA-1-60 (IgM) 1:100 Abcam Cat# ab16288, RRID:AB_778563

RE anti-OCT4_PE 1:25 Miltenyi Biotec Cat# 130-105-554, RRID:AB_2653085 RE anti-NANOG_APC 1:25 Miltenyi Biotec Cat# 130-105-049, RRID:AB_2652991 RE anti-TRA-1-60_PE 1:25 Miltenyi Biotec Cat# 130-100-347, RRID:AB_2654227 RE anti-SSEA4_VioBlue 1:25 Miltenyi Biotec Cat# 130-098-366, RRID:AB_2653521 Differentiation Markers mouse anti-TUBB3 (IgG2a) 1:400 Millipore Cat# 05-559, RRID:AB_309804

rabbit anti-PAX6 (IgG) 1:300 BioLegend Cat# 901301, RRID:AB_2565003 rabbit anti-FOXA2 (IgG) 1:100 Millipore Cat# 07-633, RRID:AB_390153 goat anti-SOX17 (IgG) 1:200 R&D Systems Cat# AF1924, RRIP:AB_355060

moues anti-ACTA2 (IgG2a) 1:100 Santa Cruz Biotechnology Cat #sc-130616, RRID: AB_1561784 mouse anti-DESMIN (IgG1) 1:20 ARP American Research Products Cat# 03-10519, RRID:AB1541098 Secondary antibodies CyTM3-AffiniPure donkey anti-mouse IgG 1:200 Jackson ImmunoResearch Labs Cat# 715-165-150, RRID:AB_2340813

Cy™3-AffiniPure Donkey anti-Mouse IgM 1:200 Jackson ImmunoResearch Labs Cat# 715-165-020, RRID:AB_2340811 AF488-AffiniPure donkey anti-goat IgG 1:200 Jackson ImmunoResearch Labs Cat# 705-545-147, RRID:AB_2336933 CyTM3-AffiniPure donkey anti-rabbit IgG 1:200 Jackson ImmunoResearch Labs Cat# 711-165-152, RRID:AB_2307443 Primers

Target Forward/Reverse primer (5-3)

Genotyping MYH7 (640 bp) Fwd_tcctacttccttcttgccaca

Rev_ggaaagagatggtggggatt Silencing of reprogramming factors dTomato transgene Fwd_AGCGCGTGATGAACTTCGAG

Rev_CCCAGCCCATCGTCTTCTTC β-Actin Fwd_ATTGCCGACAGGATGCAGAA

Rev_GGGCCGGACTCGTCATACTC

Endogenous OCT4 Fwd_TCCCATGCATTCAAACTGAGG

Rev_CCTTTGTGTTCCCAATTCCTTCC

(5)

to manufactory instructions.

Declaration of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgements

We thank T. Scheper from the Institute for Technical Chemistry, Leibniz University Hannover, for providing b-FGF and A. Schambach from MHH for providing the 4-in-1 transgenic reprogramming cassette.

This work was funded by the German Research Foundation (DFG) including grants: Cluster of Excellence REBIRTH (EXC62/3), project “In vitro model for Familial Hypertrophic Cardiomyopathy based on car- diomyocytes from human induced pluripotent stem cells” (ZW64/4-1, MA2331/16-1, KR1187/21-1, BR849/31-1). We further acknowledge support by the DFG for the Open Access Publication Fund of Hannover Medical School.

Appendix A. Supplementary data

Supplementary data to this article can be found online at https://doi.

org/10.1016/j.scr.2021.102208.

References

Enjuto, M., Francino, A., Navarro-Lopez, F., Viles, D., Pare, J.C., Ballesta, A.M., 2000.

Malignant hypertrophic cardiomyopathy caused by the Arg723Gly mutation in beta- myosin heavy chain gene. J. Mol. Cell Cardiol. 32, 2307–2313.

Kraft, T., Montag, J., Radocaj, A., Brenner, B., 2016. Hypertrophic cardiomyopathy: cell- to-cell imbalance in gene expression and contraction force as trigger for disease phenotype development. Circ. Res. 119 (9), 992995.

Warlich, E., Kuehle, J., Cantz, T., Brugman, M.H., Maetzig, T., Galla, M., Filipczyk, A.A., Halle, S., Klump, H., Sch¨oler, H.R., Baum, C., Schroeder, T., Schambach, A., 2011.

Lentiviral vector design and imaging approaches to visualize the early stages of cellular reprogramming. Mol. Ther. 19 (4), 782789.

Weber, N., Schwanke, K., Greten, S., Wendland, M., Iorga, B., Fischer, M., Geers- Knorr, C., Hegermann, J., Wrede, C., Fiedler, J., et al., 2016. Stiff matrix induces switch to pure beta-cardiac myosin heavy chain expression in human ESC-derived cardiomyocytes. Basic Res. Cardiol. 111, 68.

Referenzen

ÄHNLICHE DOKUMENTE

We generated induced pluripotent stem cell (iPSC) lines from three adult LDS-patients (two male, one female) of a family with a heterozygous point mutation in exon 4 of

a Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), REBIRTH-Research Center for Translational and Regenerative Medicine, Hannover Medical School,

Generation of two human induced pluripotent stem cell lines (MHHi017-A, MHHi017-B) from a patient with primary ciliary dyskinesia carrying a homozygous mutation (c.7915C >

To check for ISG15 expression, the knockout iPSC clones (ISG15 KO ) as well as their wild type mother clone (ISG15 WT ) were differentiated into endothelial cells (EC)

We identified two major types of encounter between second- generation Eritreans and newcomers through which the sec- ond generation starts to reflect about their identity, their

In order to validate whether a mutation in one of these genes might be responsible for OI, we genotyped three gene associated microsatellite markers derived from the surrounding

Loss of function associated with novel mutations of the SCN5A gene in patients with Brugada syndrome. Fever and

Nanog-deficient murine ES cells lost pluripotency and differentiated in to other lineages (Mitsui et al., 2003). Another factor is to determine suitable culture