[go: up one dir, main page]

Academia.eduAcademia.edu
10.1161/CIRCULATIONAHA.121.055313 Cardiovascular Progerin Suppression and lamin A Restoration Rescues Hutchinson-Gilford Progeria Syndrome Running title: Sánchez-López et al.; Cardiovascular Progerin Targeting Extends Lifespan Amanda Sánchez-López, PhD1,2†; Carla Espinós-Estévez, MS1†; Cristina González-Gómez BS1,2; Pilar Gonzalo, PhD1,2, María J. Andrés-Manzano1,2; Víctor Fanjul, PhD1,2; Raquel Riquelme-Borja, MS1; Magda R. Hamczyk, PhD1,2††; Álvaro Macías, PhD1,2; Lara del Campo, PhD1,2; Emilio Camafeita, PhD1,2; Jesús Vázquez, PhD1,2; Anna Barkaway, PhD3; Loïc Rolas, PhD3; Sussan Nourshargh, PhD3; Beatriz Dorado, PhD1,2; Ignacio Benedicto. PhD1; Vicente Andrés, PhD1,2,* 1 Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Downloaded from http://ahajournals.org by on October 27, 2021 2 CIBER de Enfermedades Cardiovasculares (CIBERCV), Spain; 3Centre for Microvascular Research, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK † Co-first authors with equal contribution Current address: Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, 33006 Oviedo, Spain †† Address of Correspondence: Vicente Andrés CNIC, Melchor Fernández Almagro 3, 28029 Madrid, Spain Phone: +34-91 453 12 00 (Ext. 1502) E-mail: vandres@cnic.es *This article is published in its accepted form, it has not been copyedited and has not appeared in an issue of the journal. Preparation for inclusion in an issue of Circulation involves copyediting, typesetting, proofreading, and author review, which may lead to differences between this accepted version of the manuscript and the final, published version. 1 10.1161/CIRCULATIONAHA.121.055313 Abstract Background: Hutchinson-Gilford progeria syndrome (HGPS) is a rare disorder characterized by premature aging and death mainly due to myocardial infarction, stroke, or heart failure. The disease is provoked by progerin, a variant of lamin A expressed in most differentiated cells. Patients look healthy at birth, and symptoms typically emerge in the first or second year of life. Assessing the reversibility of progerin-induced damage and the relative contribution of specific cell types is critical to determining the potential benefits of late treatment and to developing new therapies. Methods: We used CRISPR/Cas9 technology to generate LmnaHGPSrev/HGPSrev (HGPSrev) mice engineered to ubiquitously express progerin while lacking lamin A and allowing progerin suppression and lamin A restoration in a time- and cell-type-specific manner upon Cre recombinase activation. We characterized the phenotype of HGPSrev mice and crossed them with Cre transgenic lines to assess the effects of suppressing progerin and restoring lamin A ubiquitously at different disease stages as well as specifically in vascular smooth muscle cells (VSMCs) and cardiomyocytes. Downloaded from http://ahajournals.org by on October 27, 2021 Results: Like HGPS patients, HGPSrev mice appear healthy at birth and progressively develop HGPS symptoms, including failure to thrive, lipodystrophy, VSMC loss, vascular fibrosis, electrocardiographic anomalies, and precocious death (median lifespan of 15 months versus 26 months in wild-type controls, p<0.0001). Ubiquitous progerin suppression and lamin A restoration significantly extended lifespan when induced in 6-month-old mildly symptomatic mice and even in severely ill animals aged 13 months, although the benefit was much more pronounced upon early intervention (84.5% lifespan extension in mildly symptomatic mice, p<0.0001, and 6.7% in severely ill mice, p<0.01). Remarkably, major vascular alterations were prevented and lifespan normalized in HGPSrev mice when progerin suppression and lamin A restoration were restricted to VSMCs and cardiomyocytes. Conclusions: HGPSrev mice constitute a new experimental model for advancing knowledge of HGPS. Our findings suggest that it is never too late to treat HGPS, although benefit is much more pronounced when progerin is targeted in mice with mild symptoms. Despite the broad expression pattern of progerin and its deleterious effects in many organs, restricting its suppression to VSMCs and cardiomyocytes is sufficient to prevent vascular disease and normalize lifespan. Key Words: Hutchinson-Gilford progeria syndrome; Progerin; Vvascular Smooth Muscle Cell, Cardiomyocyte 2 10.1161/CIRCULATIONAHA.121.055313 Non-standard Abbreviations and Acronyms Downloaded from http://ahajournals.org by on October 27, 2021 BSA CNIC ECG h H&E HGPS HGPSrev HRP IP LC-MS/MS min MRI NGS OT o/n PRM RT SD SEM SEMS SM22α-Cre SMA TBST-T Ubc-CreERT2tg/+ VSMC WT Bovine serum albumin Centro Nacional de Investigaciones Cardiovasculares Electrocardiography Hour(s) Hematoxylin-eosin Hutchinson-Gilford progeria syndrome LmnaHGPSrev/HGPSrev mouse Horseradish peroxidase Immunoprecipitation Liquid chromatography coupled to targeted tandem mass spectrometry Minute(s) Magnetic resonance imaging Normal goat serum Off-target Overnight Precursor-reaction monitoring Room temperature Standard deviation Standard error of the mean Spin-echo multi-slice B6.Cg-Tg(Tagln-cre)1Her/J mouse Smooth muscle α-actin Tris-buffered saline supplemented with 0.2% Tween 20 B6.Cg Ndor1Tg(UBC-cre/ERT2)1Ejb/1J mouse Vascular smooth muscle cell Wild-type 3 10.1161/CIRCULATIONAHA.121.055313 Clinical Perspective What is new? • We have generated a new HGPS-like mouse model that ubiquitously expresses progerin, lacks lamin A, and allows progerin suppression and lamin A restoration in a time- and cell-type-specific manner upon Cre recombinase activation. • Progerin suppression and lamin A restoration extended lifespan in mice with mild symptoms and even in severely ill animals, although the benefit was much more apparent in mildly symptomatic animals. • Despite the broad expression pattern of progerin and its deleterious effects in many organs, restricting its suppression and lamin A restoration to VSMCs and cardiomyocytes prevented the development of vascular pathology and normalized lifespan in HGPSrev mice. What are the clinical implications? Downloaded from http://ahajournals.org by on October 27, 2021 • Our preclinical studies demonstrate that it is never too late to treat HGPS, although the benefit was much more pronounced when progerin suppression and lamin A restoration were achieved at early stages of disease progression. • Strategies to treat HGPS through gene therapy or RNA therapy should consider targeting VSMCs and cardiomyocytes. 4 10.1161/CIRCULATIONAHA.121.055313 Introduction HGPS is an ultra-rare genetic disorder (estimated prevalence 1 in 18-20 million people; https://www.progeriaresearch.org/) characterized by accelerated aging and premature death (average lifespan: 14.6 years)1-3. Most HGPS patients are heterozygous for a de novo synonymous mutation in the LMNA gene (c.1824C>T; p.G608G) that activates the use of a cryptic splice donor site in exon 114, 5. In normal cells, LMNA expression mostly generates the alternatively-spliced isoforms lamin A and lamin C. The HGPS-causing mutation creates an aberrant LMNA mRNA that lacks 150 nucleotides in exon 11; this is translated into progerin, a permanently-farnesylated lamin A variant that exerts a dominant-negative effect1. Growth failure and alopecia manifest in HGPS patients as the first disease symptoms typically in the first or second year of life. Additional symptoms develop and worsen over time, including dermal and bone abnormalities, joint contractures, and loss of subcutaneous fat. The main medical problem in HGPS is severe Downloaded from http://ahajournals.org by on October 27, 2021 cardiovascular disease, including generalized atherosclerosis and vascular calcification and stiffness, which ultimately provoke myocardial infarction, stroke, or heart failure, the causes of death in most HGPS patients6, 7. The US Food and Drug Administration recently approved the treatment of HGPS patients with lonafarnib (marketed as Zokinvy™), a repurposed farnesyltransferase inhibitor that has extended the lifespan of HGPS patients by 2.5 years (17% increase)8-10. Nevertheless, there is a great need for better therapies to improve and eventually cure HGPS. HGPS patients are typically diagnosed when symptoms are present or even severe, and treatment has historically been initiated at different disease stages6. It is therefore important to ascertain how late in life treatment can be initiated in symptomatic individuals while still yielding clinical benefit. Moreover, because progerin is expressed in most differentiated cells, it is vital to identify the cell types that would most benefit from treatment. To address these questions, here we generated the LmnaHGPSrev/HGPSrev (HGPSrev) mouse model, engineered to ubiquitously express 5 10.1161/CIRCULATIONAHA.121.055313 progerin and lamin C and lack lamin A, while allowing progerin suppression and lamin A restoration upon Cre recombinase activation. We have characterized this model to assess the reversibility of progerin-induced damage by targeting progerin at early and late disease stages. Moreover, we have examined the consequences of suppressing progerin and restoring lamin A specifically in vascular smooth muscle cells (VSMCs) and cardiomyocytes, the major progerin targets. Methods Additional methods are provided in the Supplemental Methods section online. Data, analytical methods, and study materials will be made available to other researchers for the purposes of reproducing these results or replicating these procedures upon reasonable request directed to the authors’ laboratories. Study approval and mouse models Downloaded from http://ahajournals.org by on October 27, 2021 Mice used in this study were housed in the animal facilities at the Centro Nacional de Investigaciones Cardiovasculares (CNIC) under specific pathogen-free conditions at a constant temperature of 23°C, relative humidity 58%, and a 12-hour (h) dark/light cycle. Mouse health was monitored in a blinded manner at regular intervals throughout the study. Mouse handling and experimental procedures were performed to conform with current EU guidelines (Directive 2010/63/EU) and Recommendation 2007/526/EC regarding the protection of animals used for scientific purposes, enforced in Spanish law under Real Decreto 1201/2005; all procedures were approved by the Animal Protection Area of the Comunidad Autónoma de Madrid (PROEX 051/18) and the CNIC Ethics Review Board. To maximize information and minimize the number of animals used, we followed the 3Rs principles11 and the ARRIVE guidelines12 throughout this study. 6 10.1161/CIRCULATIONAHA.121.055313 Studies were carried out in C57BL/6J mice fed a regular rodent chow diet. Equal numbers of females and males were used. Ubc-CreERT2tg/+ mice (B6.Cg Ndor1Tg(UBC-cre/ERT2)1Ejb/1J)13 and LmnaG609G/G609G mice14 were kindly provided by Dr. Mariano Barbacid and Dr. Carlos LópezOtín, respectively. The generation of HGPSrev mice is explained below. HGPSrev mice were crossed with Ubc-CreERT2tg/+ mice to generate LmnaHGPSrev/HGPSrev Ubc-CreERT2tg/+ mice with the Cre transgene in heterozygosis and exhibiting time-conditional Cre activity (referred to as HGPSrev-Ubc-CreERT2 mice). SM22α-Cre mice (B6.Cg-Tg(Tagln-cre)1Her/J)15 (The Jackson Laboratory, Bar Harbor, ME USA) were crossed with HGPSrev mice to generate LmnaHGPSrev/HGPSrev-SM22α-Cre mice (HGPSrev-SM22α-Cre mice). Double stranded (dsDNA) donor template design for LmnaHGPSrev strain generation For homology directed repair, a 2,494 bp dsDNA donor template flanked by EcoRI and NotI recognition sites was synthesized (Figure IA in the Supplement) and inserted into the pcDNA3.1 vector (Genscript; Piscataway, NJ USA). The dsDNA donor template contains a 938-bp left Downloaded from http://ahajournals.org by on October 27, 2021 homology arm (Lmna intron 9, exon 10 and part of intron 10), a 672-bp insert harboring a loxPflanked cDNA containing exons 11 and 12 from Lmna∆150 (exon11∆150 and the coding sequence of exon 12), followed by a bovine growth hormone polyadenylation transcriptional stop signal (BGH-polyA), and an 877-bp right homology arm (part of Lmna intron 10, exon 11, intron 11 and part of exon 12). Oocyte microinjection and implantation into pseudo-pregnant females Hormonal superovulation was induced in 10 immature female mice (3-5 weeks old, C57BL/6J genetic background) by intraperitoneal hormone injection. Mice first received an injection of 0.1 mL (5 IU) of pregnant mare serum gonadotropin, followed 48 h later by an injection of 0.1 mL (5 IU) of human chorionic gonadotropin. Immediately after the second injection, animals were mated with appropriate stud males. One day after mating, females were checked for vaginal plugs, and those with a positive result were sacrificed. Oviducts from sacrificed females were extracted and 7 10.1161/CIRCULATIONAHA.121.055313 transferred to M2 culture medium (M7167; Sigma-Aldrich, St Louis, MO USA) containing 350 µg/mL hyaluronidase (H3884, Sigma-Aldrich). Each ampulla was localized and opened to release the cumulus mass, and oocytes were separated after incubation at 37 ºC for 1-2 minutes (min) and then transferred to fresh M2 medium for washing. Zygotes were incubated in Evolve-KSOM culture medium (ZEKS-050; Zenith Biotech, Cork, Ireland) at 37 ºC in a 5% CO2/5% O2 atmosphere, until they were ready for pronuclear microinjection16. Zygotes were microinjected with 1-2 pL of a microinjection solution containing guide RNAs, dsDNA donor template and Cas9 endonuclease (Table I in the Supplement). Zygotes were incubated overnight (o/n) at 37 ºC and 5% CO2/5% O2 in Evolve-KSOM medium (ZEKS-050; Zenith Biotech) to reach the 2-cell stage16. Embryos at the 2-cell stage were transferred to pseudo-pregnant female mice by passing a sterile glass needle through the infundibulum. Three weeks later, 34 pups were weaned from their gestational mothers. Identification of founder HGPSrev mice Downloaded from http://ahajournals.org by on October 27, 2021 To identify mice carrying the LmnaHGPSrev allele, we extracted genomic DNA from the tails of the 34 mouse pups following a standard protocol with Proteinase K (EO0492; Thermo Fisher, Waltham, MA USA) and performed PCR with specific primers (Table II in the Supplement Founders: PCR-1). Additional PCR reactions were run to identify mice carrying a single copy of the mutant allele at the proper location in the Lmna locus (Table II in the Supplement Founders: PCR-2 and PCR-3). Genomic DNA from the 4 pups carrying a single copy of the edited allele was amplified by PCR and sequenced at the Sequencing Service of the Centro Nacional de Investigaciones Oncológicas (CNIO, Madrid, Spain) (Figure IB in the Supplement). Genotyping of HGPSrev mice To genotype HGPSrev, HGPSrev-Ubc-CreERT2 and HGPSrev-SM22α-Cre mice, genomic DNA was extracted from the tail following a standard Proteinase K protocol (EO0492; Thermo Fisher) 8 10.1161/CIRCULATIONAHA.121.055313 and PCR reactions were performed with specific primers (Table II in the Supplement Genotyping). Isolation, immortalization and transfection of mouse embryonic fibroblasts (MEFs) Embryos isolated at embryonic day 13.5 were minced and incubated for 20 min in 2X trypsinEDTA (0.5% Trypsin, 0.53 mM EDTA•4Na) (15400-054; Invitrogen, Carlsbad, CA USA). MEFs from each embryo were plated separately and incubated at 37 °C in complete growth medium (DMEM supplemented with 10% heat-inactivated fetal bovine serum, 5% non-essential amino acids, 5% penicillin/streptomycin, and 5% L-glutamine (v/v)). To generate virus to immortalize MEFs, HEK293T cells were seeded and transfected with pCL-Puro-SV40 LT retroviral vector (13970; Addgene, Watertown, MA USA) and pCL-ECO retroviral packaging plasmid (kindly provided by Dr. Manuel Serrano) using Fugene 6 (E2692; Promega, Madison, WI USA). Supernatants containing retroviral particles were harvested every 12 h over 2 days, filtered through 0.45 µm pores, mixed with polybrene (8 µg/mL) (9268-5G; Sigma-Aldrich), and used to Downloaded from http://ahajournals.org by on October 27, 2021 infect 2.5-5x105 MEFs in 100 mm dishes. Infected MEFs were serially passaged to select immortalized cells by adding 2 µg/mL puromycin (P8833, Sigma-Aldrich). Immortalized MEFs were seeded at ≈80% confluence and were transfected with 1.5 µg pPB CAG ER-Cre-ER IRES Zeocin17 and 4.5 µg pCMV-hyPBase18 mixed with 18 µL TransITLT1 Transfection Reagent (MIR2300; GeneFlow, Staffordshire, UK) in DMEM. Zeocin-resistant cells were selected by incubation for at least 10 days in the presence of 400 μg/mL zeocin (R25005; Thermo Fisher). Longevity studies Starting at 4-8 weeks of age, mice were weighed and inspected for health and survival twice a week. Health status was examined by a blinded veterinarian. Animals that met humane end-point criteria were euthanized and the deaths recorded for the survival curve analysis. The ROUT test was performed in all survival experiments, and identified two outliers: a tamoxifen-injected 9 10.1161/CIRCULATIONAHA.121.055313 HGPSrev-Ubc-CreERT2 mouse that died at 34 months and an oil-injected mouse that died at 17 months (in both cases, administration of tamoxifen/oil started at ≈13 months). These mice were excluded from the survival curve and statistical analysis. Histology and immunofluorescence Mouse tissues were fixed in 4% formaldehyde solution (prepared from paraformaldehyde) for 24 h at 4 ºC, dehydrated through an ascending series of ethanol concentrations, and finally embedded in paraffin and cut into serial 4-μm sections. For immunofluorescence studies, tissue cross-sections were deparaffinized, rehydrated and washed in PBS. Antigen retrieval was performed by boiling the sections in 10 mM sodium citrate buffer (pH 6) for 20 min, and samples were then blocked and permeabilized for 1 h at room temperature (RT) in PBS supplemented with 0.3% Triton X-100, 5% BSA, and 5% normal goat serum (NGS) (005-000-001, Jackson ImmunoResearch, West Grove, PA USA). Heart samples were additionally blocked with 100 mM glycine (5001901000; Merck, Kenilworth, NJ USA). Downloaded from http://ahajournals.org by on October 27, 2021 Primary and secondary antibodies were diluted in PBS supplemented with 0.3% Triton X-100, 5% BSA, and 2.5% NGS. Primary antibodies included rat monoclonal anti-CD31 (1:50, DIA-310; Dianova, Hamburg, Germany) and rabbit polyclonal anti-progerin (1:800, generated by the Nourshargh laboratory using peptide immunogens and standard immunization procedures). After overnight incubation at 4ºC, samples were washed and then incubated with the appropriate fluorescently-labeled secondary antibodies for 2 h at RT (1:400 goat anti-rat Alexa Fluor 488 A11006 or 1:400 goat anti-rabbit Alexa Fluor 647 A-21245; Invitrogen) together with anti-αsmooth muscle actin-Cy3 (1:300, C6198; Sigma-Aldrich) and Hoechst nuclear stain (bisBenzimide H 33342 trihydrochloride, B2261; Sigma Aldrich). Samples were washed and then mounted in Fluoromount-G imaging medium (4958-02; eBioscience, San Diego, CA USA). Images were acquired with a Leica SP5 DMI 6000B (Leica® Microsystems, Wetzlar, Germany) or a LSM 700 Carl Zeiss (Zeiss, Oberkochen, Germany) confocal microscope. 10 10.1161/CIRCULATIONAHA.121.055313 For staining with hematoxylin-eosin (H&E) and Masson’s trichrome, tissue cross-sections were deparaffinized, rehydrated and washed in PBS. Stained sections were scanned with a NanoZoomer-RS scanner (Hamamatsu, Japan), and images were exported with NDP.view2 software (Hamamatsu) and quantified with user-customized macros in Fiji software by an operator blinded to genotype. The thickness of the subcutaneous fat layer was qualitatively scored from 1 (thinnest) to 5 (thickest) by five independent observers and mean results are presented for each mouse. For quantitative analysis, regions containing epidermis, hypodermis, and muscle layers were examined by a single blinded manner observer, and the mean thickness was calculated from 10 independent measurements per mouse. mRNA isolation and reverse transcription for PCR detection of lamin A and progerin Total RNA was extracted from powdered mouse tissue samples using TriReagent Solution (AM9738, Thermo Fisher) and processed by alcohol precipitation. RNA was quantified in a NanoDrop ND-1000 spectrophotometer and 1-2 µg were transcribed to cDNA using the HighDownloaded from http://ahajournals.org by on October 27, 2021 Capacity cDNA Reverse Transcription Kit (4368814; Applied Biosystems, Foster City, CA USA). Lamin A and progerin mRNAs were detected as previously described19. cDNA samples (100 ng) were amplified by PCR, and products were separated on a 1.5% agarose gel. Images were acquired with the Molecular Imager® Gel DocTM XR+ System (Bio-Rad). Band intensity was quantified using Fiji software. Plasma biochemistry Blood was extracted from the mandibular sinus of live mice or from the heart (by cardiac puncture) or infrarenal abdominal aorta of euthanized animals. For biochemical analysis, plasma was obtained from blood samples collected in Microvette EDTA tubes (Sarstedt, Newton, NC USA) by specialized CNIC Animal Facility staff and centrifuged at 180-200 g for 15 min at 4ºC. Biochemical variables were analyzed using a Dimension RxL Max Integrated Chemistry System (Siemens Healthineers, Erlangen, Germany). 11 10.1161/CIRCULATIONAHA.121.055313 Electrocardiography (ECG) Mice were anesthetized with 1.5-2% isoflurane, and 4 ECG electrodes were inserted subcutaneously into the limbs. ECG was recorded in the morning for approximately 2 min using a MP36R data acquisition workstation (Biopac Systems, Goleta, CA, USA). ECG data were exported with AcqKnowledge software (Biopac Systems) and automatically analyzed using custom R scripts developed to: 1) remove noise and baseline fluctuations; 2) detect heart beats, peaks and waves; 3) exclude artifacts; and 4) calculate QT intervals and T-wave steepness. Tamoxifen administration Tamoxifen (4-hydroxy-tamoxifen, H6278; Sigma Aldrich) was dissolved in ethanol for cell studies or in corn oil (C8267; Sigma Aldrich) for mouse studies. The corn-oil preparation was incubated at 55°C until the tamoxifen was fully dissolved and then passed through a 0.22 µm filter. Zeocin-resistant WT and HGPSrev MEFs were exposed to 25 nM tamoxifen and protein Downloaded from http://ahajournals.org by on October 27, 2021 lysates were prepared 24, 48 and 72 h after tamoxifen administration. Negative controls were treated with equal volume of vehicle (ethanol) for 72 h. Mice were randomized to tamoxifen or oil groups balanced for age and sex. For proof-ofconcept studies, ≈3-month-old HGPSrev-Ubc-CreERT2 mice received daily intraperitoneal oil or tamoxifen injections (2 mg/day/mouse) for 10 days; the mice were sacrificed one week after finishing the treatment and tissues were extracted for western blot analysis. For the analysis of HGPSrev-Ubc-CreERT2 mice at different disease stages, tamoxifen administration was commenced at different ages. For longitudinal studies of survival, health status and body-weight evolution, the effect on early disease was assessed by starting oil/tamoxifen administration at ≈6 months of age, coinciding with the beginning of growth failure, whereas late disease was assessed by starting treatment at ≈13 months of age, when mice had developed severe symptoms and were close to maximum survival (Table 1). For ECG and histopathological analysis, the effect on 12 10.1161/CIRCULATIONAHA.121.055313 intermediate disease (characterized by mild cardiovascular symptoms) was assessed by starting treatment at ≈9 months of age. All mice received daily intraperitoneal injections (1 mg/day/mouse) over 5 days and were sacrificed at 14.5 months of age, at which point 4 out of 10 oil-injected animals had died and only 1 out of 12 tamoxifen-injected animals had died. Statistical analysis Quantitative data are presented as the mean ± the standard error of the mean (SEM) unless otherwise stated. Statistical tests were applied after the determination of normal distribution (Shapiro-Wilk normality test) and equality of variances (F test). In experiments with two groups and normal distribution, the statistical significance of differences was assessed by unpaired twotailed Student’s t-test. For non-normally distributed data in experiments with two groups, we used the Mann-Whitney test. In experiments with more than two groups of normally distributed populations, we applied one-way ANOVA followed by the post hoc Tukey test. For non-normally distributed data, the non-parametric equivalent Kruskal-Wallis test was performed. When more Downloaded from http://ahajournals.org by on October 27, 2021 than 2 groups were assessed over time without sphericity, we used mixed-effects analysis (twoway ANOVA) with the Geisser-Greenhouse correction and Sidák’s multiple comparisons. Kaplan-Meier survival curves were compared by the log-rank (Mantel-Cox) test. Data on bodyweight evolution were analyzed by unpaired multiple t-tests with the Holm-Sidák correction. All statistical tests were run in GraphPad Prism® 9.0.0. Differences were considered statistically significant when p-values were below 0.05: *, p<0.05; **, p<0.01; ***, p<0.001; ****, p<0.0001. Results HGPSrev mice develop progeroid symptoms and allow Cre-dependent progerin suppression and lamin A restoration 13 10.1161/CIRCULATIONAHA.121.055313 We used the CRISPR-Cas9 strategy to generate the HGPSrev mouse model, engineered to ubiquitously express progerin and lamin C and to lack lamin A, while allowing progerin suppression and lamin A restoration upon Cre recombinase activation (Figure 1A, and Figure IA,B in the Supplement). Western blot analysis in protein lysates from the tails of founder HGPSrev mice revealed progerin and lamin C expression and undetectable lamin A (Figure IC in the Supplement). HGPSrev mice obtained after breeding founder mice expressed progerin in all tissues tested, as assessed by reverse transcriptase-PCR (Figure ID in the Supplement: compare wild-type (WT) with HGPSrev), immunofluorescence (Figure 1B, and Figure II in the Supplement: compare WT with HGPSrev), and western blot (Figure 1C). Analysis of multiple tissues by semiquantitative PCR showed lower progerin mRNA content in HGPSrev mice than in LmnaG609G/G609G mice, a widely used HGPS model14 (Figure ID in the Supplement: compare HGPSrev with G609G). We next examined progerin farnesylation in HGPSrev and LmnaG609G/G609G mice, using WT Downloaded from http://ahajournals.org by on October 27, 2021 mice as a negative control. Proteins were immunoprecipitated from heart lysates with an antilamin A/C antibody that recognizes lamin A/C and progerin, and samples were analyzed by western blot and liquid chromatography coupled to targeted tandem mass spectrometry (LCMS/MS) using a high-resolution precursor-reaction monitoring (PRM) method (Figure 2A and Supplemental Methods). Compared with the initial lysates, WT heart immunoprecipitates were enriched in lamin A/C (Figure 2B, WT (+) versus WT Initial lysate), HGPSrev heart immunoprecipitates were enriched in progerin and lamin C and contained no detectable lamin A (Figure 2B, HGPSrev (+) versus HGPSrev Initial lysate), and LmnaG609G/G609G heart immunoprecipitates were enriched in progerin and lamin A/C (Figure 2B, G9609G (+) versus G609G Initial lysate). For PRM, we monitored a peptide common to lamin A and progerin (IC, internal control) and peptides specific for lamin A (LA) and farnesylated progerin (FP) (Figure 2C). The MS/MS spectra confirmed the presence of the farnesyl moiety in the monitored FP 14 10.1161/CIRCULATIONAHA.121.055313 peptide (Figure 2D). MS2 extracted ion chromatograms obtained from the time-scheduled PRM assay showed similar amounts of FP in HGPSrev and LmnaG609G/G609G hearts relative to the total amount of A-type lamin isoforms in these mutant strains, whereas FP was undetectable in WT hearts (Figure 2E). Consistent with this finding, LA was strongly depleted in HGPSrev and LmnaG609G/G609G hearts relative to WT hearts (Figure 2E). To examine potential off-target (OT) effects in HGPSrev mice, we used the online OffSpotter tool (https://cm.jefferson.edu/Off-Spotter/). Compared with the 20 mer sgRNA used for CRISPR/Cas9-dependent editing, this analysis identified 184 mouse genomic sequences containing 3, 4, or 5 mismatches (2, 16, and 166 sequences, respectively) (Figure IIIA in the Supplement). We selected 3-mismatch sequences (OT-1 and OT-2) and 6 of the 4-mismatch sequences (OT-3, OT-4, OT-5, OT-6, OT-7, and OT-8) (Figure IIIB in the Supplement), which were amplified by PCR using as template genomic DNA of WT and HGPSrev mice (n=5 per genotype). The PCR products had identical DNA sequence in all mice for all genomic regions Downloaded from http://ahajournals.org by on October 27, 2021 examined (Figure IIIC in the Supplement), indicating the absence of off-target effects. HGPSrev mice looked normal at birth and maintained a normal appearance until aged ≈5 months, when both males and females stopped gaining weight (Figure 3A). From this age, HGPSrev mice also exhibited kyphosis (Figure 3B) and showed a significant fat loss, as assessed from subcutaneous fat histology (Figure 3C) and in vivo magnetic resonance imaging (MRI) (Figure 3D). At ≈8 months of age, HGPSrev mice had below-normal levels of plasma low-density lipoprotein, but other lipids were normal (Figure IVA in the Supplement). In ≈13-month-old HGPSrev mice, plasma triglycerides, total cholesterol, and high-density-lipoprotein levels were reduced (Figure IVB in the Supplement), consistent with observations in other progerinexpressing mouse models20. Progerin expression was also associated with premature death in HGPSrev mice, which had a median lifespan of 15 months versus 26 months in WT controls (43.5% reduction) (Figure 3E). 15 10.1161/CIRCULATIONAHA.121.055313 HGPS patients and animal models are both characterized by VSMC loss and collagen accumulation in the artery wall, as well as electrocardiographic alterations7, 14, 20-30. In ≈8-monthold HGPSrev mice, medial VSMC content appeared normal in both the aortic arch and thoracic aorta (Figure 4A); however, the media of the aortic arch showed increased collagen accumulation compared with age-matched WT mice (Figure 4B). Disease progression manifested as severe VSMC depletion in the aortic arch and thoracic aorta in ≈13-month-old HGPSrev mice (Figure 5A), which also had a significantly elevated collagen content in both the media and adventitia of the aortic arch and in the media of the thoracic aorta (Figure 5B). Likewise, longitudinal ECG assessment of HGPSrev mice revealed an age-dependent reduction in T-wave steepness from 8 months of age and an increase in the QT interval from 10 months (Figure 5C). These findings demonstrate that HGPSrev mice, like HGPS patients, appear healthy at birth and progressively develop the main features of the human disease, including cardiovascular alterations and premature death. Downloaded from http://ahajournals.org by on October 27, 2021 Ubiquitous progerin suppression and lamin A restoration extends lifespan when induced in mildly symptomatic and in severely ill HGPSrev mice Treatment of HGPS patients has been initiated at widely differing ages and disease stages6, but how late treatment can be started and still ameliorate symptoms remains unknown. The HGPSrev model was designed to address this key question by taking advantage of Cre recombinase expression to remove the progerin-expressing cassette and restore lamin A expression (Figure 1A). We first performed in vitro studies in WT and HGPSrev MEFs that were stably transfected with a vector encoding a tamoxifen-inducible Cre recombinase and a zeocin-resistance cassette (Figure 6A). Western blot assays of zeocin-resistant cells showed complete progerin suppression and lamin A restoration in HGPSrev MEFs 72 hours after tamoxifen administration, with no effects in tamoxifen-treated WT MEFs (Figure 6B). To test the system in vivo, we generated HGPSrev-Ubc-CreERT2 mice by crossing HGPSrev mice with transgenic Ubc-CreERT2-tg/+ mice, 16 10.1161/CIRCULATIONAHA.121.055313 which ubiquitously express a tamoxifen-inducible Cre recombinase13. Compared with agematched oil (vehicle)-injected controls, ≈3-month-old HGPSrev-Ubc-CreERT2 mice injected with tamoxifen and sacrificed 1 week later displayed progerin downregulation and lamin A expression in all tissues tested (Figure 6C, and quantification in Figure V in the Supplement). Tamoxifen injection in ≈13-month-old HGPSrev-Ubc-CreERT2 mice also resulted in progerin downregulation and lamin A restoration (Figure 6D). To assess the impact of in vivo systemic progerin suppression and lamin A restoration starting at different stages of HGPS progression, we defined two ages for the start of tamoxifen administration, representing early disease (≈6 months: beginning of growth failure, one of the earliest symptoms in HGPS patients) and late disease (≈13 months: close to maximum survival) (Figure 7A and Table 1). Treating ≈6-month-old HGPSrev-Ubc-CreERT2 mice with tamoxifen prevented the body-weight loss observed in oil-injected controls at late disease stages (Figure 7B, top left graph), but body weight remained below WT values in mice older than 12 months (see Downloaded from http://ahajournals.org by on October 27, 2021 WT in Figure 3A). Despite this, progeroid mice injected with tamoxifen at ≈6 months showed an 84.5% increase in median lifespan (p<0.0001 versus oil; experiment ongoing at the time of manuscript submission: all oil-treated controls had died by ≈15 months of age, whereas 13 out of 22 tamoxifen-treated mice were still alive and in good health at ≈27 months of age) (Figure 7B, bottom left graph). Beginning tamoxifen administration in ≈13-month-old HGPSrev-UbcCreERT2 mice with severe symptoms resulted in progerin downregulation and lamin A expression (Figure 6D), and increased median lifespan by 6.7% (p<0.05 versus oil-treated mice) (Figure 7B, bottom right graph). These results demonstrate that administering tamoxifen to HGPSrev-UbcCreERT2 mice even at very advanced disease stages significantly increases lifespan, although the benefit is much more pronounced when progerin suppression and lamin A restoration are achieved in mildly symptomatic mice. 17 10.1161/CIRCULATIONAHA.121.055313 We next investigated how suppressing progerin and restoring lamin A expression affects the cardiovascular phenotype of HGPSrev-Ubc-CreERT2 mice. Tamoxifen was administered to HGPSrev mice at ≈9 months of age (Table 1), an intermediate disease stage when the mice already showed a clear reduction in body weight (Figure 3A) and had begun to develop cardiovascular alterations (Figure 4B and Figure 5C). Mice were sacrificed at ≈14.5 months of age (at which stage 4 out of 10 oil-injected animals but only 1 out of 12 tamoxifen-injected animals had died). Longitudinal studies showed no statistically-significant differences in ECG parameters between both groups of mice at all ages tested (Figure 7C). In contrast, histological analysis revealed increased VSMC number and decreased medial collagen content in the aortic arch and thoracic aorta of tamoxifen-injected HGPSrev-Ubc-CreERT2 mice relative to oil-injected controls, although tamoxifen did not normalize these parameters to the values seen in WT mice (Figure 7D). Progerin suppression and lamin A restoration in vascular smooth muscle cells and Downloaded from http://ahajournals.org by on October 27, 2021 cardiomyocytes is sufficient to prevent vascular damage and to normalize lifespan in HGPSrev mice To investigate whether all affected tissues should be targeted to ameliorate disease symptoms, or whether tissue-specific therapies would be effective, we examined the effect of suppressing progerin and restoring lamin A only in VSMCs and cardiomyocytes, major progerin targets in HGPS patients and animal models2, 6, 7, 14, 20-28. For this analysis, we used the SM22α-Cre transgenic line —which allows Cre-dependent recombination in VSMCs and cardiomyocytes31— to generate HGPSrev-SM22α-Cre mice. Immunofluorescence experiments confirmed that progerin expression was undetectable in arterial VSMCs of HGPSrev-SM22α-Cre mice and was clearly reduced in non-endothelial cardiac cells (69±3.8% progerin-positive cells in HGPSrev mice versus 17±2.8% in HGPSrev-SM22α-Cre mice; p<0.0001 by unpaired two-tailed Student t-test), but remained robust in endothelial cells in aorta and heart (Figure 8A) and in non-cardiovascular 18 10.1161/CIRCULATIONAHA.121.055313 tissues (Figure II in the Supplement). Progerin elimination and lamin A restoration were also confirmed by western blot assays in HGPSrev-SM22α-Cre aorta and heart, whereas progerin and lamin C expression and lack of lamin A were observed in skeletal muscle, kidney, and spleen (Figure 8B). Although HGPSrev-SM22α-Cre mice had a higher body weight than HGPSrev controls from 12 months of age, they were significantly thinner than age-matched WT mice from ≈17 months of age (Figure 8C). Despite this, the lifespan (Figure 8D), aortic medial VSMC density (Figure 8E), and collagen content (Figure 8F) of HGPSrev-SM22α-Cre mice were undistinguishable from those of WT mice. These findings complement our recent studies showing that SM22α-Cre-driven progerin expression is sufficient to promote cardiovascular alterations in mice20, 29, 30, 32 and demonstrate that targeting progerin in VSMCs and cardiomyocytes is sufficient to normalize life expectancy despite the reduced body weight caused by progerin expression in other cell types. Downloaded from http://ahajournals.org by on October 27, 2021 Discussion We report here the generation and characterization of HGSrev mice, a new HGPS model that features ubiquitous progerin and lamin C expression and lack of lamin A, and that allows Credependent progerin suppression and lamin A restoration in a time- and cell-type-specific manner. HGPSrev mice progressively develop many symptoms characteristic of the human disease, including growth failure, lipodystrophy, VSMC loss, vascular fibrosis, electrocardiographic anomalies, and premature death. Interestingly, we found increased collagen accumulation in the aortic media of HGPSrev mice at ages when VSMC content still does not differ from that of WT mice. Likewise, previous studies in 8-week-old LmnaG609G/G609G mice that still exhibited normal VSMC content revealed alterations in genes involved in aortic fibrosis32 and higher collagen III and lysyl oxidase expression in carotid arteries33, suggesting that progerin-induced extracellular matrix alterations precede VSMC death. 19 10.1161/CIRCULATIONAHA.121.055313 Unlike HGPS patients and LmnaG609G/G609G and LMNA G608G transgenic mice, two widelyused HGPS models14, 26, HGPSrev mice exhibit undetectable lamin A expression; however, mice that ubiquitously express lamin C and lack lamin A appeared normal14, 34, 35, suggesting that lamin A absence is unlikely to provoke progeroid symptoms in HGPSrev mice. Indeed, HGPSrev mice develop disease symptoms more slowly and die later than homozygous LMNA G608G mice36 and LmnaG609G/G609G mice14. Our proteomic studies in HGPSrev and LmnaG609G/G609G mice revealed similar amounts of farnesylated progerin in both models when normalized to the total amount of A-type lamin isoforms, but HGPSrev mice exhibited lower progerin mRNA level in all tissues tested, which may explain, at least in part, their milder phenotype. Because HGPS is a progressive disease and children with this condition are diagnosed after the appearance of symptoms6, it is critical to determine the reversibility of progerin-induced damage and the optimal time window for treatment, chief questions that remain unanswered. In progerin-expressing mice, the progeroid phenotype can be ameliorated and lifespan increased by Downloaded from http://ahajournals.org by on October 27, 2021 CRISPR-Cas937, 38 and base-editing approaches36 to ubiquitously correct the HGPS-causing mutation or by the delivery of antisense oligonucleotides to block pathogenic splicing of mutant lamin A transcripts14, 39, 40; however, these treatments were administered to asymptomatic neonates and young animals. We found an 84.5% extension in median lifespan when progerin was suppressed and lamin A restored in mildly symptomatic ≈6-month-old HGPSrev mice. This benefit in survival occurred despite the below-normal body weight of these animals, consistent with previous mouse studies and clinical trials demonstrating lifespan extension without bodyweight normalization after the administration of various treatments (e.g., 3, 8, 14, 32, 36-40). Future studies are warranted to test whether early disease stages feature irreversible adipocyte death that causes persistent lipodystrophy. Importantly, progerin suppression and lamin A restoration also prolonged lifespan in severely affected progeroid HGPSrev mice. Although phenotypic amelioration was much more pronounced when progerin suppression and lamin A restoration were 20 10.1161/CIRCULATIONAHA.121.055313 achieved in early disease stages, these results strongly suggest that it is never too late to start treatment for HGPS. Indeed, tipifarnib has been found to prevent the late progression of existing cardiovascular defects when administered to progeroid mice with overt disease symptoms41. Moreover, lonafarnib improved carotid-femoral pulse wave velocity and other outcome measures in HGPS clinical-trial participants who started treatment at advanced disease stages8 and also prolonged survival in a trial population with an average age of enrollment of 8.4 years3. In a background of normal expression of endogenous wild-type Lmna, Eriksson and collaborators generated transgenic mice with doxycycline-inducible expression of progerin in skin or osteoblasts/odontoblasts, which showed skin or bone/teeth defects upon doxycycline administration, respectively42, 43. Remarkably, the defects in these mutant mice were normalized by turning off progerin expression after the appearance of the phenotype. These seminal studies in mice demonstrated that ectopic progerin expression in skin and bone does not cause irreversible damage to these tissues in the context of normal endogenous lamin A expression. Nonetheless, it Downloaded from http://ahajournals.org by on October 27, 2021 is important to note that HGPS patients express below-normal levels of lamin A and that progerin is expressed in a broad range of tissues. Moreover, a major medical problem in HGPS is severe cardiovascular disease. Thus, uncertainty remained about the ability of progerin suppression and lamin A restoration to halt disease progression and increase lifespan when administered only in cardiovascular cells, the major progerin targets. We found that vascular abnormalities and premature death are both prevented in HGPSrev-SM22α-Cre mice with progerin suppression and lamin A restoration restricted to VSMCs and cardiomyocytes. This benefit occurred despite sustained broad progerin expression in other cell types, which was associated with significantly reduced body weight compared with age-matched WT mice. Although our model does not differentiate between effects in VSMCs and cardiomyocytes, we found that lifespan extension after ubiquitous progerin suppression and lamin A restoration in symptomatic progeroid mice was associated with reductions in both VSMC loss and collagen accumulation in the aortic media but 21 10.1161/CIRCULATIONAHA.121.055313 did not ameliorate electrocardiographic defects. Moreover, HGPS is not known to be a cardiomyopathy, and massive VSMC loss and accumulation of extracellular matrix components have been observed in the arterial wall of HGPS mouse models and patients7, 14, 20, 23, 25-30. Taken together, these findings suggest a major role of vascular disease in HGPS. Further studies using the HGPSrev model and Cre transgenic lines specifically targeting VSMCs or cardiomyocytes will permit to conclusively dissect out the relative contribution of these cell types to disease progression and premature death, which may help optimize gene therapy or RNA therapy to treat HGPS. Acknowledgments We thank Simon Bartlett for English editing, and Azim Surani (University of Cambridge, UK), Steve P. Jackson (University of Cambridge, UK), Manuel Serrano (Institute for Research in Biomedicine, Barcelona, Spain), Mariano Barbacid (CNIO, Madrid, Spain), Carlos López-Otín Downloaded from http://ahajournals.org by on October 27, 2021 (Universidad de Oviedo, Spain) and Enrique Lara-Pezzi (CNIC, Madrid, Spain) for providing the pPB CAG ER-Cre-ER IRES Zeocin, pCMV-hyPBase and pCL-ECO plasmids, the Ubc-CreERT2tg/+ mice, the LmnaG609G/G609G mice, and the HEK293T cells, respectively. We also thank David Filgueiras (Hospital Clínico San Carlos; CNIC, Spain) for advice with electrocardiography, Jacob F Bentzon (Aarhus University, DK; CNIC, Spain) for critical reading of the manuscript, Delphine Larrieu and Josep Vicent Forment (University of Cambridge, UK) for advice on MEF experiments, Davide Seruggia and Lluís Montoliu (CNB-CSIC, Spain) for advice on CRISPRCas9 strategy design, and the excellent support of the CNIC Animal Facility (with special thanks to Eva Santos, Marta García, and Belén Ricote), Histology Service, Microscopy Unit (with special thanks to Verónica Labrador), and Advanced Imaging Unit. Author contributions: AS-L and VA conceived, designed and planned the overall study. AS-L generated the HGPSrev mouse model. AS-L and CE-E designed and performed experiments and analyzed and interpreted results. CG-G, 22 10.1161/CIRCULATIONAHA.121.055313 PG, MJA-M, and RR-B carried out experiments. VF and CE-E designed, analyzed and interpreted ECG experiments. AB, LR, and SN generated, characterized, and provided the anti-progerin antibody. EC and JV designed and performed targeted tandem mass spectrometry and interpreted the results. VF, MRH, LdC, BD, IB and AM provided advice and scientific discussion. VA interpreted the results and supervised the overall study. CE-E, IB, and VA wrote the manuscript. All authors discussed the results and critically reviewed the manuscript. Sources of Funding This study was supported by grants to VA from the Spanish Ministerio de Ciencia e Innovación (MCIN)/Agencia Estatal de Investigación (AEI)/10.13039/501100011033 (grants SAF201679490-R, PID2019-108489RB-I00) and the Instituto de Salud Carlos III (ISCIII; grant AC17/00067 as coordinator of TREAT-HGPS, a project in the E-Rare Joint Transnational call, European Union Horizon 2020 Framework Programme 2017), with co-funding from Fondo Downloaded from http://ahajournals.org by on October 27, 2021 Europeo de Desarrollo Regional (“A way to build Europe”). Additional funding: MICIN (grant SVP-2014-068334) and Asociación Apadrina la Ciencia-Ford España-Ford Motor Company Fund (ASL); Fundación "la Caixa” grants LCF/BQ/DR19/1170012 (CEE) and LCF/BQ/DE14/10320024 (VF); Comunidad Autónoma de Madrid grant 2017-T1/BMD-5247 (IB); MICIN/AEI/10.13039/501100011033 grant FJCI-2017-33299 (MRH); and Wellcome Trust grant 098291/Z/12/Z (SN). The CNIC is supported by the MICIN, the ISCIII, the Pro-CNIC Foundation, and is a Severo Ochoa Center of Excellence (grant CEX2020-001041-S funded by MICIN/AEI/10.13039/501100011033). Disclosures 23 10.1161/CIRCULATIONAHA.121.055313 The authors declare no conflict of interest. The funders had no role in the design of the study; the collection, analysis, or interpretation of data; the writing of the manuscript; or the decision to publish the results. Supplemental Materials Supplemental Methods Supplemental Tables I – II Supplemental Figures I – V Downloaded from http://ahajournals.org by on October 27, 2021 24 10.1161/CIRCULATIONAHA.121.055313 References Downloaded from http://ahajournals.org by on October 27, 2021 1. Dorado B and Andrés V. A-type lamins and cardiovascular disease in premature aging syndromes. Curr Opin Cell Biol. 2017;46:17-25. 2. Merideth MA, Gordon LB, Clauss S, Sachdev V, Smith ACM, Perry MB, Brewer CC, Zalewski C, Kim HJ, Solomon B et al. Phenotype and Course of Hutchinson–Gilford Progeria Syndrome. New Engl J Med. 2008;358:592-604. 3. Gordon LB, Shappell H, Massaro J, D'Agostino RB, Brazier J, Campbell SE, Kleinman ME and Kieran MW. Association of Lonafarnib Treatment vs No Treatment With Mortality Rate in Patients With Hutchinson-Gilford Progeria Syndrome. JAMA. 2018;319:1687. 4. De Sandre-Giovannoli A, Bernard R, Cau P, Navarro CL, Amiel J, Boccaccio I, Lyonnet S, Stewart CL, Munnich A, Le Merrer M et al. Lamin a truncation in Hutchinson-Gilford progeria. Science. 2003;300:2055. 5. Eriksson M, Brown WT, Gordon LB, Glynn MW, Singer J, Scott L, Erdos MR, Robbins CM, Moses TY, Berglund P et al. Recurrent de novo point mutations in lamin A cause Hutchinson-Gilford progeria syndrome. Nature. 2003;423:293-298. 6. Ullrich NJ and Gordon LB. Hutchinson–Gilford progeria syndrome. Handb Clin Neurol. 2015;132:249-264. 7. Hamczyk MR, del Campo L and Andrés V. Aging in the Cardiovascular System: Lessons from Hutchinson-Gilford Progeria Syndrome. Ann Rev Physiol. 2018;80:27-48. 8. Gordon LB, Kleinman ME, Miller DT, Neuberg DS, Giobbie-Hurder A, Gerhard-Herman M, Smoot LB, Gordon CM, Cleveland RH, Snyder BD et al. Clinical trial of a farnesyltransferase inhibitor in children with Hutchinson-Gilford progeria syndrome. Proc Nat Acad Sci USA. 2012;109:16666-16671. 9. Gordon LB, Massaro J, D'Agostino RB, Campbell SE, Brazier J, Brown WT, Kleinman ME and Kieran MW. Impact of farnesylation inhibitors on survival in Hutchinson-Gilford Progeria Syndrome. Circulation. 2014;130:27-34. 10. Gordon LB, Kleinman ME, Massaro J, D'Agostino RB, Shappell H, Gerhard-Herman M, Smoot LB, Gordon CM, Cleveland RH, Nazarian A et al. Clinical trial of the protein farnesylation inhibitors Lonafarnib, Pravastatin, and Zoledronic Acid in children with Hutchinson-Gilford Progeria Syndrome. Circulation. 2016;134:114-125. 11. Russell WMS and Burch RL. The principles of humane experimental technique. London, UK: Methuen; 1959. 12. Kilkenny C, Browne WJ, Cuthill IC, Emerson M and Altman DG. Improving bioscience research reporting: the ARRIVE guidelines for reporting animal research. PLoS Biol. 2010;8:e1000412. 13. Ruzankina Y, Pinzon-Guzman C, Asare A, Ong T, Pontano L, Cotsarelis G, Zediak VP, Velez M, Bhandoola A and Brown EJ. Deletion of the developmentally essential gene ATR in adult mice leads to age-related phenotypes and stem cell loss. Cell Stem cell. 2007;1:113-126. 25 10.1161/CIRCULATIONAHA.121.055313 14. Osorio FG, Navarro CL, Cadiñanos J, López-Mejía IC, Quirós PM, Bartoli C, RiveraTorres J, Tazi J, Guzmán G, Varela I et al. Splicing-directed therapy in a new mouse model of human accelerated aging. Sci Trans Med. 2011;3:106-107. 15. Holtwick R, Gotthardt M, Skryabin B, Steinmetz M, Potthast R, Zetsche B, Hammer RE, Herz J and Kuhn M. Smooth muscle-selective deletion of guanylyl cyclase-A prevents the acute but not chronic effects of ANP on blood pressure. Proc Nat Acad Sci USA. 2002;99:7142-7147. 16. Behringer R, Gertsenstein M, Vintersten K and Nagy A. Manipulating the mouse embryo: a laboratory manual. Fourth Edition: Cold Spring Harbor Laboratory Press; 2014. 17. Cheetham SW, Gruhn WH, van den Ameele J, Krautz R, Southall TD, Kobayashi T, Surani MA and Brand AH. Targeted DamID reveals differential binding of mammalian pluripotency factors. Development. 2018;145:dev170209. 18. Yusa K, Zhou L, Li MA, Bradley A and Craig NL. A hyperactive piggyBac transposase for mammalian applications. Proc Nat Acad Sci USA. 2011;108:1531-1536. 19. Yang SH, Qiao X, Farber E, Chang SY, Fong LG and Young SG. Eliminating the synthesis of mature lamin A reduces disease phenotypes in mice carrying a HutchinsonGilford progeria syndrome allele. J Biol Chem. 2008;283:7094-7099. 20. Hamczyk MR, Villa-Bellosta R, Gonzalo P, Andrés-Manzano MJ, Nogales P, Bentzon JF, López-Otín C and Andrés V. Vascular smooth muscle-specific progerin expression accelerates atherosclerosis and death in a mouse model of Hutchinson-Gilford Progeria Syndrome. Circulation. 2018;138:266-282. Downloaded from http://ahajournals.org by on October 27, 2021 21. Rivera-Torres J, Calvo CJ, Llach A, Guzmán-Martínez G, Caballero R, González-Gómez C, Jiménez-Borreguero LJ, Guadix JA, Osorio FG, López-Otín C et al. Cardiac electrical defects in progeroid mice and Hutchinson-Gilford progeria syndrome patients with nuclear lamina alterations. Proc Nat Acad Sci USA. 2016;113:E7250. 22. Fanjul V, Jorge I, Camafeita E, Macías A, González-Gómez C, Barettino A, Dorado B, Andrés-Manzano MJ, Rivera-Torres J, Vázquez J et al. Identification of common cardiometabolic alterations and deregulated pathways in mouse and pig models of aging. Aging Cell. 2020;19:e13203. 23. Dorado B, Pløen GG, Barettino A, Macías A, Gonzalo P, Andrés-Manzano MJ, GonzálezGómez C, Galán-Arriola C, Alfonso JM, Lobo M et al. Generation and characterization of a novel knockin minipig model of Hutchinson-Gilford progeria syndrome. Cell Discov. 2019;5:16. 24. Macías A, Díaz-Larrosa JJ, Blanco Y, Fanjul V, González-Gómez C, Gonzalo P, AndrésManzano MJ, da Rocha AM, Ponce-Balbuena D, Allan A et al. Paclitaxel mitigates structural alterations and cardiac conduction system defects in a mouse model of Hutchinson-Gilford progeria syndrome. Cardiovasc Res. 2021; Feb 24:cvab055. doi: 10.1093/cvr/cvab055. Epub ahead of print. PMID: 33624748. 25. Stehbens WE, Delahunt B, Shozawa T and Gilbert-Barness E. Smooth muscle cell depletion and collagen types in progeric arteries. Cardiovasc Pathol. 2001;10:133-136. 26. Varga R, Eriksson M, Erdos MR, Olive M, Harten I, Kolodgie F, Capell BC, Cheng J, Faddah DA, Perkins S et al. Progressive vascular smooth muscle cell defects in a mouse 26 10.1161/CIRCULATIONAHA.121.055313 model of Hutchinson-Gilford progeria syndrome. Proc Nat Acad Sci USA. 2006;103:32503255. 27. Lee JM, Nobumori C, Tu Y, Choi C, Yang SH, Jung H-J, Vickers TA, Rigo F, Bennett CF, Young SG et al. Modulation of LMNA splicing as a strategy to treat prelamin A diseases. J Clin Invest. 2016;126:1592-1602. 28. Olive M, Harten I, Mitchell R, Beers JK, Djabali K, Cao K, Erdos MR, Blair CD, Funke B, Smoot LB et al. Cardiovascular pathology in Hutchinson-Gilford progeria: correlation with the vascular pathology of aging. Arterioscl Thromb Vasc Biol. 2010;30:2301-2309. 29. del Campo L, Sánchez‐López A, Salaices M, von Kleeck RA, Expósito E, González‐ Gómez C, Cussó L, Guzmán‐Martínez G, Ruiz‐Cabello J, Desco M et al. Vascular smooth muscle cell specific progerin expression in a mouse model of Hutchinson–Gilford progeria syndrome promotes arterial stiffness: Therapeutic effect of dietary nitrite. Aging Cell. 2019;18:e12936. 30. Del Campo L, Sánchez-López A, González-Gómez C, Andrés-Manzano MJ, Dorado B and Andrés V. Vascular Smooth Muscle Cell-Specific Progerin Expression Provokes Contractile Impairment in a Mouse Model of Hutchinson-Gilford Progeria Syndrome that Is Ameliorated by Nitrite Treatment. Cells. 2020;9:656. 31. Lepore JJ, Cheng L, Min Lu M, Mericko PA, Morrisey EE and Parmacek MS. Highefficiency somatic mutagenesis in smooth muscle cells and cardiac myocytes in SM22alpha-Cre transgenic mice. Genesis. 2005;41:179-84. Downloaded from http://ahajournals.org by on October 27, 2021 32. Hamczyk MR, Villa-Bellosta R, Quesada V, Gonzalo P, Vidak S, Nevado RM, AndrésManzano MJ, Misteli T, López-Otín C and Andrés V. Progerin accelerates atherosclerosis by inducing endoplasmic reticulum stress in vascular smooth muscle cells. EMBO Mol Med. 2019;11:e9736. 33. von Kleeck R, Roberts E, Castagnino P, Bruun K, Brankovic SA, Hawthorne EA, Xu T, Tobias JW and Assoian RK. Arterial stiffness and cardiac dysfunction in HutchinsonGilford Progeria Syndrome corrected by inhibition of lysyl oxidase. Life Sci Alliance. 2021;4:e202000997. 34. Fong LG. Prelamin A and lamin A appear to be dispensable in the nuclear lamina. J Clin Invest. 2006;116:743-752. 35. López-Mejía IC, de Toledo M, Chavey C, Lapasset L, Cavelier P, López-Herrera C, Chebli K, Fort P, Beranger G, Fajas L et al. Antagonistic functions of LMNA isoforms in energy expenditure and lifespan. EMBO Rep. 2014;15:529-539. 36. Koblan LW, Erdos MR, Wilson C, Cabral WA, Levy JM, Xiong ZM, Tavarez UL, Davison LM, Gete YG, Mao X et al. In vivo base editing rescues Hutchinson-Gilford progeria syndrome in mice. Nature. 2021;589:608-614. 37. Santiago-Fernández O, Osorio FG, Quesada V, Rodríguez FJ, Basso S, Maeso D, Rolas L, Barkaway A, Nourshargh S, Folgueras AR et al. Development of a CRISPR/Cas9-based therapy for Hutchinson–Gilford progeria syndrome. Nat Med. 2019;25:423-426. 38. Beyret E, Liao HK, Yamamoto M, Hernandez-Benitez R, Fu Y, Erikson G, Reddy P and Izpisua Belmonte JC. Single-dose CRISPR-Cas9 therapy extends lifespan of mice with Hutchinson-Gilford progeria syndrome. Nat Med. 2019;25:419-422. 27 10.1161/CIRCULATIONAHA.121.055313 39. Erdos MR, Cabral WA, Tavarez UL, Cao K, Gvozdenovic-Jeremic J, Narisu N, Zerfas PM, Crumley S, Boku Y, Hanson G et al. A targeted antisense therapeutic approach for Hutchinson-Gilford progeria syndrome. Nat Med. 2021;27:536-545. 40. Puttaraju M, Jackson M, Klein S, Shilo A, Bennett CF, Gordon L, Rigo F and Misteli T. Systematic screening identifies therapeutic antisense oligonucleotides for HutchinsonGilford progeria syndrome. Nat Med. 2021;27:526-535. 41. Capell BC, Olive M, Erdos MR, Cao K, Faddah DA, Tavarez UL, Conneely KN, Qu X, San H, Ganesh SK et al. A farnesyltransferase inhibitor prevents both the onset and late progression of cardiovascular disease in a progeria mouse model. Proc Nat Acad Sci USA. 2008;105:15902-15907. 42. Sagelius H, Rosengardten Y, Schmidt E, Sonnabend C, Rozell B and Eriksson M. Reversible phenotype in a mouse model of Hutchinson-Gilford progeria syndrome. J Med Genet. 2008;45:794-801. 43. Strandgren C, Nasser HA, McKenna T, Koskela A, Tuukkanen J, Ohlsson C, Rozell B and Eriksson M. Transgene silencing of the Hutchinson-Gilford progeria syndrome mutation results in a reversible bone phenotype, whereas resveratrol treatment does not show overall beneficial effects. FASEB J. 2015;29:3193-3205. 44. Sanger F, Nicklen S and Coulson AR. DNA sequencing with chain-terminating inhibitors. Proc Natl Acad Sci USA. 1977;74:5463-5467. 45. Bradford MM. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem. 1976;72:248-254. Downloaded from http://ahajournals.org by on October 27, 2021 46. Bonzon-Kulichenko E, Pérez-Hernández D, Nuñez E, Martínez-Acedo P, Navarro P, Trevisán-Herraz M, Ramos MC, Sierra S, Martínez-Martínez S, Ruiz-Meana M et al. A robust method for quantitative high-throughput analysis of proteomes by 18O labeling. Mol Cell Proteomics. 2011;10:M110 003335. 28 10.1161/CIRCULATIONAHA.121.055313 Table 1. Age of LmnaHGPSrev-Ubc-CreERT2 mice at the time of initiation of oil or tamoxifen administration Disease stage (age initiation oil/tamoxifen administration) Age at initiation of oil/tamoxifen administration (mean ± SD) Experiments Early disease (≈6-month-old; symptoms emerging) 24.3 ± 2.15 weeks Body-weight evolution and survival Intermediate disease (≈9-month-old; mild symptoms) 35.9 ± 3.2 weeks Electrocardiography and histopathology Late disease (≈13-month-old; severe symptoms) 54.4 ± 1.97 weeks Body-weight evolution and survival SD, standard deviation Downloaded from http://ahajournals.org by on October 27, 2021 29 10.1161/CIRCULATIONAHA.121.055313 Figure legends Figure 1. LmnaHGPSrev/HGPSrev (HGPSrev) mice exhibit ubiquitous progerin expression and undetectable lamin A expression. (A) CRISPR-Cas9 strategy for generating HGPSrev mice (see details in Methods and Supplemental Fig. S1A). Cre activity generates a Lmna “reverted” allele that causes progerin suppression and lamin A restoration. (B) Representative immunofluorescence images showing progerin expression (white) and nuclei (blue) in WT and HGPSrev mice. Scale bar, 25 µm. (C) Western blot of lamin A/C, progerin and GAPDH in 2-month-old WT and HGPSrev mice. Six mice of each genotype were analyzed, and representative images are shown of two mice of each genotype. The graphs show the relative amount of progerin normalized using lamin C and GAPDH as controls. (n=3-13 WT mice; n=4-12 HGPSrev mice). Statistical analysis was performed by two-tailed t-test. *, p<0.05; **, p<0.01; ***, p<0.001; ****, p<0.0001. A, Downloaded from http://ahajournals.org by on October 27, 2021 adventitia; L, lumen; M, media; SKM, skeletal muscle. Figure 2: Targeted precursor-reaction monitoring (PRM) analysis to examine progerin farnesylation in mouse heart lysates. (A) Workflow for the LC-MS/MS analysis of proteins extracted from mouse hearts and immunoprecipitated with anti-lamin A/C antibodies that recognize lamin A, lamin C, and progerin. For each genotype, each sample was the pool of 3 hearts. WT, wild-type mice; G609G, LmnaG609G/G609G mice; HGPSrev, LmnaHGPSrev/HGPSrev mice. (B) Western blots using anti-lamin A/C antibody to check the enrichment of lamin A, lamin C, and progerin in the immunoprecipitated material and supernatant (+, immunoprecipitated; -, supernatant). Controls included samples containing only beads and antibody (CT1) and only beads and protein extract (CT2). A 10 µL aliquot of each sample was loaded onto the gel; see details in Supplemental 30 10.1161/CIRCULATIONAHA.121.055313 Methods. (C) Surrogate peptides used to detect mature lamin A and progerin: IC, internal control peptide (present in both lamin A and progerin); LA, lamin A peptide (specific for lamin A); FP, farnesylated progerin peptide (specific for progerin). (D) MS2 fragmentation spectrum from FP obtained in the PRM assay. The insert shows ion ascription to the main fragment-ion series (Cterminal y-series and N-terminal b-series). (E) MS2 extracted ion chromatograms of IC, LA, and FP peptides obtained from the time-scheduled PRM assay for the detection of lamin A and progerin. The ion traces were obtained using fragment ion y+9 from IC, y+8 from LA, and b+8 from FP. Figure 3. Progeroid phenotype in LmnaHGPSrev/HGPSrev (HGPSrev) mice with ubiquitous progerin expression. (A) Postnatal body-weight curves (n=14 WT; n=22 HGPSrev). Differences were analyzed by Downloaded from http://ahajournals.org by on October 27, 2021 unpaired multiple t-tests and the Holm-Sidák correction. (B) Representative images of ≈8- and ≈13-month-old mice. Scale bar, 2 cm. (C) Representative images of hematoxylin & eosin-stained skin from ≈13-month-old mice and the results of subcutaneous fat layer (SFL) score and thickness quantification (see Methods) (n=13-14 WT; n=11-12 HGPSrev). Statistical analysis was performed by two-tailed t-test. Scale bar, 500 µm. (D) Representative images of sagittal whole-body cross-sections obtained by MRI (fat shown in white) and quantification of body-fat mass and percentage fat content in ≈13-month-old mice (n=23 WT; n=10 HGPSrev). Differences were analyzed by two-tailed t-test. (E) Kaplan-Meier survival curve (n=13 WT; n=22 HGPSrev). Differences were analyzed by the Mantel-Cox test. ***, p<0.001; ****, p<0.0001. Data are mean±SEM. Each symbol represents one animal. 31 10.1161/CIRCULATIONAHA.121.055313 Figure 4. Vascular smooth muscle cell (VSMC) content and collagen deposition in the aortas of ≈8-month-old HGPSrev mice. (A) Representative immunofluorescence of cross-sections of aortic arch (LEFT) and thoracic aorta (RIGHT) stained with anti-smooth muscle α-actin (SMA) antibody (red) and Hoechst 33342 (blue) to visualize VSMCs and nuclei, respectively. Graphs show quantification of VSMC content in the media as either the percentage (%) of SMA-positive area or nuclear density (n=6-8 WT; n=5-7 HGPSrev). Scale bar, 150 µm. (B) Representative images and quantification of Masson’s trichrome staining to visualize medial and adventitial collagen content in crosssections of aortic arch (LEFT) and thoracic aorta (RIGHT) (n=11-13 WT; n=11-13 HGPSrev). Scale bar, 50 µm. Data are mean±SEM. Each symbol represents one animal. Statistical analysis was performed by two-tailed t-test (*, p<0.05). A, adventitia; L, lumen; M, media. Downloaded from http://ahajournals.org by on October 27, 2021 Figure 5: Cardiovascular abnormalities in ≈13-month-old HGPSrev mice. (A) Representative immunofluorescence images of aortic arch (LEFT) and thoracic aorta (RIGHT). Specimens were co-stained with anti-smooth muscle α-actin (SMA) antibody (red) and Hoechst 33342 (blue) to visualize vascular smooth muscle cells (VSMCs) and nuclei, respectively. Graphs show quantification of VSMC content in the media as either the percentage (%) of SMA-positive area or nuclear density (n=4-5 WT; n=7 HGPSrev). Scale bar, 150 µm. Data are mean±SEM. Statistical analysis was performed by two-tailed t-test (**, p<0.01; ***, p<0.001; ****, p<0.0001). (B) Representative images and quantification of Masson’s trichrome staining to visualize medial and adventitial collagen content in aortic arch (LEFT) and thoracic aorta (RIGHT) of ≈13-month-old mice (n=13-17 WT; n=13-14 HGPSrev). Scale bar, 50 µm. Data are mean±SEM. Statistical analysis was performed by two-tailed t-test (***, p<0.001; ****, p<0.0001). (C) Longitudinal electrocardiography (ECG) assessment (n=12-23 WT; n=14- 32 10.1161/CIRCULATIONAHA.121.055313 19 HGPSrev). Data are median with interquartile range±minima and maxima. Differences were analyzed by mixed-effects analysis using the Geisser-Greenhouse correction and Sidák’s multiple comparisons test. Differences over time within each genotype: §, p<0.05; §§, p<0.01; §§§§, p<0.0001. Differences between genotypes at each timepoint: *, p<0.05; ***, p<0.001; ****, p<0.0001. Each symbol represents one animal. A, adventitia; M, media; L, lumen. Figure 6. In vitro and in vivo tamoxifen-induced Cre-dependent progerin suppression and lamin A restoration. (A) Wild-type (WT) and HGPSrev mouse embryonic fibroblasts (MEFs) were co-transfected with plasmids to confer resistance to zeocin and express a tamoxifen-inducible Cre recombinase. (B) Zeocin-resistant MEFs were analyzed by western blot to examine lamin A/C, progerin and GAPDH expression. Equal volumes of ethanol or tamoxifen (25 nM final concentration) were added to the cells as indicated. The graph shows the relative amount of progerin and lamin A in Downloaded from http://ahajournals.org by on October 27, 2021 HGPSrev MEFs (normalized to lamin C content). (C, D) Western blot analysis of tissues of LmnaHGPSrev/HGPSrev Ubc-CreERT2-tg/+ mice which received vehicle (oil) or tamoxifen beginning at the age of ≈3 months (C, n=4 each group) and ≈13 months (D, n=6 each group). Mice in C were euthanized 1 week after oil or tamoxifen administration, and mice in D when they met human end-point criteria. Yellow arrowheads in D indicate one animal in which tamoxifen administration did not suppress progerin or induce lamin A and that died 2 days after the end of tamoxifen administration (see Figure 7B, bottom right). Quantification of the relative amounts of lamin A and progerin in the blots in C is shown in Supplemental Figure S5). The graphs in D show the relative amount of progerin and lamin A normalized to lamin C and GAPDH content. Statistical analysis to compare genotypes was performed by two-tailed t-test. *, p<0.05; **, p<0.01; ***, p<0.001; ****, p<0.0001. 33 10.1161/CIRCULATIONAHA.121.055313 Figure 7: Ubiquitous progerin suppression and lamin A restoration extends lifespan in both mildly and severely symptomatic HGPSrev-Ubc-CreERT2 mice. (A) Experimental protocol for studies with HGPSrev-Ubc-CreERT2 mice, showing the age at which oil or tamoxifen administration started (details in Table 1). (B) Oil or tamoxifen were administered at ≈6 months (LEFT: n=18 Oil and n=22 Tamoxifen) or ≈13 months (RIGHT: n=720 Oil and n=9-23 Tamoxifen). The graphs show the results from two independent experiments. Differences were analyzed by unpaired multiple t-tests and the Holm-Sidák correction in bodyweight studies and by the Mantel-Cox test in Kaplan-Meier survival curves. (C) Oil or tamoxifen were administered at ≈9 months of age, and ECG was performed at the indicated ages (n=7-10 Oil; n=11-12 Tamoxifen). Data are medians with interquartile range±minima and maxima. Differences were analyzed by mixed-effects analysis with the Geisser-Greenhouse correction and Sidák’s multiple comparisons test. (D) Hematoxylin & eosin (H&E) and Masson’s trichrome staining of aortic cross-sections from mice receiving oil/tamoxifen at ≈9 months and sacrificed at Downloaded from http://ahajournals.org by on October 27, 2021 14.5 months of age (n=5-6 Oil; n=11 Tamoxifen). A group of age-matched untreated WT mice was included for comparison (n=7-9). Scale bar, 50 µm. Data are mean±SEM. Differences were analyzed by one-way ANOVA and the post-hoc Tukey test. **, p<0.01; ***, p<0.001; ****, p<0.0001. Each symbol represents one animal. A, adventitia; L, lumen; M, media; NS, nonspecific band. Figure 8: Normal vascular phenotype and lifespan in HGPSrev-SM22α-Cre mice with progerin suppression and lamin A restoration restricted to vascular smooth muscle cells (VSMCs) and cardiomyocytes. (A) Representative immunofluorescence images of thoracic aorta and hearts of ≈13-month-old mice. Cross-sections were co-stained with antibodies against CD31 (green), smooth muscle αactin (SMA) (red) and progerin (white) and with Hoechst 33342 (blue) to visualize endothelial 34 10.1161/CIRCULATIONAHA.121.055313 cells, VSMCs, progerin, and nuclei, respectively. (B) Western blot of lamin A/C, progerin and GAPDH in tissues of ≈13-month-old mice. (C) Body-weight curves (n=9 WT; n=13 HGPSrev; n=11 HGPSrev-SM22α-Cre). Differences were analyzed by unpaired multiple t-tests and the Holm-Sídák correction. Red asterisks denote differences between HGPSrev-SM22α-Cre and HGPSrev mice. Black asterisks denote differences between HGPSrev-SM22α-Cre and WT mice. (D) Kaplan-Meier survival curve (n=15 WT; n=15 HGPSrev; n=11 HGPSrev-SM22α-Cre). Median lifespan was 13.73 months in HGPSrev mice, 22.4 months in HGPSrev-SM22α-Cre mice, and 22.97 months in WT mice. Differences were analyzed with the Mantel-Cox test. (E, F) Representative images of aortic arch stained with hematoxylin & eosin (H&E) and Masson’s trichrome, to quantify VSMCs and fibrosis, respectively, in ≈13-month-old WT mice (n=6), HGPSrev mice (n=5-6), and HGPSrev-SM22α-Cre mice (n=5). Differences were analyzed by one-way ANOVA with the post-hoc Tukey test. *, p<0.05; ****, p<0.0001. Each symbol represents one animal. Data are mean±SEM. A, adventitia; L, lumen; M, media. Scale bars, 25 Downloaded from http://ahajournals.org by on October 27, 2021 µm (except the ones in the tile scans in panel A, where they account for 200 µm). 35 DNA repair template (2,487 bp) Lmna ∆150 cDNA E11-12 10 RHA 11 Cas9 gRNA Lmna ∆150 cDNA E11-12 allele (WT) 1 2 LmnaHGPSrev allele (HGPSrev) 1 3 4 5 6 7 8 9 10 Intron 10 11 Progerin cDNA LoxP site Transcription stop signal 12 Lmna “reverted” allele 2 3 4 5 6 7 8 9 10 Lmna ∆150 cDNA E11-12 STOP Lmna+ Lmna exons LHA: Left Homology Arm (938 bp) RHA: Right Homology Arm (877 bp) 12 STOP Insert (672 bp) LHA STOP A 11 Cre 12 1 2 3 4 5 9 11 10 Lamin C Lamin C Lamin A Progerin B 6 7 8 WT HGPSrev L L M Thoracic aorta A A Adv. M 25 µm Progerin Heart Hoechst Aorta 75 37 Heart 75 37 Skeletal muscle 75 37 WT HGPSrev Lamin A Progerin Lamin C WT GAPDH Progerin/Lamin C ratio kDa HGPSrev 2.5 **** 2.0 ** * **** 1.5 **** *** 1.0 0.5 0.0 Aorta Heart SKM Kidney Spleen Liver 37 Spleen 75 37 75 37 Progerin/GAPDH ratio Kidney 75 Liver Downloaded from http://ahajournals.org by on October 27, 2021 C Liver 2.0 **** 1.5 ** *** * 1.0 * *** Heart SKM 0.5 0.0 Aorta Kidney Spleen Liver 12 A WT G609G Initial lysate (pool 3 hearts per genotype) HGPSrev Western blot Immunoprecipitation of lamin A, lamin C and progerin Supernatant (-) Immunoprecipitate (+) LC-MS/MS analysis 1) Anti-lamin A/C antibody binding to magnetic beads 2) Incubation with lysates 3) Immunoprecipitation of target antigen B Lamin A Lamin C 75 kDa Lamin A Progerin Lamin C 75 kDa 50 kDa 50 kDa C LA FP SVGGSGGGSFGDNLVTR AAGGAGAQSSQNCΔ Farnesyl Lamin A WT mouse Downloaded from http://ahajournals.org by on October 27, 2021 IC C-OCH3 TVLC#GTC#GQPADK IC TVLC#GTC#GQPADK Peptide Description RT schedule m/z z Amino acid sequence IC Internal control peptide: common sequence to progerin and mature Lamin A 0 - 25 min 703.8238 +2 TVLC#GTC#GQPADK LA Mature Lamin A peptide 25 - 50 min 783.8791 +2 SVGGSGGGSFGDNLVTR FP Farnesylated and O-methylated progerin peptide 50 - 100 min 670.3374 +2 AAGGAGAQSSQNCΔ C#, carbamidomethylated Cys; CΔ, farnesylated and O-methylated Cys; PRM, precursor-reaction monitoring; RT, retention time. 40 35 b+3 25 25 b8 b10 b9 b11 y+2 20 15 b+10 b+ 11 10 b+12 5 00 100 200 200 300 400 400 500 600 600 m/z 700 800 800 900 HGPSrev 19.7 19.8 703.82 > 933.41 IC 50 b+8 b+9 G609G 19.7 CH3 b12 b+7 y+ 1 30 b7 1000 1000 1100 1200 1200 Relative intensity 45 200.10 50 50 b6 983.42 b+12–NH3 1000.45 55 b5 868.39 b+11–H2O 886.40 b+5 60 b4 740.33 b+10–H2O 758.34 328.16 340.23 367.17 b+6–H2O 385.18 65 438.21 b+7–H2O 456.22 b+6 70 b3 1100.45 [M+H-farnesyl-NH3-H2O]+ 1118.46 [M+H-farnesyl-NH3]+ 1135.48 [M+H-farnesyl]+ b2 75 75 653.30 b+9–H2O 671.31 80 WT y1 AAGGAGAQSSQNC 566.27 b+8–H2O 584.28 85 y2 100 b+4 454.27 90 257.12 95 E FP b+2 143.08 100 100 Relative abundance D Progerin HGPS mouse 240 30.0 783.88 > 921.48 LA 120 30.0 30.1 670.34 > 584.28 2 74.9 74.9 FP 1 25 50 75 25 50 75 Retention time / min 25 50 75 B *** 40 30 C WT HGPSrev SFL SFL SFL score 6 10 4 8 12 16 20 WT HGPSrev WT HGPSrev **** 4 2 0 WT E HGPSrev 8 **** 6 4 2 0 25 **** 400 200 0 100 80 20 15 10 5 60 **** -43.5% 40 20 0 WT 600 **** % Survival Downloaded from http://ahajournals.org by on Octo Age (months) Fat content (%) 0 SFL average SFLaverage thickness (m) thickness(µm) 20 0 D  13 months  8 months Body fat mass (g) Body weight (g) A HGPSrev 0 0 4 8 12 16 20 24 Age (months) 28 32 A Hoechst SMA M L A M L L WT A M M HGPSrev A L L L M A M A Thoracic aorta L 40 20 0.003 0.002 0.001 0.000 0 B 40 20 0.004 0.003 0.002 0.001 0.000 0 HGPSrev WT Masson’s Trichrome % SMA+ area (media) Nuclei/Area (media) 0.004 Nuclei/Area (media) 60 WT HGPSrev L L L M L A M M M A A A Aortic arch 6 4 2 0 80 4 60 40 20 0 3 2 1 0 WT HGPSrev % collagen (adventitia) 8 * Thoracic aorta % collagen (media) % collagen (media) 10 % collagen (adventitia) Downloaded from http://ahajournals.org by on October 27, 2021 + M % SMA area (media) Aortic arch 60 40 20 0 A SMA WT Hoechst HGPSrev M A L M A L L M A A M L Thoracic aorta 40 30 20 10 0 0.003 0.002 0.001 0.000 B HGPSrev **** *** 0.004 30 20 10 0.003 0.002 0.001 0.000 0 WT HGPSrev A A A M M M L L L L M A **** 40 20 0 C 80 20 *** % collagen (media) % collagen (adventitia) 60 60 40 20 0 % collagen (adventitia) Thoracic aorta Aortic arch % collagen (media) Downloaded from http://ahajournals.org by on October 27, 2021 Masson’s Trichrome WT 40 Nuclei/Area (media) ** 0.004 % SMA+ area (media) *** 50 Nuclei/Area (media) % SMA+ area (media) Aortic arch **** 15 10 5 0 10 * *** 20 **** 5 50 40 30 § 25 §§ QT (ms) T-wave steepness (mV/s) §§§§ 60 * **** 10 13 15 10 5 0 0 4 8 10 13 4 8 Age (months) Age (months) WT HGPSrev A B MEFs (WT & HGPSrev) 75 kDa Lamin A Progerin Lamin C 37 kDa Progerin/Lamin C ratio Vehicle + zeocin Cre-ER (zeocin resistant) 24h 72h after tamoxifen 48h 0.4 0.8 0.3 0.6 0.2 0.4 0.1 0.2 0.0 0.0 Vehicle 24 48 72 Lamin A/Lamin C ratio pPB-CAG-ER-CreER-IRES-Zeocin pCMV-hyPBase (Transposase) GAPDH Time after tamoxifen (h) C Oil Tamoxifen Start at ≈ 3 months Tamoxifen Oil NS 75 kDa Aorta Heart 75 kDa Lamin A Progerin Lamin C 37 kDa GAPDH 37 kDa NS 37 kDa 37 kDa Oil HGPSrev-UbCreERT2 WT HGPSrev-UbCreERT2 Tamoxifen HGPSrev-UbCreERT2 Start at ≈ 13 months 75 kDa Tamoxifen Oil 75 kDa Lamin A Progerin Lamin C Liver Liver 37 kDa 75 kDa 75 kDa GAPDH Heart Heart 37 kDa 37 kDa 37 kDa 0.6 **** *** 0.4 0.2 0.0 Liver Heart 0.8 0.6 HGPSrev-UbCreERT2 *** *** 0.4 0.2 0.0 Liver Heart Oil 0.5 ** 0.4 0.3 * 0.2 0.1 0.0 Liver Heart Tamoxifen Lamin A/GAPDH ratio 0.8 Progerin/GAPDH ratio Progerin/Lamin C ratio HGPSrev-UbCreERT2 Lamin A/Lamin C ratio Downloaded from http://ahajournals.org by on October 27, 2021 D 75 kDa Kidney Liver 75 kDa 0.4 * 0.3 0.2 ** 0.1 0.0 Liver Heart 6 months No symptoms C 20 10 15 40 30 20 10 5 10 0 0 0 4 8 12 16 20 24 28 0 4 8 Age (months) 20 24 10 28 80 80 % Survival 100 **** +84.5% * +6.7% 40 20 0 0 12 16 20 24 28 0 4 8 Age (months) L Oil M A L L 0 10 M **** ** **** 13 15 Age (months) Masson’s Trichrome A 80 60 L M **** ** ** 40 20 0 L M A M A L A L M A M L A 2 2 Nuclei/mm (media) 2 0 28 WT A A 4 5 L M 6 24 10 H&E M L Tamoxifen Aortic arch WT L A 20 Masson’s Trichrome M A 16 15 15 Age (months) H&E D 12 Nuclei/mm (media) 8 Oil 4 Thoracic aorta 0 % collagen (media) Downloaded from http://ahajournals.org by on October 27, 2021 20 60 13 Age (months) Oil or Tamoxifen (13 months) 100 40 16 Age (months) Oil or Tamoxifen (6 months) 60 12 T-wave steepness (mV/s) 0 % Survival 20 Oil or Tamoxifen (13 months) Body weight (g) 30 * Body weight (g) Oil or Tamoxifen (6 months) 40 WT Tamoxifen B HGPSrev UBC-Cre + Tamoxifen Death QT (ms) HGPSrev UBC-Cre Late disease Early disease HGPSrev UBC-Cre + Oil 5 4 3 2 1 0 L M **** **** ** A % collagen (media) A 13 months Oil or Tamoxifen 80 60 40 20 0 M **** **** *** A B Thoracic aorta WT HGPSrev HGPSrev-SM22αCre HGPSrevHGPSrev SM22αCre WT kDa 75 L M M A 37 GAPDH L A 75 M Heart A Aorta L Lamin A Progerin Lamin C Heart 37 75 Skeletal muscle 3 L 37 2 75 HGPSrev Kidney 1 37 200 µm 2 Spleen 75 1 3 L 37 C Body weight (g) 3 Downloaded from http://ahajournals.org by on October 27, 2021 2 40 30 20 ** 10 0 0 4 8 1 12 16 20 24 D 100 L 200 µm 1 2 3 80 60 **** 40 +63.1% 20 0 0 4 L Myocardium Progerin CD31 SMA H&E WT Hoechst HGPSrev L 12 16 20 24 HGPSrev HGPSrev-SM22αCre HGPSrev-SM22αCre L A A L M M M 4 **** **** 3 2 1 L M L A A L M M % collagen (media) Masson’s Trichrome 0 A 28 Age (months) Endocardium WT A 8 Nuclei/mm2 (media) Epicardium E F 28 Age (months) % Survival HGPSrev-SM22αCre * 50 40 30 20 10 0 **** ****