Overexpression of Wild-Type TMEM43 Improves Cardiac Function in Arrhythmogenic Right Ventricular Cardiomyopathy Type 5
VIEW EDITORIAL:TMEM-ing the Tide: Gene Therapy Holds Promise for ARVC5
Abstract
BACKGROUND:
Arrhythmogenic right ventricular cardiomyopathy type 5 (ARVC5) is the most aggressive type of ARVC, caused by a fully penetrant missense mutation (p.S358L) in TMEM43 (transmembrane protein 43). Pathologically, the disease is characterized by dilation of the cardiac chambers and fibrofatty replacement of the myocardium, which results in heart failure and sudden cardiac death. Current therapeutic options are limited, and no specific therapies targeting the primary cause of the disease have been proposed.
METHODS:
We investigated whether overexpression of wild-type (WT) TMEM43 could overcome the detrimental effects of the mutant form. We used transgenic mouse models overexpressing either WT or mutant (S358L) TMEM43 to generate a double transgenic mouse line overexpressing both forms of the protein. In addition, we explored if systemic delivery of a codon-optimized self-complementary adeno-associated virus bearing WT-TMEM43 could improve disease progression assessed by ECG and echocardiography.
RESULTS:
Double transgenic mice overexpressing both WT and mutant TMEM43 forms showed delayed ARVC5 onset, improved cardiac contraction, and reduced ECG abnormalities compared with mice expressing S358L-TMEM43. In addition, cardiomyocyte death and myocardial fibrosis were reduced, with an overall increase in survival. Finally, we demonstrated that a single systemic administration of an adeno-associated virus carrying codon-optimized WT-TMEM43 prevents ventricular dysfunction and ECG abnormalities induced by S358L-TMEM43.
CONCLUSIONS:
Overexpression of WT-TMEM43 improves the pathological phenotype in a mouse model of ARVC5. Adeno-associated virus-mediated delivery of WT-TMEM43 offers a promising and specific therapy for patients suffering from this highly lethal disease.
Graphical Abstract

Novelty and Significance
What Is Known?
•
Arrhythmogenic Right Ventricular Cardiomyopathy type 5 (ARVC5) is a severe cardiac disorder caused by the p.S358L mutation in TMEM43 (transmembrane protein 43).
•
Current treatments for ARVC5 are focused on the management of symptoms but fail to address the underlying cause of the disease.
What New Information Does This Article Contribute?
•
The p.S358L mutation in TMEM43 does not affect its self-binding ability.
•
Overexpression of wild-type TMEM43 significantly improves cardiac function, reduces arrhythmic events, and extends survival in mice.
•
This study introduces an effective targeted gene therapy strategy to treat ARVC5 that uses adeno-associated virus (AAVs) to deliver codon-optimized wild-type TMEM43 specifically to cardiomyocytes, offering a potential alternative to palliative treatments.
This study tackles the urgent need for specific therapies in ARVC5 by demonstrating that overexpression of wild-type TMEM43 can counteract the deleterious effects of the p.S358L mutation. Using double transgenic mice and an AAV-based gene delivery system, the authors show marked improvements in cardiac contractility, electrical stability, and survival, alongside reductions in myocardial fibrosis. Importantly, the findings suggest that increasing the proportion of wild-type to mutant TMEM43 within protein complexes may be a key mechanism for rescuing cardiac function. This novel therapeutic approach not only deepens our understanding of ARVC5 pathogenesis but also contributes to the rapidly emerging field of gene therapy strategies to treat inherited cardiac disorders.
Meet the First Author, see p 804
Editorial, see p 845
Arrhythmogenic right ventricular cardiomyopathy (ARVC) is an inherited cardiac disorder characterized by chamber dilation and fibrofatty replacement of the myocardium. Individuals present with dizziness, ventricular arrhythmias, and sudden cardiac death.1 A majority of ARVC-associated genetic variants affect proteins of the cardiac desmosome, triggering aberrant signaling pathways that elicit fibrosis and inflammation.2,3 Genetic variants of TMEM43 (transmembrane protein 43) are responsible for the most aggressive subtype of the disease, commonly referred to as ARVC type 5 or ARVC5.4,5 This highly lethal disease has a poor prognosis, especially in men who show a median survival of only 42 years.6,7 Subjects harboring genetic variants in TMEM43 present an autosomal dominant, fully penetrant form of ARVC, with common left ventricle (LV) involvement.6–8
ARVC5 is caused by a point substitution (p.S358L) in a highly conserved transmembrane region of TMEM43.5 This mutation was initially identified in 2008 in Newfoundland (Canada), but subsequent cases have been diagnosed around the globe, including in Spain, the United Kingdom, Germany, and Denmark.5,8–10 TMEM43 has 4 transmembrane domains, and the affected serine in ARVC5 (Ser358) lies within the third of these domains. In a previous work, our group reported a transgenic mouse model of ARVC5 that recapitulates the main characteristics of the human disease.11 Currently, there are no targeted therapies for ARVC5, and only the implantation of an implantable cardioverter-defibrillator (ICD) is offered as a prophylactic treatment to prevent sudden cardiac death. If patients survive ventricular arrhythmia and sudden cardiac death, they often develop end-stage heart failure, and their only alternative is heart transplantation.2,3
Here, we investigated the efficacy of wild-type (WT) TMEM43 overexpression as a potential therapy for the treatment of ARVC5. We observed that overexpression of WT-TMEM43 delays the onset of the ARVC5 phenotype in our transgenic mouse model, significantly improving cardiac contraction, reducing ECG abnormalities and fibrosis, and increasing life span. As a first step toward the translation of this finding to the clinics, we generated an adeno-associated virus (AAV) expressing a codon-optimized WT-TMEM43 that improved the functional and ECG abnormalities caused by S358L-TMEM43. Overall, these data support the therapeutic potential of WT-TMEM43 overexpression in ARVC5 and provide a promising translational tool that could serve as an alternative to current palliative treatments in this highly lethal disease.
Methods
Please see the Major Resources Table in the Supplemental Material.
Data Availability
The data that support the findings of this study are available from the corresponding author upon reasonable request.
Mice
WT male and female C57BL/6J mice were obtained from the animal facility of Centro Nacional de Investigaciones Cardiovasculares. TMEM43wt and TMEM43mut mice, in the C57BL/6J background, express human WT and human S358L-TMEM43, respectively, specifically in cardiomyocytes under the control of the α-myosin heavy chain promoter, as described previously.11 Double transgenic mice were obtained by crossing TMEM43wt and TMEM43mut mice. WT littermates were used as controls. Male and female mice were used throughout the study. Genotyping was performed by Sanger sequencing after polymerase chain reaction (PCR) amplification of human TMEM43 with the following primers: human TMEM43 forward 5’-TCCCAAGTATCCAGAGGTGGGAGACT-3’ and human TMEM43 reverse 5’-AGGCCGGCAATGAGGAGGG-3’ (450 bp amplicon size). Thermocycler conditions were set as follows: initial denaturation at 95 °C for 5 minutes, followed by 37 cycles of denaturation at 98 °C for 30 seconds, annealing at 63 °C for 30 seconds, and extension at 72 °C for 40 seconds.
Mice were housed in an air-conditioned room with a 12-hour light/dark cycle and free access to water and chow. Mice were euthanized in a carbon dioxide chamber. After sacrifice, tibial length was measured, and hearts were weighed. Power calculations were conducted using G*Power software to determine the appropriate sample size required to detect a 10% improvement in ejection fraction, with an SD of ±6.5%, according to previous data.11 A significance level of 0.05 and 80% power was used for the calculations. Based on these parameters, the required sample size per group was found to be at least 8 animals. Given that ARVC5 often presents differently in women and men, a minimum of 8 animals per sex and group were included in the studies. All procedures were approved by the Ethics Committees of the Centro Nacional de Investigaciones Cardiovasculares and the Regional Government of Madrid (PROEX191.5/22).
Echocardiography
Transthoracic echocardiography was performed in mice aged 5, 10, 16, and 24 weeks. Mice were placed on a heating pad and kept under light anesthesia with isoflurane administered via a nose cone. The anesthesia was adjusted to obtain a target heart rate of 500±50 bpm. Cardiac function, chamber dilation, and wall thickness were analyzed from 2-dimensional and M-mode acquisitions. Image acquisitions were performed by a blinded operator using a high-frequency ultrasound system with a 30-MHz probe (Vevo 2100, Visualsonics). Left ventricular ejection fraction (LVEF) and LV end-diastolic volume measurements were obtained from the long axis view, and LV posterior wall thickness in diastole values were analyzed in the short axis view. Right ventricular systolic function was assessed indirectly from the tricuspid annular plane systolic excursion, estimated from maximum lateral tricuspid annulus movement obtained from a 2-dimensional 4-chamber apical view. Images were analyzed offline by an expert blinded to genotype and treatment using the Vevo 2100 Workstation software.
Electrocardiograms and Flecainide Administration
Mice were anesthetized with isoflurane via nose cone for echocardiography. Surface ECGs were obtained by using bipolar limb leads (leads I, II, and III) and unipolar limb leads (leads aVR, aVL, and aVF) for 90 seconds. Recordings were acquired and analyzed by a blinded operator using Acqknowledge 4.1.1 for MP36R (BIOPAC Systems Inc). Mean values of P-wave duration and amplitude, R-wave and S-wave amplitudes, and QRS duration and amplitude were calculated from 3 nonconsecutive beats from lead II. Mice that exhibited premature ventricular contractions in at least one 90-second ECG recording during follow-up were quantified. For experiments with flecainide, 90-second baseline ECG recordings were obtained before flecainide administration via intraperitoneal injection (40 mg/kg). ECG recordings continued for ≈5 minutes postadministration.
Cell Culture and Immunoprecipitation
TMEM43 co-immunoprecipitation experiments were performed in the P19 cell line. P19 cells were transfected with both HA-tagged and GFP (green fluorescent protein)-tagged TMEM43 WT and S358L constructs using Lipofectamine 2000 (11668019; Thermo Fisher Scientific). Cells were lysed with tris-buffered saline buffer supplemented with 1% IGEPAL, 5 mmol/L EDTA, 5 mmol/L MgCl2, and protease and phosphatase inhibitors (5892791001 and 04906845001, respectively, from Roche) for 30 minutes at room temperature. After centrifugation, the supernatants were precleared with Protein G Dynabeads (10003D; Thermo Fisher Scientific). Proteins were immunoprecipitated with Protein G Dynabeads coupled with a rabbit anti-GFP antibody (632592; Takara Bio), rotating at 4 °C overnight. Beads were washed 3× with 0.05% IGEPAL lysis buffer and 5× with lysis buffer without added detergent. Proteins were eluted from beads by boiling for 5 minutes at 95 °C in Laemmli sample buffer. Input, supernatant, and output samples from the co-immunoprecipitation were resolved by SDS-PAGE.
For TMEM43 codon-optimization experiments, HL1 cells were transfected with plasmids carrying a chicken cardiac troponin T promoter driving GFP, human codon-optimized TMEM43, or human WT TMEM43 expression using jetPRIME transfection reagent (POL101000046; Polyplus).
Western Blots
LV myocardium samples or transfected HL1 cells were homogenized by manual grinding in radioimmunoprecipitation assay buffer in the presence of protease and phosphatase inhibitors (5892791001 and 04906845001, respectively, from Roche). Lysates were separated on 10% SDS-PAGE gels (acrylamide:bisacrylamide 29:1), transferred to polyvinylidene fluoride membranes, and blocked with 3% nonfat dry milk in 1X tris-buffered saline for 1 hour. Membranes were incubated with primary antibodies overnight, followed by appropriate horseradish peroxidase-labeled secondary antibodies (anti-mouse P044701 and anti-rabbit P044801-2; Dako). Horseradish peroxidase activity was detected using Radiance ECL chemiluminescent horseradish peroxidase substrate (AC2204; Azure Biosystems). Primary antibodies were as follows: TMEM43 (ab184164; Abcam), GFP (11814460001; Roche), HA (11583816001; Roche), and vinculin (V4505; Sigma). Blot images were obtained and quantified with the Invitrogen iBright CL750 Imaging System and analysis software (Thermo Fisher Scientific).
RNA Isolation and Quantitative Real-Time PCR
LV samples were snap-frozen in liquid nitrogen right after sacrifice. Total RNA was isolated using TRIzol reagent (15596026; Thermo Fisher Scientific). First-strand cDNA was synthesized using 100 ng of total RNA and a High Capacity cDNA Reverse Transcription Kit (4368814; Thermo Fisher Scientific). Quantitative reverse-transcribed PCR was performed in a QuantStudio 5 real-time PCR thermocycler (Thermo Fisher Scientific) using SYBR Green (4367659; Thermo Fisher Scientific) for double-stranded DNA detection. Primer pairs used for quantitative reverse-transcribed PCR are LOX (lysyl oxidase) forward 5’-GCTGCGGAAGAAAACTGC-3’ and LOX reverse 5’-CCTTGGTTCTTCACTCTTTGC-3’ (104 bp amplicon), POSTN (periostin) forward 5’-AACGTCTGTGCCCTCCAG-3’ and POSTN reverse 5’-AGCCTTTCATCCCTTCCATT-3’ (151 bp amplicon), COL1A1 (collagen type I alpha 1 chain) forward 5’-GTGCCACTCTGACTGGAAGA-3’ and COL1A1 reverse 5’-CTGACCTGTCTCCATGTTGC-3’ (100 bp amplicon), COL3A1 (collagen type III alpha 1 chain) forward 5’-CACCCTTCTTCATCCCACTC-3’ and COL3A1 reverse 5’-ATGTCATCGCAAAGGACAGA-3’ (159 bp amplicon), ACTA1 (alpha actin 1) forward 5’-GACCACAGCTGAACGTGAGA-3’, ACTA1 reverse 5’-TGTTGTAGGTGGTCTCATGGAT-3’ (239 bp amplicon), and Luciferase forward 5’-TATCATGGCCTCGTGAAATCC-3’ and Luciferase reverse 5’-TCCTGGGTCCGATTCAATAAAC-3’ (131 bp amplicon). Results were analyzed with the LinReg PCR software,12 which calculates the PCR efficiency of each sample independently. The average efficiency of each mRNA was then used to calculate the relative expression of each gene, which was normalized to that of β-actin.
Histological Analysis, Immunohistochemistry, and Immunofluorescence
Hearts were fixed in 4% paraformaldehyde in PBS for 48 hours, washed in PBS, dehydrated, included in paraffin, and sliced in 5 µm sections. The collagen content was analyzed using Masson trichrome protocol.13 For immunofluorescence, heart sections were subjected to sodium citrate antigen retrieval, permeabilized with 0.1% Tween-20/PBS for 20 minutes, and blocked with blocking solution (5% goat serum, 2% MgCl2, 3% BSA in 0.3% Tween-20/PBS). Sections were incubated with anti-TMEM43 (ab184164; Abcam) diluted 1:100, followed by secondary antibody Alexa 568-labeled goat anti-rabbit (A-11011; Thermo Fisher Scientific) diluted 1:200, and wheat germ agglutinin (WGA) Alexa Fluor 488 conjugate (W11261, Thermo Fisher Scientific) diluted 1:2000. Nuclei were stained with DAPI (4′,6-diamidino-2-phenylindole; D1306; Thermo Fisher Scientific). Samples were mounted in Vectashield mounting medium (H-1000; Vector Laboratories). Antibody-negative tissue sections were used as a control for the primary antibody, while a secondary-only control was included to account for nonspecific binding of the secondary antibody.
Immunofluorescence images were acquired with a Leica SP5 confocal microscope, an HCX PL APO CS 10× 0.4 dry objective, and Leica LAS-AF 2.7.3 software. Images for immunohistochemistry were digitized with NanoZoomer-2.0RS (Hamamatsu). All images were analyzed offline with Fiji, and brightness and contrast were linearly adjusted.
Brillouin Spectroscopy
Brillouin spectra were acquired with a home-modified Olympus BX51 reflected light microscope with a diode-pumped solid-state (DPSS) laser at 532 nm (SpectraPhysics) and a light power below 3.5 mW on the sample coupled to a 3+3 TandemFabry-Perot spectrometer (J. Sandercock, Table Stable Ltd.).14,15 An Olympus MPlan 10X objective with a numerical aperture of 0.25 was used to focus the incident light beam and to collect the scattered light simultaneously. Measurements were performed on dried samples to avoid having to account for water content variability. The tip of the heart apex was carefully sliced to allow the entry of agarose into the ventricular lumen, and samples were embedded in agarose (0.2 g/10 mL distilled water) and sliced into 500 µm sections using a vibratome (Campdem Instruments). A cover slip was placed on top of the sample without sealing. Transparent nail polish was applied on the sides to fix the cover slip. Samples were dried out at room temperature for 5 hours and stored at 4 °C overnight. Brillouin frequency shift, its error, and the half width half maximum were obtained from the Brillouin spectra by a nonlinear least squares fitting procedure using a Pseudo-Voigt function and a cubic polynomial background in MATLAB environment. Laboratory temperature monitoring, displacement of the Märzhäuser Wetzler automated table, and the GHOST multichannel analyzer were controlled with our own developed software using LABVIEW 4 platform.
AAV Vector Production and Delivery
Codon optimization of human TMEM43 was performed using VectorBuilder design studio. Self-complementary AAV vectors codon-optimized wild-type TMEM43 AAV vector (AAV-WT-OPT), AAV vector expressing S358L-TMEM43 (AAV-MUT), and AAV vector expressing luciferase (AAV-LUC) expressing optimized human WT-TMEM43, S358L-TMEM43, and luciferase, respectively, were constructed by VectorBuilder. Centro Nacional de Investigaciones Cardiovasculares’s Viral Vector Unit produced the recombinant AAVs with the AAVMYO myotropic capsid16 in-house using a modified Rep/Cap vector. Briefly, the cap gene from the AAV9 variant in the pAAV2/9n helper plasmid (gift from James M. Wilson, addgene number 112865) was replaced by digestion and ligation with a 3014-bp SwaI+SbfI fragment encoding the AAVMYO viral particle 1 (VP1) gene (obtained by synthesis from the public GenBank sequence deposited under accession: MN365014).
For viral administration, neonatal mice at postnatal day 1 (P1) were lightly anesthetized with ice, and 30 uL of the viral preparation, including 5e10 VP, were injected through the temporal vein in a single injection (1.7e13 VP/kg). Neonates recovered on a heating mat before placing them back together with the dams.
For experiments involving symptomatic adult mice, a subgroup of AAV-MUT mice was randomized into 2 subgroups based on LVEF and QRS amplitude at 10 weeks of age. Mice without signs of disease at this time were excluded from the study (n=7). These subgroups were then treated with 1.7e13 VP/kg of either AAV-LUC or AAV-WT-OPT via the femoral vein.
Statistical Analysis
All statistical analyses were performed using GraphPad Prism (version 10.0.0; GraphPad Software). Data sets were first assessed for normality using the Shapiro-Wilk test. For data meeting the assumption of normality, statistical significance was evaluated using regular 1-way or 2-way ANOVA, followed by Bonferroni posttest for multiple comparisons. For non-normally distributed data, the Kruskal-Wallis test followed by Dunn multiple comparisons test, or multiple Mann-Whitney U tests with Bonferroni-Dunn posttest for multiple comparisons, were used as specified in the figure legends. Survival curves were analyzed using the log-rank (Mantel-Cox) test.
Statistical significance was defined as P<0.05, and exact P values were provided where applicable. Data are presented as mean±SD.
Results
WT-TMEM43 Overexpression in the Heart Improves Cardiac Contractility, Reduces Electrophysiological Abnormalities, and Extends Life Span
We have previously shown using in silico models that WT-TMEM43 and S358L-TMEM43 can form heterodimers. To confirm the models experimentally, we conducted IP experiments using tagged proteins to increase specificity. We found that GFP-tagged S358L-TMEM43 co-immunoprecipitated with HA-tagged S358L-TMEM43 as well as with HA-tagged WT-TMEM43 (Figure 1A). This result validated the in silico models and led us to hypothesize that the overexpression of WT-TMEM43 might quench the deleterious dominant effects of the mutant protein by forming a heterodimer.

To explore whether WT-TMEM43 overexpression could reduce the toxicity of S358L-TMEM43, we crossed our ARVC5 mouse model overexpressing human S358L-TMEM43 (TMEM43mut) with mice overexpressing human WT-TMEM43 (TMEM43wt mice) to generate a double transgenic mouse line that overexpresses both forms of the human protein in a cardiac-specific manner. The functional effects were evaluated through echocardiography and ECG over 24 weeks, and double transgenic mice were compared with TMEM43mut, TMEM43wt, and WT littermates.
The life span of double transgenic mice increased significantly by >10 weeks compared with mice overexpressing the mutant TMEM43, with a median life span of 34.6 versus 23.9 weeks, respectively (Figure 1B). Echocardiography analysis revealed improved LV contraction (LVEF) and reduced dilation in double transgenic mice (Figure 1C and 1D, respectively), while LV wall thickness remained unaltered in all of the groups (Figure 1E). Right ventricular contractility was also improved in double transgenic mice, without any significant deterioration observed until 24 weeks of age (Figure 1F). In addition, double transgenic mice exhibited reduced heart weight to tibial length ratio compared with TMEM43mut mice (Figure 1G).
Overexpression of WT-TMEM43 also improved ECG abnormalities caused by the mutant protein (Figure 2A through 2D). In contrast to the progressive P-wave prolongation observed in TMEM43mut mice, double transgenic mice exhibited normal P-wave duration (Figure 2B). Similarly, double transgenic mice showed reduced QRS duration and increased QRS amplitude compared with TMEM43mut mice at both 16 and 24 weeks of age (Figure 2C and 2D). Furthermore, the proportion of mice showing premature ventricular contractions, a common pathological manifestation in ARVC5 patients, was reduced to levels found in WT mice among double transgenic mice (≈12.5%), compared with TMEM43mut mice (≈35%) (Figure S1A and S1B). In line with these results, the mislocalization of connexin 43 observed in TMEM43mut mice was also partially prevented in double-transgenic mice (Figure S2).

WT-TMEM43 Overexpression Reduces Myocardial Fibrosis
As we previously demonstrated, ARVC5 mice develop extensive myocardial fibrosis as a result of cardiomyocyte death, evidenced by an increase in circulating cardiac troponin I levels.11 As shown in Figure 3A, double transgenic mice showed a delayed rise in circulating troponin I compared with mutant mice, in agreement with the delayed onset of the disease. Moreover, double transgenic mice presented reduced expression of fibrosis markers at week 16, including LOX, which is responsible for collagen and elastin crosslinking, POSTN, and collagens 1 and 3 (COL1A1 and COL3A1), together with a decrease of the cardiac stress marker ACTA1 (Figure 3B through 3F). Masson trichrome staining confirmed a reduced fibrotic content in the heart of double transgenic mice compared with TMEM43mut mice at both 16 and 24 weeks (Figure 4A through 4E). Consistent with these results, TMEM43mut mice showed an increased Brillouin frequency shift, indicating increased stiffness of the myocardial tissue compared with double transgenic, WT, and TMEM43wt mice (Figure 4F). Together, these results demonstrate that WT-TMEM43 overexpression reduces cardiomyocyte death, fibrotic replacement, and myocardial stiffness, leading to improved cardiac function.


WT-TMEM43 Expression Is Enhanced by Codon Optimization
After the positive impact of WT-TMEM43 overexpression in our ARVC5 mice, we developed a gene therapy tool based on AAVs carrying WT-TMEM43 to further explore the therapeutic potential of this approach. To enhance vector efficiency, we first investigated the influence of cDNA codon optimization on the expression of human WT-TMEM43. This process includes substituting rare or nonpreferred codons in the original cDNA sequence with synonymous codons that are more frequently used by the host, likely improving mRNA stability and protein translation efficiency without altering the encoded protein’s amino acid sequence.17 HL1 cells were transfected with control (GFP), human TMEM43 (TMEM43WT), and codon-optimized human TMEM43 (TMEM43WT-OPT) constructs, and protein expression was assessed through western blot. Our results revealed a positive impact of codon optimization on TMEM43 protein expression (Figure S3A and S3B), prompting the utilization of this optimized sequence for the development of a self-complementary myotropic AAV. This vector expresses codon-optimized TMEM43 specifically in the cardiac tissue under the control of the cardiac troponin T promoter, hereafter referred to as AAV-WT-OPT (Figure S3C).
AAV-WT-OPT Prevents Systolic Dysfunction and ECG Abnormalities Caused by Mutant TMEM43
To determine the efficacy of our gene therapy approach and ensure a similar ratio of WT to mutant human TMEM43, we used systemic delivery of myotropic AAVs in neonates through the temporal vein and followed the animals for 24 weeks (Figure 5A). Each neonate received a total of 5e10 VP distributed as follows: 2.5e10 VP of AAV-WT-OPT+2.5e10 VP of AAV-LUC, 2.5e10 VP of AAV carrying S358L-TMEM43 (AAV-MUT)+2.5e10 VP of AAV-LUC, or 2.5e10 VP of AAV-WT-OPT+2.5e10 VP of AAV-MUT.

Western blot and PCR analyses revealed sustained TMEM43 overexpression in mice infected with AAV-WT-OPT or AAV-MUT (Figure 5B; Figure S4). Similarly, mice infected or co-infected with AAV-LUC expressed high levels of luciferase mRNA, as determined by quantitative reverse-transcribed PCR (Figure 5C). Immunofluorescence analysis of human TMEM43 showed overexpression of the protein in 10% to 30% of cardiomyocytes (Figure 5D through 5K), indicating wide biodistribution of the viral infection and cell type-specific protein expression.
Consistent with our TMEM43mut transgenic mice, mice transduced with AAV-MUT+AAV-LUC showed reduced life span (Figure 6A) and a progressive decline in LVEF as assessed by echocardiography (Figure 6B). This cardiac function impairment and life span decline were successfully prevented in mice co-infected with AAV-WT-OPT+AAV-MUT, providing additional support for the therapeutic potential of WT-TMEM43 overexpression. Right ventricular function showed similar trends, although the differences were milder and did not reach statistical significance (Figure 6C). AAV-MUT mice also developed ECG abnormalities, including increased P-wave duration, progressive widening of the QRS, and a gradual decline of the QRS amplitude, mirroring the pathological changes observed in our TMEM43mut transgenic mice. Remarkably, coadministration of AAV-WT-OPT proved sufficient to prevent all these ECG abnormalities (Figure 6D through 6G). In addition, the proportion of mice showing premature ventricular contractions during follow-up was reduced in AAV-WT-OPT+AAV-MUT coinfected mice compared with AAV-MUT mice (Figure S5).

AAV-WT-OPT Prevents Cardiomyocyte Necrosis and Upregulation of Fibrosis-Related Genes
AAV-MUT mice also showed a significant increase in circulating cardiac troponin I levels, albeit to lower levels than TMEM43mut transgenic mice (Figure 7A). This cardiomyocyte death was accompanied by mild but significant increases in the expression of genes related to fibrosis, including LOX, POSTN, and collagens 1 and 3, and the cardiac stress marker ACTA1 (Figure 7B through 7F). Importantly, coinfection with AAV-WT-OPT+AAV-MUT strongly reduced cardiomyocyte death and partially prevented the induction of fibrosis and stress genes (Figure 7A through 7F). Collectively, these findings support that AAV-WT-OPT gene therapy can prevent cardiomyocyte necrosis and the upregulation of fibrosis-related genes, improving cardiac contraction and reducing ECG abnormalities, in agreement with the results obtained in the double transgenic mice.

Treatment of Adult Symptomatic ARVC5 Mice With AAV-WT-OPT Improves Systolic Function and Reduces Arrhythmia Susceptibility
To date, we have demonstrated the potential of WT-TMEM43 overexpression as a preventive therapeutic strategy. To investigate whether late administration of the therapy could also ameliorate the ARVC5 phenotype, we administered AAV-WT-OPT to adult AAV-MUT mice after overt symptoms of the disease were already evident (10 weeks of age), including low LVEF, reduced QRS amplitude, and increased QRS duration. These mice were randomized into 2 groups with matched median LVEF and QRS amplitude. Mice from each group received the same dose of either AAV-LUC or AAV-WT-OPT used in neonates (1.7e13 VP/kg) via the femoral vein (Figure 8A). PCR analyses from 24-week-old mouse hearts exhibited sustained AAV-MUT and AAV-WT-OPT expression in both groups (Figure S6B).

Mice injected with AAV-WT-OPT showed a significant increase in LVEF compared with AAV-LUC-treated mice, lasting for up to 24 weeks (Figure 8B). Regarding QRS amplitude and duration, no significant improvement was observed (Figure 8C and 8D). During the 24-week follow-up period, none of the AAV-WT-OPT treated mice died, whereas 3 of 20 mice (15%) in the AAV-LUC group were found dead (Figure 8E), in line with our previous results at this age (Figure 6A). To uncover any subtle changes in electrical conduction, we administered a supratherapeutic dose of flecainide (40 mg/kg), a class I antiarrhythmic drug that can induce arrhythmias and produce AV block at high doses.18 As shown in Figure 8F and 8G, AAV-WT-OPT treatment reduced the incidence of AV blocks in response to flecainide compared with treatment with AAV-LUC. These findings further support the potential of WT-TMEM43 overexpression therapy in improving cardiac function and electrical manifestations in ARVC5, even when administered in adult symptomatic mice.
Discussion
Gene therapy approaches carrying functional copies of the affected genes, like the one used in this study, have shown to be a powerful tool for tailored therapy in a variety of genetic diseases. Some of these products have already been authorized for clinical practice,19 including AAV-mediated expression of RPE65 for the treatment of retinal dystrophy, SMN1 for spinal muscular atrophy patients, or AADC for aromatic L-amino acid decarboxylase deficiency, among others.20–22 Although no gene therapy has been approved for cardiac diseases yet, several gene therapy approaches are currently being developed for heart failure and genetic cardiomyopathies.23 Specifically in ARVC, the potential of WT copy overexpression in PKP2-ARVC, the most common subtype of ARVC, has recently been demonstrated.24,25 In fact, several products based on this new gene therapy strategy have received Food and Drug Administration approval for phase 1/2 clinical trials (https://www.clinicaltrials.gov; NCT05885412, NCT06109181, and NCT06228924).
Gene therapy approaches are usually designed as replacement therapies for biallelic or haploinsufficient diseases. When addressing dominant negative mutations like p.S358L in TMEM43, therapeutic strategies have mostly focused on antisense oligonucleotides and siRNAs that can downregulate the expression of both WT and affected gene copies, like inotersen, patisiran, and vutrisiran, authorized for targeting the TTR (transthyretin) mRNA in hereditary TTR-mediated amyloidosis.26 However, silencing completely both the affected and unaffected gene copies might not be feasible in certain cardiac diseases, and alternative approaches are warranted. In this sense, our data suggest that functional copy overexpression should also be considered for the treatment of dominant negative diseases, such as ARVC5, without the need for knocking down the mutant allele.
ARVC5 is an incurable disease, and current therapeutic options are limited to preventive or symptomatic treatments, aiming to avoid sudden cardiac death and heart failure.3 In this regard, our group previously showed the potential of enalapril, a commonly used drug in heart failure patients, to preventively treat ARVC5.27 However, although several models have been reported to date, no specific in vivo therapies targeting the primary cause of the disease have been proposed.11,28–31
The data presented here underscore the therapeutic potential of TMEM43 overexpression in ARVC5, with no apparent detrimental effects of TMEM43 overexpression in the heart under healthy conditions. In a first proof-of-concept approach, we generated a double transgenic mouse that overexpresses both WT and mutant TMEM43 in cardiomyocytes. In a second, highly translational approach, we used AAVs to express WT and mutant human TMEM43 under the cardiac troponin T promoter. Both the transgenic mutant model and the mice infected with AAV-MUT recapitulated the main characteristics of the human disease, including cardiomyocyte death, ventricular dysfunction, and ECG abnormalities. Importantly, WT-TMEM43 overexpression successfully improved or delayed most of ARVC5 features in both settings, significantly increasing survival. This scenario resembles heterozygous mutations in channelopathies that lead to heteromeric assemblies carrying both WT and mutated subunits. In that context, the functional changes of the protein complex show a linear dependence on the number of mutant subunits in the complex.32 Given these findings, we can speculate that TMEM43 works as a homomeric assembly and that overexpressing WT-TMEM43 in ARVC5 would increase the ratio of WT to mutant subunits, thereby reducing the functional impact of the mutant protein on the complex (see graphical abstract).
In addition to the p.S358L mutation, our optimized AAV-bearing WT-TMEM43 could potentially be applied to patients harboring different pathogenic or likely pathogenic variants in the TMEM43 gene. Besides the S358L-TMEM43 variant causing ARVC5, 2 heterozygous missense variants, p.Glu85Lys and p.Ile91Val, affecting the hydrophilic domain of TMEM43, showed a phenotype of Emery-Dreifuss-related myopathy involving muscle atrophy with possible cardiac involvement.33 In addition, a p.Arg372Ter nonsense mutation has been associated with deafness in an autosomal dominant auditory neuropathy spectrum disorder.34 These variants showed decreased TMEM43 protein levels, suggesting that restoration of normal protein levels is likely to improve or prevent the pathological phenotype. Furthermore, gene therapy aimed at restoring TMEM43 expression could potentially be applied to other heart diseases that present TMEM43 downregulation, including cardiac hypertrophy or sepsis-induced cardiomyopathy.35,36
Here, we provide evidence that WT-TMEM43 overexpression improves cardiac function in ARVC5 by forming a WT-TMEM43-S358L-TMEM43 heterodimer and that a single systemic administration of our self-complementary myotropic AAV bearing codon-optimized WT-TMEM43 can prevent cardiomyocyte death and improve the disease in mice. While we acknowledge that more research is needed to comprehensively understand TMEM43 function and its role in heart disease, these results collectively suggest the therapeutic benefit of overexpressing WT-TMEM43 in cardiac and, potentially, noncardiac diseases beyond ARVC5.
Study Limitations
Although knock-in models are generally preferred for therapy development, heterozygous TMEM43-S358L knock-in models often show inconsistent results, with an exceedingly delayed onset or even an absence of cardiac phenotypes. Consequently, we used both transgenic and AAV-based ARVC5 models, which reliably reproduce the key functional and electrophysiological abnormalities of the disease. Notably, the AAV-based model exhibited a milder phenotype, which we attribute to lower mutant protein expression compared with the transgenic model.
We propose that overexpression of WT TMEM43 confers a therapeutic benefit by increasing the ratio of WT to mutant subunits within TMEM43 oligomers; however, it remains unclear whether this benefit stems from the oligomerization process itself or from competitive interactions between WT and mutant subunits.
Furthermore, while our mouse gene therapy models have provided promising insights, significant challenges remain in translating these findings to larger mammals and ultimately to human patients. Differences in tissue structure, immune responses, and cardiac electrophysiology may impact the efficiency of AAV-based gene delivery, making future studies in larger animal models essential for clinical application.
Acknowledgments
We thank the Centro Nacional de Investigaciones Cardiovasculares animal facility for mouse breeding, the Viral Vectors Unit for AAV production and the Microscopy Unit for their support during image acquisition and analysis.
Footnote
Nonstandard Abbreviations and Acronyms
- AAV
- adeno-associated virus
- AAV-LUC
- AAV vector expressing luciferase
- AAV-MUT
- AAV vector expressing S358L-TMEM43
- AAV-WT-OPT
- codon-optimized wild-type TMEM43 AAV vector
- ACTA1
- alpha actin 1
- ARVC5
- arrhythmogenic right ventricular cardiomyopathy type 5
- AV
- atrioventricular
- COL1A1
- collagen type I alpha 1 chain
- COL3A1
- collagen type III alpha 1 chain
- GFP
- green fluorescent protein
- HA
- hemagglutinin
- LOX
- lysyl oxidase
- LV
- left ventricle
- LVEF
- left ventricular ejection fraction
- POSTN
- periostin
- TMEM43
- transmembrane protein 43
- TTR
- transthyretin
- VP
- viral particles
- WT
- wild-type
Supplemental Material
References
1.
Corrado D, Basso C, Judge DP. Arrhythmogenic cardiomyopathy. Circ Res. 2017;121:785–802.
2.
Arbelo E, Protonotarios A, Gimeno JR, Arbustini E, Barriales-Villa R, Basso C, Bezzina CR, Biagini E, Blom NA, de Boer RA, et al; ESC Scientific Document Group. 2023 ESC guidelines for the management of cardiomyopathies: developed by the task force on the management of cardiomyopathies of the European Society of Cardiology (ESC). Eur Heart J. 2023;44:3503–3626. doi: 10.1093/eurheartj/ehad194
3.
Marcus FI, McKenna WJ, Sherrill D, Basso C, Bauce B, Bluemke DA, Calkins H, Corrado D, Cox MGPJ, Daubert JP, et al. Diagnosis of arrhythmogenic right ventricular cardiomyopathy/dysplasia: proposed modification of the task force criteria. Eur Heart J. 2010;31:806–814. doi: 10.1093/eurheartj/ehq025
4.
James CA, Jongbloed JDH, Hershberger RE, Morales A, Judge DP, Syrris P, Pilichou K, Domingo AM, Murray B, Cadrin-Tourigny J, et al. International evidence based reappraisal of genes associated with arrhythmogenic right ventricular cardiomyopathy using the clinical genome resource framework. Circ Genom Precis Med. 2021;14:e003273. doi: 10.1161/CIRCGEN.120.003273
5.
Merner ND, Hodgkinson KA, Haywood AFM, Connors S, French VM, Drenckhahn JD, Kupprion C, Ramadanova K, Thierfelder L, McKenna W, et al. Arrhythmogenic right ventricular cardiomyopathy type 5 is a fully penetrant, lethal arrhythmic disorder caused by a missense mutation in the TMEM43 gene. Am J Hum Genet. 2008;82:809–821. doi: 10.1016/j.ajhg.2008.01.010
6.
Hodgkinson K, Connors SP, Merner N, Haywood A, Young TL, McKenna WJ, Gallagher B, Curtis F, Bassett AS, Parfrey PS. The natural history of a genetic subtype of arrhythmogenic right ventricular cardiomyopathy caused by a p.S358L mutation in TMEM43. Clin Genet. 2013;83:321–331. doi: 10.1111/j.1399-0004.2012.01919.x
7.
Hodgkinson KA, Howes AJ, Boland P, Shen XS, Stuckless S, Young TL, Curtis F, Collier A, Parfrey PS, Connors SP. Long-term clinical outcome of arrhythmogenic right ventricular cardiomyopathy in individuals with a p.S358L mutation in TMEM43 following implantable cardioverter defibrillator therapy. Circ Arrhythm Electrophysiol. 2016;9:e003589. doi: 10.1161/CIRCEP.115.003589
8.
Dominguez F, Zorio E, Jimenez-Jaimez J, Salguero-Bodes R, Zwart R, Gonzalez-Lopez E, Molina P, Bermúdez-Jiménez F, Delgado JF, Braza-Boïls A, et al. Clinical characteristics and determinants of the phenotype in TMEM43 arrhythmogenic right ventricular cardiomyopathy type 5. Heart Rhythm. 2020;17:945–954. doi: 10.1016/j.hrthm.2020.01.035
9.
Haywood AFM, Merner ND, Hodgkinson KA, Houston J, Syrris P, Booth V, Connors S, Pantazis A, Quarta G, Elliott P, et al. Recurrent missense mutations in TMEM43 (ARVD5) due to founder effects cause arrhythmogenic cardiomyopathies in the UK and Canada. Eur Heart J. 2013;34:1002–1011.
10.
Milting H, Klauke B, Christensen AH, Müsebeck J, Walhorn V, Grannemann S, Münnich T, Šarić T, Rasmussen TB, Jensen HK, et al. The TMEM43 Newfoundland mutation p.S358L causing ARVC-5 was imported from Europe and increases the stiffness of the cell nucleus. Eur Heart J. 2015;36:872–881. doi: 10.1093/eurheartj/ehu077
11.
Padrón-Barthe L, Villalba-Orero M, Gómez-Salinero JM, Domínguez F, Román M, Larrasa-Alonso J, Ortiz-Sánchez P, Martínez F, López-Olañeta M, Bonzón-Kulichenko E, et al. Severe cardiac dysfunction and death caused by arrhythmogenic right ventricular cardiomyopathy type 5 are improved by inhibition of glycogen synthase kinase-3β. Circulation. 2019;140:1188–1204. doi: 10.1161/CIRCULATIONAHA.119.040366
12.
Ruijter JM, Ramakers C, Hoogaars WMH, Karlen Y, Bakker O, van den Hoff MJB, Moorman AFM. Amplification efficiency: linking baseline and bias in the analysis of quantitative PCR data. Nucleic Acids Res. 2009;37:e45. doi: 10.1093/nar/gkp045
13.
Sridharan D, Pracha N, Dougherty JA, Akhtar A, Alvi SB, Khan M. A one-stop protocol to assess myocardial fibrosis in frozen and paraffin sections. Methods Protoc. 2022;5:13. doi: 10.3390/mps5010013
14.
Rioboó RJJ, Gontán N, Sanderson D, Desco M, Gómez-Gaviro MV. Brillouin spectroscopy: from biomedical research to new generation pathology diagnosis. Int J Mol Sci . 2021;22:8055. doi: 10.3390/ijms22158055
15.
Villalba-Orero M, Jiménez-Riobóo RJ, Gontán N, Sanderson D, López-Olañeta M, García-Pavía P, Desco M, Lara-Pezzi E, Gómez-Gaviro MV. Assessment of myocardial viscoelasticity with Brillouin spectroscopy in myocardial infarction and aortic stenosis models. Sci Rep. 2021;11:24484. doi: 10.1038/s41598-021-04261-0
16.
Weinmann J, Weis S, Sippel J, Tulalamba W, Remes A, El Andari J, Herrmann AK, Pham QH, Borowski C, Hille S, et al. Identification of a myotropic AAV by massively parallel in vivo evaluation of barcoded capsid variants. Nat Commun. 2020;11:1–12.
17.
Inouye S, Sahara-Miura Y, Sato JI, Suzuki T. Codon optimization of genes for efficient protein expression in mammalian cells by selection of only preferred human codons. Protein Expr Purif. 2015;109:47–54.
18.
Cerrone M, Noorman M, Lin X, Chkourko H, Liang FX, van der Nagel R, Hund T, Birchmeier W, Mohler P, van Veen TA, et al. Sodium current deficit and arrhythmogenesis in a murine model of plakophilin-2 haploinsufficiency. Cardiovasc Res. 2012;95:460–468. doi: 10.1093/cvr/cvs218
19.
Shchaslyvyi AY, Antonenko SV, Tesliuk MG, Telegeev GD. Current state of human gene therapy: approved products and vectors. Pharmaceuticals (Basel). 2023;16:1416. doi: 10.3390/ph16101416
20.
Pearson TS, Gupta N, San Sebastian W, Imamura-Ching J, Viehoever A, Grijalvo-Perez A, Fay AJ, Seth N, Lundy SM, Seo Y, et al. Gene therapy for aromatic L-amino acid decarboxylase deficiency by MR-guided direct delivery of AAV2-AADC to midbrain dopaminergic neurons. Nat Commun. 2021;12:1–12.
21.
Aslesh T, Yokota T. Restoring SMN expression: an overview of the therapeutic developments for the treatment of spinal muscular atrophy. Cells. 2022;11:417. doi: 10.3390/cells11030417
22.
Maguire AM, Bennett J, Aleman EM, Leroy BP, Aleman TS. Clinical perspective: treating RPE65-associated retinal dystrophy. Mol Ther. 2021;29:442–463. doi: 10.1016/j.ymthe.2020.11.029
23.
Argirò A, Ding J, Adler E. Gene therapy for heart failure and cardiomyopathies. Rev Esp Cardiol (Engl Ed). 2023;76:1042–1054. doi: 10.1016/j.rec.2023.06.009
24.
Bradford WH, Zhang J, Gutierrez-Lara EJ, Liang Y, Do A, Wang TM, Nguyen L, Mataraarachchi N, Wang J, Gu Y, et al. Plakophilin 2 gene therapy prevents and rescues arrhythmogenic right ventricular cardiomyopathy in a mouse model harboring patient genetics. Nat Cardiovasc Res. 2023;2:1246–1261. doi: 10.1038/s44161-023-00370-3
25.
Kyriakopoulou E, Versteeg D, de Ruiter H, Perini I, Seibertz F, Döring Y, Zentilin L, Tsui H, van Kampen SJ, Tiburcy M, et al. Therapeutic efficacy of AAV-mediated restoration of PKP2 in arrhythmogenic cardiomyopathy. Nat Cardiovasc Res. 2023;2:1262–1276. doi: 10.1038/s44161-023-00378-9
26.
Urits I, Swanson D, Swett MC, Patel A, Berardino K, Amgalan A, Berger AA, Kassem H, Kaye AD, Viswanath O. A review of patisiran (ONPATTRO®) for the treatment of polyneuropathy in people with hereditary transthyretin amyloidosis. Neurol Ther. 2020;9:301–315. doi: 10.1007/s40120-020-00208-1
27.
Domínguez F, Lalaguna L, López-Olañeta M, Villalba-Orero M, Padrón-Barthe L, Román M, Bello-Arroyo E, Briceño A, Gonzalez-Lopez E, Segovia-Cubero J, et al. Early preventive treatment with enalapril improves cardiac function and delays mortality in mice with arrhythmogenic right ventricular cardiomyopathy type 5. Circ Heart Fail. 2021;14:e007616. doi: 10.1161/CIRCHEARTFAILURE.120.007616
28.
Klinke N, Meyer H, Ratnavadivel S, Reinhardt M, Heinisch JJ, Malmendal A, Milting H, Paululat A. A Drosophila melanogaster model for TMEM43-related arrhythmogenic right ventricular cardiomyopathy type 5. Cell Mol Life Sci. 2022;79:444. doi: 10.1007/s00018-022-04458-0
29.
Rouhi L, Cheedipudi SM, Chen SN, Fan S, Lombardi R, Chen X, Coarfa C, Robertson MJ, Gurha P, Marian AJ. Haploinsufficiency of Tmem43 in cardiac myocytes activates the DNA damage response pathway leading to a late-onset senescence-associated pro-fibrotic cardiomyopathy. Cardiovasc Res. 2021;117:2377–2394. doi: 10.1093/cvr/cvaa300
30.
Gu Q, Xu F, Orgil BO, Khuchua Z, Munkhsaikhan U, Johnson JN, Alberson NR, Pierre JF, Black DD, Dong D, et al. Systems genetics analysis defines importance of TMEM43/ LUMA for cardiac- and metabolic-related pathways. Physiol Genomics. 2022;54:22–35. doi: 10.1152/physiolgenomics.00066.2021
31.
Orgil BO, Munkhsaikhan U, Pierre JF, Li N, Xu F, Alberson NR, Johnson JN, Wetzel GT, Boukens BJD, Lu L, et al. The TMEM43 S358L mutation affects cardiac, small intestine, and metabolic homeostasis in a knock-in mouse model. Am J Physiol Heart Circ Physiol. 2023;324:H866–H880. doi: 10.1152/ajpheart.00712.2022
32.
Brelidze TI. Heteromeric wild-type/mutant potassium channel subunit composition as a major determinant of channelopathy phenotype in heterozygous patients. J Gen Physiol. 2023;155:e202313333. doi: 10.1085/jgp.202313333
33.
Liang WC, Mitsuhashi H, Keduka E, Nonaka I, Noguchi S, Nishino I, Hayashi YK. TMEM43 mutations in emery-dreifuss muscular dystrophy-related myopathy. Ann Neurol. 2011;69:1005–1013. doi: 10.1002/ana.22338
34.
Jang MW, Oh DY, Yi E, Liu X, Ling J, Kim N, Sharma K, Kim TY, Lee S, Kim AR, et al. A nonsense TMEM43 variant leads to disruption of connexin-linked function and autosomal dominant auditory neuropathy spectrum disorder. Proc Natl Acad Sci USA. 2021;118:e2019681118. doi: 10.1073/pnas.2019681118
35.
Chen Z, Cao Z, Gui F, Zhang M, Wu X, Peng H, Yu B, Li W, Ai F, Zhang J. TMEM43 protects against sepsis-induced cardiac injury via inhibiting ferroptosis in mice. Cells. 2022;11:2992. doi: 10.3390/cells11192992
36.
Gu Y, Yao YR, Ding Y, Zhang XW. Reduced expression of transmembrane protein 43 during cardiac hypertrophy leads to worsening heart failure in mice. Exp Biol Med (Maywood). 2023;248:1437–1445. doi: 10.1177/15353702231191111
Information & Authors
Information
Published In
Copyright
© 2025 The Authors.Circulation Research is published on behalf of the American Heart Association, Inc., by Wolters Kluwer Health, Inc. This is an open access article under the terms of the Creative Commons Attribution Non-Commercial-NoDerivs License, which permits use, distribution, and reproduction in any medium, provided that the original work is properly cited, the use is noncommercial, and no modifications or adaptations are made.
Versions
You are viewing the most recent version of this article.
History
Received: 11 November 2024
Revision received: 18 February 2025
Accepted: 2 March 2025
Published online: 17 March 2025
Published in print: 11 April 2025
Keywords
Subjects
Authors
Disclosures
The authors would like to acknowledge that this research has led to the filing of a pending patent application. Inventors: B.J. Byrne, M. Corti, E. Lara-Pezzi, L. Lalaguna, and P. García-Pavía. Application number: EP25382006.2. Title: Method for improving cardiac function in arrhythmogenic right ventricular cardiomyopathy type 5. Priority date: 08/01/2025. Applicants: Centro Nacional de Investigaciones Cardiovasculares Carlos III (F.S.P.), Fundación para la Investigación Biomédica del Hospital Universitario Puerta de Hierro Majadahonda, University of Florida Research Foundation, Incorporated.
Funding Information
Ministerio de Ciencia e Innovación (MCIN) 501100004837: PID2021-124629OB-I00
EC | NextGenerationEU (NGEU) 100031478: TED2021-129774B-C22
EC | European Regional Development Fund (ERDF) 501100008530: PLEC2022-009235
EC | Horizon Europe | å‰µæ–°çš„ææ´² | HORIZON EUROPE European Innovation Council (EIC) 100018703: 101115416
Ministerio de Ciencia e Innovación (MCIN) 501100004837: CEX2020-001041-S
This study was funded by the Pathfinder Cardiogenomics program of the European Innovation Council of the European Union (DCM-NEXT project; project number: 101115416) and grants PID2021-124629OB-I00, TED2021-129774B-C22, and PLEC2022-009235 funded by the Ministry of Science and Innovation (MCIN/AEI/10.13039/501100011033), the European Union’s NextGenerationEU/PRTR (Plan de Recuperación, Transformación y Resiliencia de España), and Fondo Europeo de Desarrollo Regional (FEDER) to E. Lara-Pezzi. The Centro Nacional de Investigaciones Cardiovasculares (CNIC) is supported by the Instituto de Salud Carlos III (ISCIII), the Ministerio de Ciencia, Innovación y Universidades (MICIU) and the Pro CNIC Foundation, and is a Severo Ochoa Center of Excellence (grant CEX2020-001041-S funded by MICIU/AEI/10.13039/501100011033).
Metrics & Citations
Metrics
Citations
Download Citations
If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Select your manager software from the list below and click Download.
- TMEM-ing the Tide: Gene Therapy Holds Promise for ARVC5, Circulation Research, 136, 8, (845-847), (2025)./doi/10.1161/CIRCRESAHA.125.326367
Loading...
View Options
Login options
Check if you have access through your login credentials or your institution to get full access on this article.
Personal login Institutional LoginPurchase Options
Purchase this article to access the full text.
eLetters(0)
eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.
Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.