59 results on '"Meraviglia V"'
Search Results
2. Toward Human Models of Cardiorenal Syndrome in vitro
- Author
-
Gabbin, B., Meraviglia, V., Mummery, C.L., Rabelink, T.J., Meer, B.J. van, Berg, C.W. van den, and Bellin, M.
- Subjects
cardiorenal syndrome ,hiPSCs ,disease modeling ,tissue-specific organoids ,organ-on-chip ,Cardiology and Cardiovascular Medicine - Abstract
Heart and kidney diseases cause high morbidity and mortality. Heart and kidneys have vital functions in the human body and, interestingly, reciprocally influence each other’s behavior: pathological changes in one organ can damage the other. Cardiorenal syndrome (CRS) is a group of disorders in which there is combined dysfunction of both heart and kidney, but its underlying biological mechanisms are not fully understood. This is because complex, multifactorial, and dynamic mechanisms are likely involved. Effective treatments are currently unavailable, but this may be resolved if more was known about how the disease develops and progresses. To date, CRS has actually only been modeled in mice and rats in vivo. Even though these models can capture cardiorenal interaction, they are difficult to manipulate and control. Moreover, interspecies differences may limit extrapolation to patients. The questions we address here are what would it take to model CRS in vitro and how far are we? There are already multiple independent in vitro (human) models of heart and kidney, but none have so far captured their dynamic organ-organ crosstalk. Advanced in vitro human models can provide an insight in disease mechanisms and offer a platform for therapy development. CRS represents an exemplary disease illustrating the need to develop more complex models to study organ-organ interaction in-a-dish. Human induced pluripotent stem cells in combination with microfluidic chips are one powerful tool with potential to recapitulate the characteristics of CRS in vitro. In this review, we provide an overview of the existing in vivo and in vitro models to study CRS, their limitations and new perspectives on how heart-kidney physiological and pathological interaction could be investigated in vitro for future applications.
- Published
- 2022
3. Cardiac mesenchymal stromal cells are a source of adipocytes in arrhythmogenic cardiomyopathy
- Author
-
Sommariva, E., Brambilla, S., Carbucicchio, C., Gambini, E., Meraviglia, V., Dello Russo, A., Farina, F.M., Casella, M., Catto, V., Pontone, G., Chiesa, M., Stadiotti, I., Cogliati, E., Paolin, A., Ouali Alami, N., Preziuso, C., dʼAmati, G., Colombo, G.I., Rossini, A., Capogrossi, M.C., Tondo, C., and Pompilio, G.
- Published
- 2016
- Full Text
- View/download PDF
4. High cardiac differentiation properties are evident in induced pluripotent stem cells obtained from atrial mesenchymal cells
- Author
-
Meraviglia, V., Wen, J., Piacentini, L., Campostrini, G., Wang, C., Florio, M.C., Azzimato, V., Fassina, L., Langes, M., Wong, J., Miragoli, M., Gaetano, C., Pompilio, G., Barbuti, A., DiFrancesco, D., Mascalzoni, D., Pramstaller, P.P., Colombo, G.I., Chen, H.S.V., and Rossini, A.
- Published
- 2015
- Full Text
- View/download PDF
5. Are human induced pluripotent stem cell derived cardiomyocytes a good cellular model for studying incomplete penetrance in arrhythmogenic cardiomyopathy?
- Author
-
Bortoli, M De, Meraviglia, V, Mackova, K, Volani, C, Frommelt, LS, Schlittler, M, Cattelan, G, Konig, E, Rauhe, W, Barbuti, A, Zacchigna, S, Pramstaller, PP, and Rossini, A
- Subjects
- *
ARRHYTHMOGENIC right ventricular dysplasia , *INDUCED pluripotent stem cells , *DELETION mutation , *ADIPOGENESIS , *PLURIPOTENT stem cells , *CARDIOMYOPATHIES , *REGIONAL development , *VENTRICULAR arrhythmia - Abstract
Funding Acknowledgements Type of funding sources: Public grant(s) – EU funding. Main funding source(s): European Regional Development Fund and Interreg V-A Italy-Austria 2014-2020 and Department of Innovation, Research and University of the Autonomous Province of Bolzano-South Tyrol (Italy). Background Human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) were previously used to model several inherited heart diseases (1). Among these, arrhythmogenic cardiomyopathy (ACM) is characterized by a fibrofatty myocardial replacement and severe ventricular arrhythmias (2). ACM is mainly caused by mutations affecting proteins of intercalated discs (3) and shows incomplete penetrance and variable expressivity (4). Purpose To evaluate whether hiPSC-CMs provide a good in vitro model to study incomplete penetrance in ACM. Material and methods hiPSCs were generated from buffy coats of 6 close relatives. Three of them were ACM patients carrying a deletion of exon 4 in the PKP2 gene causing a premature stop codon, 2 were asymptomatic (ASY) PKP2 mutation carriers and 1 was a healthy control (CTR). Whole exome sequencing of the 6 individuals was performed. The purified hiPSC-CMs were cultured in basal and adipogenic medium and examined by ddPCR, western blot, Wes™ immunoassay system, patch clamp and immunofluorescence. Results All family members tested negative for additional mutations in ACM genes. As expected, half the amount of wild type (wt) PKP2 mRNA was found in ACM and ASY than in CTR hiPSC-CMs (2.25±0.27vs4.29±0.70 wtPKP2/HPRT1; p=0.028) (2.08±0.38vs4.29±0.70 wtPKP2/HPRT1; p=0.052). Of note, the mutated (mut) PKP2 mRNA was detected and significantly more expressed in ACM than in ASY hiPSC-CMs (0.33±0.04vs0.11±0.06 mutPKP2/HPRT1; p=0.029). While we confirmed a reduced amount of wt plakophillin-2 protein in ACM and ASY compared to CTR hiPSC-CMs (0.19±0.07vs0.81±0.28 wtPKP2/GAPDH; p=0.016), (0.25±0.07vs0.81±0.28 wtPKP2/GAPDH; p=0.11), the truncated protein was not detected. Moreover, a significantly lower expression of the active form of β-catenin (ABC) has been shown in ACM and ASY than in CTR hiPSC-CMs (0.93±0.18vs1.30±0.09 ABC/GAPDH; p=0.057), (0.76±0.10vs1.30±0.09 ABC/GAPDH; p=0.0057). In basal medium, ACM hiPSC-CMs had a significant lower overall sodium current density compared to CTR (-49.82±2.8 vs -69.38±4.8 pA/pF; p<0.0001) and to ASY (-49.82±2.8 vs -68.92±4.3 pA/pF; p<0.0001) hiPSC-CMs. In adipogenic medium, ACM hiPSC-CMs showed a higher lipid accumulation and a higher sarcomere disorganization than CTR (1580±219vs615±199 intensity/nuclei; p=0.0351) (0.019±0.001vs0.028±0.003 myofibril alignment index; p=0.039) and ASY (1580±219vs689±146 intensity/nuclei; p=0.0059) (0.019±0.001vs0.027±0.001 myofibril alignment index; p=0.010) hiPSC-CMs. Conclusions Here we report that ACM express a significantly higher amount of mutated PKP2 mRNA than ASY iPSC-CMs. Even though no differences were detected for plakophilin-2 and active β-catenin proteins between ACM and ASY iPSCM-CMs, the ACM showed a fatty phenotype and altered electrical activity that differed significantly from the CTR as well as from the ASY hiPSC-CMs, demonstrating that these cells provide a valid model to study incomplete penetrance in ACM. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
6. Evidence of mitochondrial alterations in primary cardiac stromal cells from arrhythmogenic cardiomyopathy hearts.
- Author
-
Philippe, R, Volani, C, Medici, A, Texler, B, Pagliaro, A, Stadiotti, I, Meraviglia, V, Bortoli, M De, Guarino, A, Blumer, M, Pompilio, G, Pramstaller, PP, Sommariva, E, Troppmair, J, and Rossini, A
- Subjects
HEART cells ,STROMAL cells ,WESTERN immunoblotting ,MITOCHONDRIA ,FATTY acid oxidation - Abstract
Funding Acknowledgements Type of funding sources: Public grant(s) – National budget only. Main funding source(s): This research was funded by the Department of Innovation, Research and University of the Autonomous Province of Bolzano-South Tyrol (Italy), and by the Joint Project Südtirol- FWF (Italy-Austria) for A.R. J.T. A.M. R.P.. Background Arrhythmogenic cardiomyopathy (ACM) is a genetic disease associated with sudden cardiac death and fibro-fatty replacement of myocardium. Recently, it has been demonstrated that oxidized lipid contributes to cardiac adipogenesis and that ACM hearts are characterized by an increase in oxidative stress. Purpose As mitochondria are an important source of reactive oxygen species (ROS) within mammalian cells, the present work aims to evaluate if increased oxidative stress observed in ACM hearts is associated with altered mitochondrial function. Methods The oxidative stress marker 4HNE was investigated together with the cardiomyocyte marker cardiac Troponin T on paraffin embedded human ventricular samples and analyzed by confocal microscopy. Human primary cardiac stromal cells (CStCs), obtained from either right ventricle biopsies of ACM patients or healthy cadaveric tissue donor (CTR), were used as cellular model as they are known for their contribution to adipogenesis in the ACM pathology. CStCs were cultured either in basal medium or adipogenic medium (ADIPO) in presence or absence of 500 nM of the ROS scavenger MitoTEMPO. After 7 days of adipogenic differentiation, intracellular lipid droplets accumulation and mitochondrial superoxide levels were measured in CStCs by confocal microscopy using BODIPY 493/503 (0.5 µM) and MitoSOX Red (5 µM) dyes, respectively. The oxidative phosphorylation (OXPHOS) and fatty acid oxidation (FAO) were also evaluated in CStCs by high resolution respirometry. Results 4HNE staining was increased in heart tissues from ACM patients compared to CTRs and particularly evident in non-cardiomyocyte cells. In agreement, a higher MitoSOX fluorescence intensity was found in ACM-CStCs compared to CTR-CStCs in ADIPO medium, confirming higher ROS presence in patient cells. The treatment with MitoTEMPO was efficient in reducing ROS abundance, strongly suggesting mitochondrial origin. Lipid accumulation in ACM-CStCs was also prevented by MitoTEMPO treatment. Preliminary evidence further indicates that mitochondrial respiratory capacity is increased in ACM-CStCs versus CTR-CStCs exposed to adipogenic medium for 7 days. However, transmission electron microscopy analysis and Western blot analysis of Mfn1/2, Opa1, Fis1 and Drp1 did not show an evident alteration neither in mitochondria ultrastructure nor in the expression of proteins regulating mitochondrial dynamics. Conclusions Our data support an alteration of mitochondrial activity in CStCs from ACM patients, apparently not linked to a modified network or morphology of mitochondria but associated with a higher ROS production. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
7. Role of water in chromosome spreading and swelling induced by acetic acid treatment: a FTIR spectroscopy study.
- Author
-
Ami, D., Di Segni, M., Forcella, M., Meraviglia, V., Baccarin, M., Doglia, S. M., and Terzoli, G.
- Published
- 2014
- Full Text
- View/download PDF
8. Expression and function of the atypical purinergic receptor GPR17 in endocrine pancreas.
- Author
-
Di Cairano, E. S., Meraviglia, V., Ulivi, A., Rosa2, P., Moretti, S., Daniele, F., Bertuzzi, F., LaRosa, S., Folli, F., Sacchi, V., and Perego, C.
- Subjects
- *
GENE expression , *PURINERGIC receptors - Abstract
An abstract of the article "Expression and function of the atypical purinergic receptor GPR17 in endocrine pancrease" by E.S. Di Cairano, V. Meraviglia, A. Ulivi and company is presented.
- Published
- 2014
9. GCN5 contributes to intracellular lipid accumulation in human primary cardiac stromal cells from patients affected by Arrhythmogenic cardiomyopathy
- Author
-
Chiara Volani, Alessandra Pagliaro, Johannes Rainer, Giuseppe Paglia, Benedetta Porro, Ilaria Stadiotti, Luisa Foco, Elisa Cogliati, Adolfo Paolin, Costanza Lagrasta, Caterina Frati, Emilia Corradini, Angela Falco, Theresa Matzinger, Anne Picard, Benedetta Ermon, Silvano Piazza, Marzia De Bortoli, Claudio Tondo, Réginald Philippe, Andrea Medici, Alexandros A. Lavdas, Michael J.F. Blumer, Giulio Pompilio, Elena Sommariva, Peter P. Pramstaller, Jakob Troppmair, Viviana Meraviglia, Alessandra Rossini, Volani, C, Pagliaro, A, Rainer, J, Paglia, G, Porro, B, Stadiotti, I, Foco, L, Cogliati, E, Paolin, A, Lagrasta, C, Frati, C, Corradini, E, Falco, A, Matzinger, T, Picard, A, Ermon, B, Piazza, S, De Bortoli, M, Tondo, C, Philippe, R, Medici, A, Lavdas, A, Blumer, M, Pompilio, G, Sommariva, E, Pramstaller, P, Troppmair, J, Meraviglia, V, and Rossini, A
- Subjects
reactive oxygen specie ,Adipogenesis ,Arrhythmogenic cardiomyopathy ,Cell Biology ,human cardiac stromal cell ,Lipids ,Death, Sudden, Cardiac ,cellular redox mechanism ,Humans ,Molecular Medicine ,Stromal Cells ,intracellular lipid accumulation ,histone acetyltransferase GCN5 ,Arrhythmogenic Right Ventricular Dysplasia - Abstract
Arrhythmogenic cardiomyopathy (ACM) is a genetic disease associated with sudden cardiac death and cardiac fibro-fatty replacement. Over the last years, several works have demonstrated that different epigenetic enzymes can affect not only gene expression changes in cardiac diseases but also cellular metabolism. Specifically, the histone acetyltransferase GCN5 is known to facilitate adipogenesis and modulate cardiac metabolism in heart failure. Our group previously demonstrated that human primary cardiac stromal cells (CStCs) contribute to adipogenesis in the ACM pathology. Thus, this study aims to evaluate the role of GCN5 in ACM intracellular lipid accumulation. To do so, CStCs were obtained from right ventricle biopsies of ACM patients and from samples of healthy cadaveric donors (CTR). GCN5 expression was increased both in ex vivo and in vitro ACM samples compared to CTR. When GCN5 expression was silenced or pharmacologically inhibited by the administration of MB-3, we observed a reduction in lipid accumulation and a mitigation of reactive oxygen species (ROS) production in ACM CStCs. In agreement, transcriptome analysis revealed that the presence of MB-3modified the expression of pathways related to cellular redox balance. Altogether, our findings suggest that GCN5 inhibition reduces fat accumulation in ACM CStCs, partially by modulating intracellular redox balance pathways.
- Published
- 2022
10. Metabolic signature of arrhythmogenic cardiomyopathy
- Author
-
Alessandra Rossini, Giulio Pompilio, Chiara Volani, Vinicius Veri Hernandes, Elena Sommariva, Johannes Rainer, Giuseppe Paglia, Michela Casella, Viviana Meraviglia, Peter P. Pramstaller, Sigurður Vidir Smárason, Volani, C, Rainer, J, Hernandes, V, Meraviglia, V, Pramstaller, P, Smarason, S, Pompilio, G, Casella, M, Sommariva, E, Paglia, G, and Rossini, A
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Nitric oxide (NO) ,Arginine ,Endothelium ,Endocrinology, Diabetes and Metabolism ,lcsh:QR1-502 ,Cardiomyopathy ,Metabolomic ,030204 cardiovascular system & hematology ,Biochemistry ,Article ,lcsh:Microbiology ,Sudden cardiac death ,03 medical and health sciences ,0302 clinical medicine ,Metabolomics ,Internal medicine ,biocrates ,medicine ,Metabolome ,Biocrate ,Molecular Biology ,Beta oxidation ,business.industry ,ACM ,medicine.disease ,metabolomics ,Penetrance ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Asymmetric dimethylarginine (ADMA) ,business - Abstract
Arrhythmogenic cardiomyopathy (ACM) is a genetic-based cardiac disease accompanied by severe ventricular arrhythmias and a progressive substitution of the myocardium with fibro-fatty tissue. ACM is often associated with sudden cardiac death. Due to the reduced penetrance and variable expressivity, the presence of a genetic defect is not conclusive, thus complicating the diagnosis of ACM. Recent studies on human induced pluripotent stem cells-derived cardiomyocytes (hiPSC-CMs) obtained from ACM individuals showed a dysregulated metabolic status, leading to the hypothesis that ACM pathology is characterized by an impairment in the energy metabolism. However, despite efforts having been made for the identification of ACM specific biomarkers, there is still a substantial lack of information regarding the whole metabolomic profile of ACM patients. The aim of the present study was to investigate the metabolic profiles of ACM patients compared to healthy controls (CTRLs). The targeted Biocrates AbsoluteIDQ® p180 assay was used on plasma samples. Our analysis showed that ACM patients have a different metabolome compared to CTRLs, and that the pathways mainly affected include tryptophan metabolism, arginine and proline metabolism and beta oxidation of fatty acids. Altogether, our data indicated that the plasma metabolomes of arrhythmogenic cardiomyopathy patients show signs of endothelium damage and impaired nitric oxide (NO), fat, and energy metabolism.
- Published
- 2021
11. Acetylation mediates Cx43 reduction caused by electrical stimulation
- Author
-
Laura Gennaccaro, Anna Binda, Giulio Pompilio, Claudia Colussi, Maurizio C. Capogrossi, Michele Miragoli, Alice Panariti, Peter P. Pramstaller, Andrea Barbuti, Silvia Suffredini, Valerio Azzimato, Alessandra Rossini, Fabio A. Recchia, Maria Cristina Florio, Paul D. Lampe, Khaled Qanud, Viviana Meraviglia, Ilaria Rivolta, Carlo Gaetano, Meraviglia, V, Azzimato, V, Colussi, C, Florio, M, Binda, A, Panariti, A, Qanud, K, Suffredini, S, Gennaccaro, L, Miragoli, M, Barbuti, A, Lampe, P, Gaetano, C, Pramstaller, P, Capogrossi, M, Recchia, F, Pompilio, G, Rivolta, I, and Rossini, A
- Subjects
Cell signaling ,Heart Ventricles ,Gap junction ,Electrical field stimulation ,Gap junctions, Acetylation, Connexin43 ,Stimulation ,Histone Deacetylase 1 ,Cell Communication ,Biology ,Article ,Acetylation ,Connexin43 ,Electrical field stimulation ,Gap junctions ,chemistry.chemical_compound ,Mice ,Dogs ,MG132 ,Animals ,Humans ,Myocytes, Cardiac ,RNA, Messenger ,Molecular Biology ,Histone Acetyltransferases ,Molecular biology ,HDAC1 ,Electric Stimulation ,Anacardic acids ,Cell biology ,Anacardic Acids ,chemistry ,Connexin 43 ,cardiovascular system ,Histone deacetylase ,sense organs ,Cardiology and Cardiovascular Medicine - Abstract
Communication between cardiomyocytes depends upon gap junctions (GJ). Previous studies have demonstrated that electrical stimulation induces GJ remodeling and modifies histone acetylase (HAT) and deacetylase (HDAC) activities, although these two results have not been linked. The aim of this work was to establish whether electrical stimulation modulates GJ-mediated cardiac cell-cell communication by acetylation-dependent mechanisms. Field stimulation of HL-1 cardiomyocytes at 0.5. Hz for 24. h significantly reduced connexin43 (Cx43) expression and cell-cell communication. HDAC activity was down-regulated whereas HAT activity was not modified resulting in increased acetylation of Cx43. Consistent with a post-translational mechanism, we did not observe a reduction in Cx43 mRNA in electrically stimulated cells, while the proteasomal inhibitor MG132 maintained Cx43 expression. Further, the treatment of paced cells with the HAT inhibitor Anacardic Acid maintained both the levels of Cx43 and cell-cell communication. Finally, we observed increased acetylation of Cx43 in the left ventricles of dogs subjected to chronic tachypacing as a model of abnormal ventricular activation.In conclusion, our findings suggest that altered electrical activity can regulate cardiomyocyte communication by influencing the acetylation status of Cx43.
- Published
- 2015
12. Human mesenchymal stromal cells: how the tissue of origin influences plastic properties and microRNA expression profile
- Author
-
MERAVIGLIA, VIVIANA and Meraviglia, V
- Subjects
microRNA ,Mesenchymal stromal cell ,differentiation ,BIO/11 - BIOLOGIA MOLECOLARE - Abstract
MicroRNAs are key modulators at molecular level in different biological processes, including determination of cell fate and differentiation. Herein, microRNA expression profiling experiments were performed on syngeneic cardiac and bone marrow mesenchymal stromal cells cultured in standard growth medium and then in vitro exposed to adipogenic, osteogenic, cardiomyogenic and endothelial differentiation media. Analysis identified a tissue-specific microRNA signature composed by 16 microRNAs that univocally discriminated cell type of origin and that were completely unaffected by in vitro differentiation media: 4 microRNAs were over-expressed in cardiac stromal cells, and 12 were overexpressed or present only in bone marrow stromal cells. Further, results revealed microRNA subsets specifically modulated by each differentiation medium, independently from the cell type of origin, and a subset of 7 microRNAs that were downregulated by all media with respect to growth medium. Finally, we identified 16 microRNAs that were differentially modulated by the media when comparing the two tissues of origin. The existence of a tissue-specific microRNA signature surviving to any differentiation stimuli, strongly support the role if microRNAs determining cell identity related to tissue origin. Moreover, we identified microRNA subsets modulated by different culture conditions irrespective of tissue origin, pointing out their importance during differentiation processes.
- Published
- 2014
13. Role of water in chromosome spreading and swelling induced by acetic acid treatment: a FTIR spectroscopy study
- Author
-
Silvia Maria Doglia, G. Terzoli, M. Di Segni, Matilde Forcella, Diletta Ami, Marco Baccarin, Viviana Meraviglia, Ami, D, Di Segni, M, Forcella, M, Meraviglia, V, Baccarin, M, Doglia, S, and Terzoli, G
- Subjects
Male ,spectroscopy ,Histology ,Biophysics ,DNA hydration ,FIS/07 - FISICA APPLICATA (A BENI CULTURALI, AMBIENTALI, BIOLOGIA E MEDICINA) ,Biology ,Acetic acid ,chemistry.chemical_compound ,chromosome spreading and swelling ,DNA conformational transition ,Spectroscopy, Fourier Transform Infrared ,medicine ,Bound water ,Chromosomes, Human ,Humans ,chromosome ,Fourier transform infrared spectroscopy ,lcsh:QH301-705.5 ,Chromosome preparation ,Acetic Acid ,Original Paper ,FTIR microspectroscopy ,Chromosome ,Water ,Cell Biology ,DNA ,BIO/10 - BIOCHIMICA ,Chromatin ,Cytosol ,Biochemistry ,chemistry ,lcsh:Biology (General) ,Chromosome preparation, chromosome spreading and swelling, DNA conformational transition, FTIR (micro)spectroscopy, protein and DNA hydration ,FTIR (micro)spectroscopy ,Female ,Swelling ,medicine.symptom ,protein - Abstract
The so called chromosome preparation is a procedure consisting of three strictly connected stages that enables to obtain chromosomes of quality suitable for cytogenetic analysis. Interestingly, experimental evidence strongly suggested that chromosome spreading and swelling (key processes that allow their counting and detailed structural analysis) are induced in the last fixative-evaporation stage by the interaction, mediated by acetic acid, between water from the environmental humidity, and the cytoplasmic matrix and the chro-matin. However, since a considerable variation in the quality of chromosome preparations is observed, strongly depending on the environmental conditions in which the procedure takes place, a better comprehension of the mechanisms underlying chromosome preparation is required. To this aim, here we analysed intact lymphocytes before and at each stage of the chromosome preparation protocol by Fourier transform infrared (FTIR) spec-troscopy, a technique widely used for the study not only of isolated biomolecules, but also of complex biological systems, such as whole cells. Interestingly, we found that the chromosome preparation protocol induces significant structural changes of cell proteins and DNA, in particular due to the interaction with acetic acid. Moreover, noteworthy, through the monitoring of changes in the water combination band between 2300 and 1800 cm-1, we provided evidence at molecular level of the crucial role of the bound water to the cytoplasmic matrix and to the chromatin in determining the chromosome spreading and swelling. Our FTIR results, therefore, underline the need to perform the last fixative-evaporation stage in standardized and optimized temperature and relative humidity conditions, thus providing chromosomes of high quality for the cytogenet-ic analysis that would lead in this way to more reliable results. © D. Ami et al., 2014.
- Published
- 2014
14. Editorial: Advances in pluripotent stem cell-based in vitro models of the human heart for cardiac physiology, disease modeling and clinical applications.
- Author
-
Prondzynski M, Pioner JM, Sala L, Bellin M, and Meraviglia V
- Abstract
Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
- Published
- 2024
- Full Text
- View/download PDF
15. Vascularized hiPSC-derived 3D cardiac microtissue on chip.
- Author
-
Arslan U, Brescia M, Meraviglia V, Nahon DM, van Helden RWJ, Stein JM, van den Hil FE, van Meer BJ, Vila Cuenca M, Mummery CL, and Orlova VV
- Published
- 2023
- Full Text
- View/download PDF
16. Patient-specific primary and pluripotent stem cell-derived stromal cells recapitulate key aspects of arrhythmogenic cardiomyopathy.
- Author
-
Maione AS, Meraviglia V, Iengo L, Rabino M, Chiesa M, Catto V, Tondo C, Pompilio G, Bellin M, and Sommariva E
- Subjects
- Humans, Stromal Cells, Induced Pluripotent Stem Cells, Pluripotent Stem Cells, Mesenchymal Stem Cells, Cardiomyopathies
- Abstract
Primary cardiac mesenchymal stromal cells (C-MSCs) can promote the aberrant remodeling of cardiac tissue that characterizes arrhythmogenic cardiomyopathy (ACM) by differentiating into adipocytes and myofibroblasts. These cells' limitations, including restricted access to primary material and its manipulation have been overcome by the advancement of human induced pluripotent stem cells (hiPSCs), and their ability to differentiate towards the cardiac stromal population. C-MSCs derived from hiPSCs make it possible to work with virtually unlimited numbers of cells that are genetically identical to the cells of origin. We performed in vitro experiments on primary stromal cells (Primary) and hiPSC-derived stromal cells (hiPSC-D) to compare them as tools to model ACM. Both Primary and hiPSC-D cells expressed mesenchymal surface markers and possessed typical MSC differentiation potentials. hiPSC-D expressed desmosomal genes and proteins and shared a similar transcriptomic profile with Primary cells. Furthermore, ACM hiPSC-D exhibited higher propensity to accumulate lipid droplets and collagen compared to healthy control cells, similar to their primary counterparts. Therefore, both Primary and hiPSC-D cardiac stromal cells obtained from ACM patients can be used to model aspects of the disease. The choice of the most suitable model will depend on experimental needs and on the availability of human source samples., (© 2023. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
17. Heart and kidney organoids maintain organ-specific function in a microfluidic system.
- Author
-
Gabbin B, Meraviglia V, Angenent ML, Ward-van Oostwaard D, Sol W, Mummery CL, Rabelink TJ, van Meer BJ, van den Berg CW, and Bellin M
- Abstract
Heart and kidney communicate with one another in an interdependent relationship and they influence each other's behavior reciprocally, as pathological changes in one organ can damage the other. Although independent human in vitro models for heart and kidney exist, they do not capture their dynamic crosstalk. We have developed a microfluidic system which can be used to study heart and kidney interaction in vitro . Cardiac microtissues (cMTs) and kidney organoids (kOs) derived from human induced pluripotent stem cells (hiPSCs) were generated and loaded into two separated communicating chambers of a perfusion chip. Static culture conditions were compared with dynamic culture under unidirectional flow. Tissue viability was maintained for minimally 72 h under both conditions, as indicated by the presence of sarcomeric structures coupled with beating activity in cMTs and the presence of nephron structures and albumin uptake in kOs. We concluded that this system enables the study of human cardiac and kidney organoid interaction in vitro while controlling parameters like fluidic flow speed and direction. Together, this "cardiorenal-unit" provides a new in vitro model to study the cardiorenal axis and it may be further developed to investigate diseases involving both two organs and their potential treatments., Competing Interests: C.L.M. is co-founder of Pluriomics (now Ncardia) and B.v.M. is co-founder and CTO of Demcon Biovitronix. The other authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (© 2023 Published by Elsevier Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
18. Role of microRNAs in arrhythmogenic cardiomyopathy: translation as biomarkers into clinical practice.
- Author
-
Alcalde M, Toro R, Bonet F, Córdoba-Caballero J, Martínez-Barrios E, Ranea JA, Vallverdú-Prats M, Brugada R, Meraviglia V, Bellin M, Sarquella-Brugada G, and Campuzano O
- Subjects
- Humans, Genetic Predisposition to Disease, Biomarkers, Death, Sudden, Cardiac etiology, MicroRNAs genetics, Arrhythmogenic Right Ventricular Dysplasia genetics, Arrhythmogenic Right Ventricular Dysplasia metabolism, Arrhythmogenic Right Ventricular Dysplasia pathology
- Abstract
Arrhythmogenic cardiomyopathy is a rare inherited entity, characterized by a progressive fibro-fatty replacement of the myocardium. It leads to malignant arrhythmias and a high risk of sudden cardiac death. Incomplete penetrance and variable expressivity are hallmarks of this arrhythmogenic cardiac disease, where the first manifestation may be syncope and sudden cardiac death, often triggered by physical exercise. Early identification of individuals at risk is crucial to adopt protective and ideally personalized measures to prevent lethal episodes. The genetic analysis identifies deleterious rare variants in nearly 70% of cases, mostly in genes encoding proteins of the desmosome. However, other factors may modulate the phenotype onset and outcome of disease, such as microRNAs. These small noncoding RNAs play a key role in gene expression regulation and the network of cellular processes. In recent years, data focused on the role of microRNAs as potential biomarkers in arrhythmogenic cardiomyopathy have progressively increased. A better understanding of the functions and interactions of microRNAs will likely have clinical implications. Herein, we propose an exhaustive review of the literature regarding these noncoding RNAs, their versatile mechanisms of gene regulation and present novel targets in arrhythmogenic cardiomyopathy., (Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
19. RagD auto-activating mutations impair MiT/TFE activity in kidney tubulopathy and cardiomyopathy syndrome.
- Author
-
Sambri I, Ferniani M, Campostrini G, Testa M, Meraviglia V, de Araujo MEG, Dokládal L, Vilardo C, Monfregola J, Zampelli N, Vecchio Blanco FD, Torella A, Ruosi C, Fecarotta S, Parenti G, Staiano L, Bellin M, Huber LA, De Virgilio C, Trepiccione F, Nigro V, and Ballabio A
- Subjects
- Humans, Autophagy genetics, HeLa Cells, Kidney metabolism, Lysosomes metabolism, Mechanistic Target of Rapamycin Complex 1 metabolism, Mutation, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors genetics, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors metabolism, Induced Pluripotent Stem Cells metabolism
- Abstract
Heterozygous mutations in the gene encoding RagD GTPase were shown to cause a novel autosomal dominant condition characterized by kidney tubulopathy and cardiomyopathy. We previously demonstrated that RagD, and its paralogue RagC, mediate a non-canonical mTORC1 signaling pathway that inhibits the activity of TFEB and TFE3, transcription factors of the MiT/TFE family and master regulators of lysosomal biogenesis and autophagy. Here we show that RagD mutations causing kidney tubulopathy and cardiomyopathy are "auto- activating", even in the absence of Folliculin, the GAP responsible for RagC/D activation, and cause constitutive phosphorylation of TFEB and TFE3 by mTORC1, without affecting the phosphorylation of "canonical" mTORC1 substrates, such as S6K. By using HeLa and HK-2 cell lines, human induced pluripotent stem cell-derived cardiomyocytes and patient-derived primary fibroblasts, we show that RRAGD auto-activating mutations lead to inhibition of TFEB and TFE3 nuclear translocation and transcriptional activity, which impairs the response to lysosomal and mitochondrial injury. These data suggest that inhibition of MiT/TFE factors plays a key role in kidney tubulopathy and cardiomyopathy syndrome., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
20. Integrated Proteomics Unveils Nuclear PDE3A2 as a Regulator of Cardiac Myocyte Hypertrophy.
- Author
-
Subramaniam G, Schleicher K, Kovanich D, Zerio A, Folkmanaite M, Chao YC, Surdo NC, Koschinski A, Hu J, Scholten A, Heck AJR, Ercu M, Sholokh A, Park KC, Klussmann E, Meraviglia V, Bellin M, Zanivan S, Hester S, Mohammed S, and Zaccolo M
- Subjects
- Humans, Proteomics, Phosphoric Diester Hydrolases, Hypertrophy, Adrenergic Agents, Myocytes, Cardiac, Cyclic AMP
- Abstract
Background: Signaling by cAMP is organized in multiple distinct subcellular nanodomains regulated by cAMP-hydrolyzing PDEs (phosphodiesterases). Cardiac β-adrenergic signaling has served as the prototypical system to elucidate cAMP compartmentalization. Although studies in cardiac myocytes have provided an understanding of the location and properties of a handful of cAMP subcellular compartments, an overall view of the cellular landscape of cAMP nanodomains is missing., Methods: Here, we combined an integrated phosphoproteomics approach that takes advantage of the unique role that individual PDEs play in the control of local cAMP, with network analysis to identify previously unrecognized cAMP nanodomains associated with β-adrenergic stimulation. We then validated the composition and function of one of these nanodomains using biochemical, pharmacological, and genetic approaches and cardiac myocytes from both rodents and humans., Results: We demonstrate the validity of the integrated phosphoproteomic strategy to pinpoint the location and provide critical cues to determine the function of previously unknown cAMP nanodomains. We characterize in detail one such compartment and demonstrate that the PDE3A2 isoform operates in a nuclear nanodomain that involves SMAD4 (SMAD family member 4) and HDAC-1 (histone deacetylase 1). Inhibition of PDE3 results in increased HDAC-1 phosphorylation, leading to inhibition of its deacetylase activity, derepression of gene transcription, and cardiac myocyte hypertrophic growth., Conclusions: We developed a strategy for detailed mapping of subcellular PDE-specific cAMP nanodomains. Our findings reveal a mechanism that explains the negative long-term clinical outcome observed in patients with heart failure treated with PDE3 inhibitors.
- Published
- 2023
- Full Text
- View/download PDF
21. Desmosomal protein degradation as an underlying cause of arrhythmogenic cardiomyopathy.
- Author
-
Tsui H, van Kampen SJ, Han SJ, Meraviglia V, van Ham WB, Casini S, van der Kraak P, Vink A, Yin X, Mayr M, Bossu A, Marchal GA, Monshouwer-Kloots J, Eding J, Versteeg D, de Ruiter H, Bezstarosti K, Groeneweg J, Klaasen SJ, van Laake LW, Demmers JAA, Kops GJPL, Mummery CL, van Veen TAB, Remme CA, Bellin M, and van Rooij E
- Subjects
- Humans, Mice, Animals, Infant, Proteolysis, Myocytes, Cardiac metabolism, Mutation genetics, Plakophilins genetics, Plakophilins metabolism, Cardiomyopathies genetics
- Abstract
Arrhythmogenic cardiomyopathy (ACM) is an inherited progressive cardiac disease. Many patients with ACM harbor mutations in desmosomal genes, predominantly in plakophilin-2 ( PKP2 ). Although the genetic basis of ACM is well characterized, the underlying disease-driving mechanisms remain unresolved. Explanted hearts from patients with ACM had less PKP2 compared with healthy hearts, which correlated with reduced expression of desmosomal and adherens junction (AJ) proteins. These proteins were also disorganized in areas of fibrotic remodeling. In vitro data from human-induced pluripotent stem cell-derived cardiomyocytes and microtissues carrying the heterozygous PKP2 c.2013delC pathogenic mutation also displayed impaired contractility. Knockin mice carrying the equivalent heterozygous Pkp2 c.1755delA mutation recapitulated changes in desmosomal and AJ proteins and displayed cardiac dysfunction and fibrosis with age. Global proteomics analysis of 4-month-old heterozygous Pkp2 c.1755delA hearts indicated involvement of the ubiquitin-proteasome system (UPS) in ACM pathogenesis. Inhibition of the UPS in mutant mice increased area composita proteins and improved calcium dynamics in isolated cardiomyocytes. Additional proteomics analyses identified lysine ubiquitination sites on the desmosomal proteins, which were more ubiquitinated in mutant mice. In summary, we show that a plakophilin-2 mutation can lead to decreased desmosomal and AJ protein expression through a UPS-dependent mechanism, which preceded cardiac remodeling. These findings suggest that targeting protein degradation and improving desmosomal protein stability may be a potential therapeutic strategy for the treatment of ACM.
- Published
- 2023
- Full Text
- View/download PDF
22. Modeling incomplete penetrance in arrhythmogenic cardiomyopathy by human induced pluripotent stem cell derived cardiomyocytes.
- Author
-
De Bortoli M, Meraviglia V, Mackova K, Frommelt LS, König E, Rainer J, Volani C, Benzoni P, Schlittler M, Cattelan G, Motta BM, Volpato C, Rauhe W, Barbuti A, Zacchigna S, Pramstaller PP, and Rossini A
- Abstract
Human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) are commonly used to model arrhythmogenic cardiomyopathy (ACM), a heritable cardiac disease characterized by severe ventricular arrhythmias, fibrofatty myocardial replacement and progressive ventricular dysfunction. Although ACM is inherited as an autosomal dominant disease, incomplete penetrance and variable expressivity are extremely common, resulting in different clinical manifestations. Here, we propose hiPSC-CMs as a powerful in vitro model to study incomplete penetrance in ACM. Six hiPSC lines were generated from blood samples of three ACM patients carrying a heterozygous deletion of exon 4 in the PKP2 gene, two asymptomatic (ASY) carriers of the same mutation and one healthy control (CTR), all belonging to the same family. Whole exome sequencing was performed in all family members and hiPSC-CMs were examined by ddPCR, western blot, Wes™ immunoassay system, patch clamp, immunofluorescence and RNASeq. Our results show molecular and functional differences between ACM and ASY hiPSC-CMs, including a higher amount of mutated PKP2 mRNA, a lower expression of the connexin-43 protein, a lower overall density of sodium current, a higher intracellular lipid accumulation and sarcomere disorganization in ACM compared to ASY hiPSC-CMs. Differentially expressed genes were also found, supporting a predisposition for a fatty phenotype in ACM hiPSC-CMs. These data indicate that hiPSC-CMs are a suitable model to study incomplete penetrance in ACM., Competing Interests: The authors declare no conflict of interest., (© 2023 The Authors.)
- Published
- 2023
- Full Text
- View/download PDF
23. GCN5 contributes to intracellular lipid accumulation in human primary cardiac stromal cells from patients affected by Arrhythmogenic cardiomyopathy.
- Author
-
Volani C, Pagliaro A, Rainer J, Paglia G, Porro B, Stadiotti I, Foco L, Cogliati E, Paolin A, Lagrasta C, Frati C, Corradini E, Falco A, Matzinger T, Picard A, Ermon B, Piazza S, De Bortoli M, Tondo C, Philippe R, Medici A, Lavdas AA, Blumer MJF, Pompilio G, Sommariva E, Pramstaller PP, Troppmair J, Meraviglia V, and Rossini A
- Subjects
- Adipogenesis physiology, Death, Sudden, Cardiac pathology, Humans, Lipids, Stromal Cells metabolism, Arrhythmogenic Right Ventricular Dysplasia genetics, Arrhythmogenic Right Ventricular Dysplasia metabolism, Arrhythmogenic Right Ventricular Dysplasia pathology
- Abstract
Arrhythmogenic cardiomyopathy (ACM) is a genetic disease associated with sudden cardiac death and cardiac fibro-fatty replacement. Over the last years, several works have demonstrated that different epigenetic enzymes can affect not only gene expression changes in cardiac diseases but also cellular metabolism. Specifically, the histone acetyltransferase GCN5 is known to facilitate adipogenesis and modulate cardiac metabolism in heart failure. Our group previously demonstrated that human primary cardiac stromal cells (CStCs) contribute to adipogenesis in the ACM pathology. Thus, this study aims to evaluate the role of GCN5 in ACM intracellular lipid accumulation. To do so, CStCs were obtained from right ventricle biopsies of ACM patients and from samples of healthy cadaveric donors (CTR). GCN5 expression was increased both in ex vivo and in vitro ACM samples compared to CTR. When GCN5 expression was silenced or pharmacologically inhibited by the administration of MB-3, we observed a reduction in lipid accumulation and a mitigation of reactive oxygen species (ROS) production in ACM CStCs. In agreement, transcriptome analysis revealed that the presence of MB-3 modified the expression of pathways related to cellular redox balance. Altogether, our findings suggest that GCN5 inhibition reduces fat accumulation in ACM CStCs, partially by modulating intracellular redox balance pathways., (© 2022 EURAC Research. Journal of Cellular and Molecular Medicine published by Foundation for Cellular and Molecular Medicine and John Wiley & Sons Ltd.)
- Published
- 2022
- Full Text
- View/download PDF
24. Toward Human Models of Cardiorenal Syndrome in vitro .
- Author
-
Gabbin B, Meraviglia V, Mummery CL, Rabelink TJ, van Meer BJ, van den Berg CW, and Bellin M
- Abstract
Heart and kidney diseases cause high morbidity and mortality. Heart and kidneys have vital functions in the human body and, interestingly, reciprocally influence each other's behavior: pathological changes in one organ can damage the other. Cardiorenal syndrome (CRS) is a group of disorders in which there is combined dysfunction of both heart and kidney, but its underlying biological mechanisms are not fully understood. This is because complex, multifactorial, and dynamic mechanisms are likely involved. Effective treatments are currently unavailable, but this may be resolved if more was known about how the disease develops and progresses. To date, CRS has actually only been modeled in mice and rats in vivo . Even though these models can capture cardiorenal interaction, they are difficult to manipulate and control. Moreover, interspecies differences may limit extrapolation to patients. The questions we address here are what would it take to model CRS in vitro and how far are we? There are already multiple independent in vitro (human) models of heart and kidney, but none have so far captured their dynamic organ-organ crosstalk. Advanced in vitro human models can provide an insight in disease mechanisms and offer a platform for therapy development. CRS represents an exemplary disease illustrating the need to develop more complex models to study organ-organ interaction in-a-dish. Human induced pluripotent stem cells in combination with microfluidic chips are one powerful tool with potential to recapitulate the characteristics of CRS in vitro . In this review, we provide an overview of the existing in vivo and in vitro models to study CRS, their limitations and new perspectives on how heart-kidney physiological and pathological interaction could be investigated in vitro for future applications., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Gabbin, Meraviglia, Mummery, Rabelink, van Meer, van den Berg and Bellin.)
- Published
- 2022
- Full Text
- View/download PDF
25. Inflammation in the Pathogenesis of Arrhythmogenic Cardiomyopathy: Secondary Event or Active Driver?
- Author
-
Meraviglia V, Alcalde M, Campuzano O, and Bellin M
- Abstract
Arrhythmogenic cardiomyopathy (ACM) is a rare inherited cardiac disease characterized by arrhythmia and progressive fibro-fatty replacement of the myocardium, which leads to heart failure and sudden cardiac death. Inflammation contributes to disease progression, and it is characterized by inflammatory cell infiltrates in the damaged myocardium and inflammatory mediators in the blood of ACM patients. However, the molecular basis of inflammatory process in ACM remains under investigated and it is unclear whether inflammation is a primary event leading to arrhythmia and myocardial damage or it is a secondary response triggered by cardiomyocyte death. Here, we provide an overview of the proposed players and triggers involved in inflammation in ACM, focusing on those studied using in vivo and in vitro models. Deepening current knowledge of inflammation-related mechanisms in ACM could help identifying novel therapeutic perspectives, such as anti-inflammatory therapy., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2021 Meraviglia, Alcalde, Campuzano and Bellin.)
- Published
- 2021
- Full Text
- View/download PDF
26. Oxidized LDL-dependent pathway as new pathogenic trigger in arrhythmogenic cardiomyopathy.
- Author
-
Sommariva E, Stadiotti I, Casella M, Catto V, Dello Russo A, Carbucicchio C, Arnaboldi L, De Metrio S, Milano G, Scopece A, Casaburo M, Andreini D, Mushtaq S, Conte E, Chiesa M, Birchmeier W, Cogliati E, Paolin A, König E, Meraviglia V, De Musso M, Volani C, Cattelan G, Rauhe W, Turnu L, Porro B, Pedrazzini M, Camera M, Corsini A, Tondo C, Rossini A, and Pompilio G
- Subjects
- Animals, Arrhythmias, Cardiac etiology, Humans, Lipoproteins, LDL, Mice, Phenotype, Arrhythmogenic Right Ventricular Dysplasia genetics
- Abstract
Arrhythmogenic cardiomyopathy (ACM) is hallmarked by ventricular fibro-adipogenic alterations, contributing to cardiac dysfunctions and arrhythmias. Although genetically determined (e.g., PKP2 mutations), ACM phenotypes are highly variable. More data on phenotype modulators, clinical prognosticators, and etiological therapies are awaited. We hypothesized that oxidized low-density lipoprotein (oxLDL)-dependent activation of PPARγ, a recognized effector of ACM adipogenesis, contributes to disease pathogenesis. ACM patients showing high plasma concentration of oxLDL display severe clinical phenotypes in terms of fat infiltration, ventricular dysfunction, and major arrhythmic event risk. In ACM patient-derived cardiac cells, we demonstrated that oxLDLs are major cofactors of adipogenesis. Mechanistically, the increased lipid accumulation is mediated by oxLDL cell internalization through CD36, ultimately resulting in PPARγ upregulation. By boosting oxLDL in a Pkp2 heterozygous knock-out mice through high-fat diet feeding, we confirmed in vivo the oxidized lipid dependency of cardiac adipogenesis and right ventricle systolic impairment, which are counteracted by atorvastatin treatment. The modulatory role of oxidized lipids on ACM adipogenesis, demonstrated at cellular, mouse, and patient levels, represents a novel risk stratification tool and a target for ACM pharmacological strategies., (© 2021 The Authors. Published under the terms of the CC BY 4.0 license.)
- Published
- 2021
- Full Text
- View/download PDF
27. Generation and characterization of three human induced pluripotent stem cell lines (EURACi007-A, EURACi008-A, EURACi009-A) from three different individuals of the same family with arrhythmogenic cardiomyopathy (ACM) carrying the plakophillin2 p.N346Lfs*12 mutation.
- Author
-
Meraviglia V, Cattelan G, De Bortoli M, Motta BM, Volpato C, Frommelt LS, Rauhe W, Di Segni M, Silipigni R, Pramstaller PP, and Rossini A
- Subjects
- Cell Differentiation, Heterozygote, Humans, Mutation, Cardiomyopathies, Induced Pluripotent Stem Cells
- Abstract
Arrhythmogenic Cardiomyopathy (ACM) is a genetically based cardiomyopathy associated with ventricular arrhythmias and fibro-fatty substitution of cardiac tissue. It is characterized by incomplete penetrance. We generated human iPSCs by episomal reprogramming of blood cells from three members of the same family: the proband, affected by ACM and carrying the heterozygous plakophillin2 p.N346Lfs*12 mutation, one asymptomatic carrier of the same mutation and one apparently healthy control. hiPSCs were characterized according to standard protocols including karyotyping, pluripotency marker expression and differentiation towards the three germ layers. These hiPSC lines can be used to study the mechanisms of ACM incomplete penetrance in vitro., (Copyright © 2021 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
28. Generation of human induced pluripotent stem cell line EURACi006-A and its isogenic gene-corrected line EURACi006-A-1 from an arrhythmogenic cardiomyopathy patient carrying the c.1643delG PKP2 mutation.
- Author
-
Meraviglia V, Ganesh S, Arendzen CH, Freund C, Sommariva E, Rossini A, and Bellin M
- Subjects
- Cell Differentiation, Heterozygote, Humans, Mutation genetics, Plakophilins genetics, Cardiomyopathies, Induced Pluripotent Stem Cells
- Abstract
Arrhythmogenic Cardiomyopathy (ACM) is a rare genetic cardiac disease predominantly associated with mutations in genes of the desmosomes and characterized by arrhythmia and fibro-fatty replacement of the myocardium. We generated human induced pluripotent stem cells (hiPSCs) from one patient affected by ACM carrying the heterozygous c.1643delG (p.G548VfsX15) PKP2 mutation and then corrected the mutation using CRISPR/Cas9 technology. Both original and corrected hiPSC lines showed typical morphology of pluripotent cells, expressed pluripotency markers, displayed a normal karyotype, and differentiated towards the three germ layers. This isogenic hiPSC pair can be used to study the role of the c.1643delG PKP2 mutation in vitro., (Copyright © 2021. Published by Elsevier B.V.)
- Published
- 2021
- Full Text
- View/download PDF
29. Generation, functional analysis and applications of isogenic three-dimensional self-aggregating cardiac microtissues from human pluripotent stem cells.
- Author
-
Campostrini G, Meraviglia V, Giacomelli E, van Helden RWJ, Yiangou L, Davis RP, Bellin M, Orlova VV, and Mummery CL
- Subjects
- Cell Differentiation, Electrophysiological Phenomena, Humans, Models, Biological, Myocytes, Cardiac cytology, Tissue Scaffolds chemistry, Heart physiology, Induced Pluripotent Stem Cells cytology, Tissue Engineering methods
- Abstract
Tissue-like structures from human pluripotent stem cells containing multiple cell types are transforming our ability to model and understand human development and disease. Here we describe a protocol to generate cardiomyocytes (CMs), cardiac fibroblasts (CFs) and cardiac endothelial cells (ECs), the three principal cell types in the heart, from human induced pluripotent stem cells (hiPSCs) and combine them in three-dimensional (3D) cardiac microtissues (MTs). We include details of how to differentiate, isolate, cryopreserve and thaw the component cells and how to construct and analyze the MTs. The protocol supports hiPSC-CM maturation and allows replacement of one or more of the three heart cell types in the MTs with isogenic variants bearing disease mutations. Differentiation of each cell type takes ~30 d, while MT formation and maturation requires another 20 d. No specialist equipment is needed and the method is inexpensive, requiring just 5,000 cells per MT.
- Published
- 2021
- Full Text
- View/download PDF
30. Metabolic Signature of Arrhythmogenic Cardiomyopathy.
- Author
-
Volani C, Rainer J, Hernandes VV, Meraviglia V, Pramstaller PP, Smárason SV, Pompilio G, Casella M, Sommariva E, Paglia G, and Rossini A
- Abstract
Arrhythmogenic cardiomyopathy (ACM) is a genetic-based cardiac disease accompanied by severe ventricular arrhythmias and a progressive substitution of the myocardium with fibro-fatty tissue. ACM is often associated with sudden cardiac death. Due to the reduced penetrance and variable expressivity, the presence of a genetic defect is not conclusive, thus complicating the diagnosis of ACM. Recent studies on human induced pluripotent stem cells-derived cardiomyocytes (hiPSC-CMs) obtained from ACM individuals showed a dysregulated metabolic status, leading to the hypothesis that ACM pathology is characterized by an impairment in the energy metabolism. However, despite efforts having been made for the identification of ACM specific biomarkers, there is still a substantial lack of information regarding the whole metabolomic profile of ACM patients. The aim of the present study was to investigate the metabolic profiles of ACM patients compared to healthy controls (CTRLs). The targeted Biocrates AbsoluteIDQ
® p180 assay was used on plasma samples. Our analysis showed that ACM patients have a different metabolome compared to CTRLs, and that the pathways mainly affected include tryptophan metabolism, arginine and proline metabolism and beta oxidation of fatty acids. Altogether, our data indicated that the plasma metabolomes of arrhythmogenic cardiomyopathy patients show signs of endothelium damage and impaired nitric oxide (NO), fat, and energy metabolism.- Published
- 2021
- Full Text
- View/download PDF
31. Generation of human induced pluripotent stem cell line LUMCi027-A and its isogenic gene-corrected line from a patient affected by arrhythmogenic cardiomyopathy and carrying the c.2013delC PKP2 mutation.
- Author
-
Meraviglia V, Arendzen CH, Tok M, Freund C, Maione AS, Sommariva E, and Bellin M
- Subjects
- Cell Differentiation, Heterozygote, Humans, Mutation, Plakophilins genetics, Cardiomyopathies, Induced Pluripotent Stem Cells
- Abstract
Arrhythmogenic Cardiomyopathy (ACM) is a rare inherited heart muscle disease characterised by progressive fibro-fatty replacement of the ventricular myocardium leading to life-threatening arrhythmias. We generated human induced pluripotent stem cells (hiPSCs) from a patient affected by ACM and carrying the heterozygous c.2013delC (p.K672Rfs) PKP2 mutation and then corrected the mutation using CRISPR/Cas9 technology. Both hiPSC lines expressed pluripotency markers, maintained a normal karyotype, and differentiated into derivatives of the three germ layers. This isogenic hiPSC pair represents a genetically controlled system to study the role of the c.2013delC PKP2 mutation in vitro., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2020 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
32. Human-iPSC-Derived Cardiac Stromal Cells Enhance Maturation in 3D Cardiac Microtissues and Reveal Non-cardiomyocyte Contributions to Heart Disease.
- Author
-
Giacomelli E, Meraviglia V, Campostrini G, Cochrane A, Cao X, van Helden RWJ, Krotenberg Garcia A, Mircea M, Kostidis S, Davis RP, van Meer BJ, Jost CR, Koster AJ, Mei H, Míguez DG, Mulder AA, Ledesma-Terrón M, Pompilio G, Sala L, Salvatori DCF, Slieker RC, Sommariva E, de Vries AAF, Giera M, Semrau S, Tertoolen LGJ, Orlova VV, Bellin M, and Mummery CL
- Subjects
- Cell Differentiation, Endothelial Cells, Humans, Myocytes, Cardiac, Stromal Cells, Heart Diseases, Induced Pluripotent Stem Cells
- Abstract
Cardiomyocytes (CMs) from human induced pluripotent stem cells (hiPSCs) are functionally immature, but this is improved by incorporation into engineered tissues or forced contraction. Here, we showed that tri-cellular combinations of hiPSC-derived CMs, cardiac fibroblasts (CFs), and cardiac endothelial cells also enhance maturation in easily constructed, scaffold-free, three-dimensional microtissues (MTs). hiPSC-CMs in MTs with CFs showed improved sarcomeric structures with T-tubules, enhanced contractility, and mitochondrial respiration and were electrophysiologically more mature than MTs without CFs. Interactions mediating maturation included coupling between hiPSC-CMs and CFs through connexin 43 (CX43) gap junctions and increased intracellular cyclic AMP (cAMP). Scaled production of thousands of hiPSC-MTs was highly reproducible across lines and differentiated cell batches. MTs containing healthy-control hiPSC-CMs but hiPSC-CFs from patients with arrhythmogenic cardiomyopathy strikingly recapitulated features of the disease. Our MT model is thus a simple and versatile platform for modeling multicellular cardiac diseases that will facilitate industry and academic engagement in high-throughput molecular screening., Competing Interests: Declaration of Interests C.L.M. is co-founder of Ncardia bv., (Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
33. Generation of two human induced pluripotent stem cell lines, LUMCi020-A and LUMCi021-A, from two patients with Catecholaminergic Polymorphic Ventricular Tachycardia carrying heterozygous mutations in the RYR2 gene.
- Author
-
Meraviglia V, Arendzen CH, Freund C, Atsma DE, Mummery CL, and Bellin M
- Subjects
- Humans, Mutation, Ryanodine Receptor Calcium Release Channel genetics, Induced Pluripotent Stem Cells, Tachycardia, Ventricular genetics
- Abstract
Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT) is a malignant channelopathy associated with exercise- and stress-induced cardiac sudden death. The autosomal dominant form of CPVT is due to mutations in the ryanodine receptor 2 (RYR2) gene. We generated induced pluripotent stem cells (hiPSCs) from skin fibroblasts of two patients carrying the c.12441 G>T and c.14885 A>G RYR2 missense mutations, respectively, using non-integrating Sendai virus. These lines show the typical morphology of pluripotent cells, express pluripotency markers, display a normal karyotype and differentiate towards the three germ layers in vitro. These lines represent a human cellular model to study the molecular basis of CPVT., Competing Interests: Declaration of Competing Interest Christine L. Mummery is co-founder of Pluriomics (now Ncardia) bv., (Copyright © 2020 The Author(s). Published by Elsevier B.V. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
34. Lipidomics, Atrial Conduction, and Body Mass Index.
- Author
-
Del Greco M F, Foco L, Teumer A, Verweij N, Paglia G, Meraviglia V, Melotti R, Vukovic V, Rauhe W, Joshi PK, Demirkan A, Felix SB, Pietzner M, Said MA, van de Vegte YJ, van der Harst P, Wright AF, Hicks AA, Campbell H, Dörr M, Snieder H, Wilson JF, Pramstaller PP, Rossini A, and Pattaro C
- Subjects
- Adult, Aged, Arteries metabolism, Electrocardiography, Female, Genome-Wide Association Study, Humans, Lipid Metabolism, Lipidomics, Male, Mendelian Randomization Analysis, Middle Aged, Risk Factors, Arteries physiopathology, Body Mass Index, Cardiovascular Diseases metabolism, Cardiovascular Diseases physiopathology, Lipids chemistry
- Abstract
Background: Lipids are increasingly involved in cardiovascular risk prediction as potential proarrhythmic influencers. However, knowledge is limited about the specific mechanisms connecting lipid alterations with atrial conduction., Methods: To shed light on this issue, we conducted a broad assessment of 151 sphingo- and phospholipids, measured using mass spectrometry, for association with atrial conduction, measured by P wave duration (PWD) from standard electrocardiograms, in the MICROS study (Microisolates in South Tyrol) (n=839). Causal pathways involving lipidomics, body mass index (BMI), and PWD were assessed using 2-sample Mendelian randomization analyses based on published genome-wide association studies of lipidomics (n=4034) and BMI (n=734 481), and genetic association analysis of PWD in 5 population-based studies (n=24 236)., Results: We identified an association with relative phosphatidylcholine 38:3 (%PC 38:3) concentration, which was replicated in the ORCADES (Orkney Complex Disease Study; n=951), with a pooled association across studies of 2.59 (95% CI, 1.3-3.9; P=1.1×10
-4 ) ms PWD per mol% increase. While being independent of cholesterol, triglycerides, and glucose levels, the %PC 38:3-PWD association was mediated by BMI. Results supported a causal effect of BMI on both PWD ( P=8.3×10-5 ) and %PC 38:3 ( P=0.014)., Conclusions: Increased %PC 38:3 levels are consistently associated with longer PWD, partly because of the confounding effect of BMI. The causal effect of BMI on PWD reinforces evidence of BMI's involvement into atrial electrical activity.- Published
- 2019
- Full Text
- View/download PDF
35. Subchronic exposure to titanium dioxide nanoparticles modifies cardiac structure and performance in spontaneously hypertensive rats.
- Author
-
Rossi S, Savi M, Mazzola M, Pinelli S, Alinovi R, Gennaccaro L, Pagliaro A, Meraviglia V, Galetti M, Lozano-Garcia O, Rossini A, Frati C, Falco A, Quaini F, Bocchi L, Stilli D, Lucas S, Goldoni M, Macchi E, Mutti A, and Miragoli M
- Subjects
- Animals, Blood Pressure drug effects, Dose-Response Relationship, Drug, Electrocardiography, Fibrosis, Heart physiopathology, Heart Rate drug effects, Hypertension physiopathology, Rats, Inbred SHR, Telemetry, Ventricular Function, Left, Heart drug effects, Hypertension pathology, Myocardium pathology, Nanoparticles toxicity, Titanium toxicity
- Abstract
Background: Non-communicable diseases, intended as the results of a combination of inherited, environmental and biological factors, kill 40 million people each year, equivalent to roughly 70% of all premature deaths globally. The possibility that manufactured nanoparticles (NPs) may affect cardiac performance, has led to recognize NPs-exposure not only as a major Public Health concern, but also as an occupational hazard. In volunteers, NPs-exposure is problematic to quantify. We recently found that inhaled titanium dioxide NPs, one of the most produced engineered nanomaterials, acutely increased cardiac excitability and promoted arrhythmogenesis in normotensive rats by a direct interaction with cardiac cells. We hypothesized that such scenario can be exacerbated by latent cardiovascular disorders such as hypertension., Results: We monitored cardiac electromechanical performance in spontaneously hypertensive rats (SHRs) exposed to titanium dioxide NPs for 6 weeks using a combination of cardiac functional measurements associated with toxicological, immunological, physical and genetic assays. Longitudinal radio-telemetry ECG recordings and multiple-lead epicardial potential mapping revealed that atrial activation times significantly increased as well as proneness to arrhythmia. At the third week of nanoparticles administration, the lung and cardiac tissue encountered a maladaptive irreversible structural remodelling starting with increased pro-inflammatory cytokines levels and lipid peroxidation, resulting in upregulation of the main pro-fibrotic cardiac genes. At the end of the exposure, the majority of spontaneous arrhythmic events terminated, while cardiac hemodynamic deteriorated and a significant accumulation of fibrotic tissue occurred as compared to control untreated SHRs. Titanium dioxide nanoparticles were quantified in the heart tissue although without definite accumulation as revealed by particle-induced X-ray emission and ultrastructural analysis., Conclusions: The co-morbidity of hypertension and inhaled nanoparticles induces irreversible hemodynamic impairment associated with cardiac structural damage potentially leading to heart failure. The time-dependence of exposure indicates a non-return point that needs to be taken into account in hypertensive subjects daily exposed to nanoparticles.
- Published
- 2019
- Full Text
- View/download PDF
36. The Histone Deacetylase Inhibitor Suberoylanilide Hydroxamic Acid (SAHA) Restores Cardiomyocyte Contractility in a Rat Model of Early Diabetes.
- Author
-
Bocchi L, Motta BM, Savi M, Vilella R, Meraviglia V, Rizzi F, Galati S, Buschini A, Lazzaretti M, Pramstaller PP, Rossini A, and Stilli D
- Subjects
- Adenosine Triphosphate metabolism, Animals, Calcium metabolism, Cells, Cultured, Diabetes Mellitus, Experimental pathology, Histone Deacetylases chemistry, Histone Deacetylases metabolism, Male, Myocytes, Cardiac metabolism, Oxidative Stress drug effects, Rats, Rats, Wistar, Reactive Oxygen Species metabolism, Histone Deacetylase Inhibitors pharmacology, Myocytes, Cardiac drug effects, Vorinostat pharmacology
- Abstract
In early diabetes, hyperglycemia and the associated metabolic dysregulation promote early changes in the functional properties of cardiomyocytes, progressively leading to the appearance of the diabetic cardiomyopathy phenotype. Recently, the interplay between histone acetyltransferases (HAT) and histone deacetylases (HDAC) has emerged as a crucial factor in the development of cardiac disorders. The present study evaluates whether HDAC inhibition can prevent the development of cardiomyocyte contractile dysfunction induced by a short period of hyperglycemia, with focus on the potential underlying mechanisms. Cell contractility and calcium dynamics were measured in unloaded ventricular myocytes isolated from the heart of control and diabetic rats. Cardiomyocytes were either untreated or exposed to the pan-HDAC inhibitor suberoylanilide hydroxamic acid (SAHA) for 90 min. Then, a fraction of each group of cells was used to evaluate the expression levels of proteins involved in the excitation-contraction coupling, and the cardiomyocyte metabolic activity, ATP content, and reactive oxygen species levels. SAHA treatment was able to counteract the initial functional derangement in cardiomyocytes by reducing cell oxidative damage. These findings suggest that early HDAC inhibition could be a promising adjuvant approach for preventing diabetes-induced cardiomyocyte oxidative damage, which triggers the pro-inflammatory signal cascade, mitochondrial damage, and ventricular dysfunction.
- Published
- 2019
- Full Text
- View/download PDF
37. Effects of smoking status, history and intensity on heart rate variability in the general population: The CHRIS study.
- Author
-
Murgia F, Melotti R, Foco L, Gögele M, Meraviglia V, Motta B, Steger A, Toifl M, Sinnecker D, Müller A, Merati G, Schmidt G, Rossini A, Pramstaller PP, and Pattaro C
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Arrhythmias, Cardiac etiology, Female, Germany epidemiology, Humans, Male, Middle Aged, Prognosis, Young Adult, Arrhythmias, Cardiac epidemiology, Heart Rate physiology, Smokers statistics & numerical data, Smoking physiopathology, Tobacco Use Disorder complications
- Abstract
Background: Heart rate variability (HRV) reflects the autonomous nervous system modulation on heart rate and is associated with several pathologies, including cardiac mortality. While mechanistic studies show that smoking is associated with lower HRV, population-based studies present conflicting results., Methods: We assessed the mutual effects of active smoking status, cumulative smoking history, and current smoking intensity, on HRV among 4751 adults from the Cooperative Health Research In South Tyrol (CHRIS) study. The HRV metrics standard deviation of normal-to-normal (NN) inter-beat intervals (SDNN), square root of the mean squared differences of consecutive NN intervals (RMSSD), total power (TP), low (LF) and high frequency (HF) power, and their ratio (LF/HF), were derived from 20-minute electrocardiograms. Smoking status, pack-years (PY), and tobacco grams/day from standardized questionnaires were the main exposures. We fitted linear mixed models to account for relatedness, non-linearity, and moderating effects, and including fractional polynomials., Results: Past smokers had higher HRV levels than never smokers, independently of PY. The association of HRV with current smoking became apparent when accounting for the interaction between smoking status and PY. In current smokers, but not in past smokers, we observed HRV reductions between 2.0% (SDNN) and 4.9% (TP) every 5 PY increase. Furthermore, current smokers were characterized by dose-response reductions of 9.8% (SDNN), 8.9% (RMSSD), 20.1% (TP), 17.7% (LF), and 19.1% (HF), respectively, every 10 grams/day of smoked tobacco, independently of common cardiometabolic conditions and HRV-modifying drugs. The LF/HF ratio was not associated with smoking status, history, or intensity., Conclusions: Smoking cessation was associated with higher HRV levels. In current smokers, heavier smoking intensity appears gradually detrimental on HRV, corroborating previous evidence. By affecting both the sympathetic and parasympathetic nervous system indexes, but not the LF/HF balance, smoking intensity seems to exert a systemic dysautonomic effect., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2019
- Full Text
- View/download PDF
38. Are Requirements to Deposit Data in Research Repositories Compatible With the European Union's General Data Protection Regulation?
- Author
-
Mascalzoni D, Bentzen HB, Budin-Ljøsne I, Bygrave LA, Bell J, Dove ES, Fuchsberger C, Hveem K, Mayrhofer MT, Meraviglia V, O'Brien DR, Pattaro C, Pramstaller PP, Rakic V, Rossini A, Shabani M, Svantesson DJB, Tomasi M, Ursin L, Wjst M, and Kaye J
- Subjects
- European Union, Humans, Biomedical Research organization & administration, Computer Security standards, Information Dissemination methods
- Published
- 2019
- Full Text
- View/download PDF
39. Derivation of human induced pluripotent stem cell line EURACi004-A from skin fibroblasts of a patient with Arrhythmogenic Cardiomyopathy carrying the heterozygous PKP2 mutation c.2569_3018del50.
- Author
-
Ermon B, Volpato CB, Cattelan G, Silipigni R, Di Segni M, Cantaloni C, Casella M, Pramstaller PP, Pompilio G, Sommariva E, Meraviglia V, and Rossini A
- Subjects
- Arrhythmias, Cardiac metabolism, Cardiomyopathies metabolism, Cell Differentiation genetics, Cell Differentiation physiology, Cells, Cultured, Fibroblasts metabolism, Heterozygote, Humans, Mutation genetics, Fibroblasts cytology, Induced Pluripotent Stem Cells cytology, Plakophilins genetics, Skin cytology
- Abstract
Arrhythmogenic Cardiomyopathy (ACM) is an inherited cardiac disease characterized by arrhythmias and fibro-fatty replacement in the ventricular myocardium. Causative mutations are mainly reported in desmosomal genes, especially in plakophilin2 (PKP2). Here, using a virus-free reprogramming approach, we generated induced pluripotent stem cells (iPSCs) from skin fibroblasts of one ACM patient carrying the frameshift heterozygous PKP2 mutation c.2569_3018del50. The iPSC line (EURACi004-A) showed the typical morphology of pluripotent cells, possessed normal karyotype and exhibited pluripotency markers and trilineage differentiation potential, including cardiomyogenic capability. Thus, this line can represent a human in vitro model to study the molecular basis of ACM., (Copyright © 2018 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
40. The arrhythmogenic cardiomyopathy-specific coding and non-coding transcriptome in human cardiac stromal cells.
- Author
-
Rainer J, Meraviglia V, Blankenburg H, Piubelli C, Pramstaller PP, Paolin A, Cogliati E, Pompilio G, Sommariva E, Domingues FS, and Rossini A
- Subjects
- Gene Expression Profiling methods, Genomics methods, Humans, Arrhythmias, Cardiac genetics, Cardiomyopathies genetics, MicroRNAs genetics, Transcriptome genetics
- Abstract
Background: Arrhythmogenic cardiomyopathy (ACM) is a genetic autosomal disease characterized by abnormal cell-cell adhesion, cardiomyocyte death, progressive fibro-adipose replacement of the myocardium, arrhythmias and sudden death. Several different cell types contribute to the pathogenesis of ACM, including, as recently described, cardiac stromal cells (CStCs). In the present study, we aim to identify ACM-specific expression profiles of human CStCs derived from endomyocardial biopsies of ACM patients and healthy individuals employing TaqMan Low Density Arrays for miRNA expression profiling, and high throughput sequencing for gene expression quantification., Results: We identified 3 miRNAs and 272 genes as significantly differentially expressed at a 5% false discovery rate. Both the differentially expressed genes as well as the target genes of the ACM-specific miRNAs were found to be enriched in cell adhesion-related biological processes. Functional similarity and protein interaction-based network analyses performed on the identified deregulated genes, miRNA targets and known ACM-causative genes revealed clusters of highly related genes involved in cell adhesion, extracellular matrix organization, lipid transport and ephrin receptor signaling., Conclusions: We determined for the first time the coding and non-coding transcriptome characteristic of ACM cardiac stromal cells, finding evidence for a potential contribution of miRNAs, specifically miR-29b-3p, to ACM pathogenesis or phenotype maintenance.
- Published
- 2018
- Full Text
- View/download PDF
41. Generation of human induced pluripotent stem cells (EURACi001-A, EURACi002-A, EURACi003-A) from peripheral blood mononuclear cells of three patients carrying mutations in the CAV3 gene.
- Author
-
Meraviglia V, Benzoni P, Landi S, Murano C, Langione M, Motta BM, Baratto S, Silipigni R, Di Segni M, Pramstaller PP, DiFrancesco D, Gazzerro E, Barbuti A, and Rossini A
- Subjects
- Caveolin 3 genetics, Cell Differentiation genetics, Cell Differentiation physiology, Cells, Cultured, Cellular Reprogramming genetics, Cellular Reprogramming physiology, Flow Cytometry, Humans, Karyotype, Mutation genetics, Mutation, Missense genetics, Reverse Transcriptase Polymerase Chain Reaction, Caveolin 3 metabolism, Induced Pluripotent Stem Cells cytology, Induced Pluripotent Stem Cells metabolism, Leukocytes, Mononuclear cytology, Leukocytes, Mononuclear metabolism
- Abstract
Caveolinopathies are a heterogeneous family of genetic pathologies arising from alterations of the caveolin-3 gene (CAV3), encoding for the isoform specifically constituting muscle caveolae. Here, by reprogramming peripheral blood mononuclear cells, we report the generation of induced pluripotent stem cells (iPSCs) from three patients carrying the ΔYTT deletion, T78K and W101C missense mutations in caveolin-3. iPSCs displayed normal karyotypes and all the features of pluripotent stem cells in terms of morphology, specific marker expression and ability to differentiate in vitro into the three germ layers. These lines thus represent a human cellular model to study the molecular basis of caveolinopathies. Resource table., (Copyright © 2017 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
42. HDAC Inhibition Improves the Sarcoendoplasmic Reticulum Ca 2+ -ATPase Activity in Cardiac Myocytes.
- Author
-
Meraviglia V, Bocchi L, Sacchetto R, Florio MC, Motta BM, Corti C, Weichenberger CX, Savi M, D'Elia Y, Rosato-Siri MD, Suffredini S, Piubelli C, Pompilio G, Pramstaller PP, Domingues FS, Stilli D, and Rossini A
- Subjects
- Acetylation, Animals, Histone Deacetylase Inhibitors pharmacology, Humans, Male, Myocytes, Cardiac enzymology, Myocytes, Cardiac metabolism, Rats, Rats, Wistar, Vorinostat, Hydroxamic Acids pharmacology, Myocytes, Cardiac drug effects, Protein Processing, Post-Translational, Sarcoplasmic Reticulum Calcium-Transporting ATPases metabolism
- Abstract
SERCA2a is the Ca
2+ ATPase playing the major contribution in cardiomyocyte (CM) calcium removal. Its activity can be regulated by both modulatory proteins and several post-translational modifications. The aim of the present work was to investigate whether the function of SERCA2 can be modulated by treating CMs with the histone deacetylase (HDAC) inhibitor suberanilohydroxamic acid (SAHA). The incubation with SAHA (2.5 µM, 90 min) of CMs isolated from rat adult hearts resulted in an increase of SERCA2 acetylation level and improved ATPase activity. This was associated with a significant improvement of calcium transient recovery time and cell contractility. Previous reports have identified K464 as an acetylation site in human SERCA2. Mutants were generated where K464 was substituted with glutamine (Q) or arginine (R), mimicking constitutive acetylation or deacetylation, respectively. The K464Q mutation ameliorated ATPase activity and calcium transient recovery time, thus indicating that constitutive K464 acetylation has a positive impact on human SERCA2a (hSERCA2a) function. In conclusion, SAHA induced deacetylation inhibition had a positive impact on CM calcium handling, that, at least in part, was due to improved SERCA2 activity. This observation can provide the basis for the development of novel pharmacological approaches to ameliorate SERCA2 efficiency., Competing Interests: The authors declare no conflict of interest.- Published
- 2018
- Full Text
- View/download PDF
43. Exploring digenic inheritance in arrhythmogenic cardiomyopathy.
- Author
-
König E, Volpato CB, Motta BM, Blankenburg H, Picard A, Pramstaller P, Casella M, Rauhe W, Pompilio G, Meraviglia V, Domingues FS, Sommariva E, and Rossini A
- Subjects
- Adult, Aged, Aged, 80 and over, Alcohol Oxidoreductases genetics, Basic Helix-Loop-Helix Transcription Factors genetics, Co-Repressor Proteins, Connectin chemistry, Connectin genetics, Dystroglycans genetics, Female, Humans, Male, Middle Aged, Models, Molecular, Mutation, Nerve Tissue Proteins genetics, Pedigree, Plakophilins genetics, Protein Domains, Repressor Proteins genetics, Exome Sequencing, ras GTPase-Activating Proteins genetics, Arrhythmogenic Right Ventricular Dysplasia genetics
- Abstract
Background: Arrhythmogenic cardiomyopathy (ACM) is an inherited genetic disorder, characterized by the substitution of heart muscle with fibro-fatty tissue and severe ventricular arrhythmias, often leading to heart failure and sudden cardiac death. ACM is considered a monogenic disorder, but the low penetrance of mutations identified in patients suggests the involvement of additional genetic or environmental factors., Methods: We used whole exome sequencing to investigate digenic inheritance in two ACM families where previous diagnostic tests have revealed a PKP2 mutation in all affected and some healthy individuals. In family members with PKP2 mutations we determined all genes that harbor variants in affected but not in healthy carriers or vice versa. We computationally prioritized the most likely candidates, focusing on known ACM genes and genes related to PKP2 through protein interactions, functional relationships, or shared biological processes., Results: We identified four candidate genes in family 1, namely DAG1, DAB2IP, CTBP2 and TCF25, and eleven candidate genes in family 2. The most promising gene in the second family is TTN, a gene previously associated with ACM, in which the affected individual harbors two rare deleterious-predicted missense variants, one of which is located in the protein's only serine kinase domain., Conclusions: In this study we report genes that might act as digenic players in ACM pathogenesis, on the basis of co-segregation with PKP2 mutations. Validation in larger cohorts is still required to prove the utility of this model.
- Published
- 2017
- Full Text
- View/download PDF
44. SNX27, a protein involved in down syndrome, regulates GPR17 trafficking and oligodendrocyte differentiation.
- Author
-
Meraviglia V, Ulivi AF, Boccazzi M, Valenza F, Fratangeli A, Passafaro M, Lecca D, Stagni F, Giacomini A, Bartesaghi R, Abbracchio MP, Ceruti S, and Rosa P
- Subjects
- Animals, Brain metabolism, Brain pathology, Cell Differentiation physiology, Cell Line, Cell Membrane metabolism, Cell Membrane pathology, Disease Models, Animal, Down Syndrome metabolism, Down Syndrome pathology, Endocytosis physiology, HEK293 Cells, Humans, Mice, Mice, Inbred C57BL, Mice, Transgenic, Oligodendroglia pathology, Sorting Nexins deficiency, Sorting Nexins genetics, Biological Transport physiology, Nerve Tissue Proteins metabolism, Oligodendroglia metabolism, Receptors, G-Protein-Coupled metabolism, Sorting Nexins metabolism
- Abstract
The G protein-coupled receptor 17 (GPR17) plays crucial roles in myelination. It is highly expressed during transition of oligodendrocyte progenitor cells to immature oligodendrocytes, but, after this stage, it must be down-regulated to allow generation of mature myelinating cells. After endocytosis, GPR17 is sorted into lysosomes for degradation or recycled to the plasma membrane. Balance between degradation and recycling is important for modulation of receptor levels at the cell surface and thus for the silencing/activation of GPR17-signaling pathways that, in turn, affect oligodendrocyte differentiation. The molecular mechanisms at the basis of these processes are still partially unknown and their characterization will allow a better understanding of myelination and provide cues to interpret the consequences of GPR17 dysfunction in diseases. Here, we demonstrate that the endocytic trafficking of GPR17 is mediated by the interaction of a type I PDZ-binding motif located at the C-terminus of the receptor and SNX27, a recently identified protein of the endosome-associated retromer complex and whose functions in oligodendrocytes have never been studied. SNX27 knock-down significantly reduces GPR17 plasma membrane recycling in differentiating oligodendrocytes while accelerating cells' terminal maturation. Interestingly, trisomy-linked down-regulation of SNX27 expression in the brain of Ts65Dn mice, a model of Down syndrome, correlates with a decrease in GPR17(+) cells and an increase in mature oligodendrocytes, which, however, fail in reaching full maturation, eventually leading to hypomyelination. Our data demonstrate that SNX27 modulates GPR17 plasma membrane recycling and stability, and that disruption of the SNX27/GPR17 interaction might contribute to pathological oligodendrocyte differentiation defects. GLIA 2016. GLIA 2016;64:1437-1460., (© 2016 Wiley Periodicals, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
45. Higher cardiogenic potential of iPSCs derived from cardiac versus skin stromal cells.
- Author
-
Meraviglia V, Wen J, Piacentini L, Campostrini G, Wang C, Florio MC, Azzimato V, Fassina L, Langes M, Wong J, Miragoli M, Gaetano C, Pompilio G, Barbuti A, DiFrancesco D, Mascalzoni D, Pramstaller PP, Colombo GI, Chen HS, and Rossini A
- Subjects
- Animals, Cells, Cultured, Humans, Induced Pluripotent Stem Cells metabolism, MicroRNAs genetics, Myocardium metabolism, Cell Differentiation, Induced Pluripotent Stem Cells cytology, Myocardium cytology
- Abstract
Prior studies have demonstrated that founder cell type could influence induced pluripotent stem cells (iPSCs) molecular and developmental properties at early passages after establishing their pluripotent state. Herein, we evaluated the persistence of a functional memory related to the tissue of origin in iPSCs from syngeneic cardiac (CStC) vs skin stromal cells (SStCs). We found that, at passages greater than 15, iPSCs from cardiac stromal cells (C-iPSCs) produced a higher number of beating embryoid bodies than iPSCs from skin stromal cells (S-iPSCs). Flow cytometry analysis revealed that dissected beating areas from C-iPSCs exhibited more Troponin-T positive cells compared to S-iPSCs. Beating areas derived from C-iPSCs displayed higher expression of cardiac markers, more hyperpolarized diastolic potentials, larger action potential amplitude and higher contractility than beaters from skin. Also, different microRNA subsets were differentially modulated in CStCs vs SStCs during the reprogramming process, potentially accounting for the higher cardiogenic potentials of C-iPSCs vs S-iPSCs. Therefore, the present work supports the existence of a founder organ memory in iPSCs obtained from the stromal component of the origin tissue.
- Published
- 2016
- Full Text
- View/download PDF
46. Acetylation mediates Cx43 reduction caused by electrical stimulation.
- Author
-
Meraviglia V, Azzimato V, Colussi C, Florio MC, Binda A, Panariti A, Qanud K, Suffredini S, Gennaccaro L, Miragoli M, Barbuti A, Lampe PD, Gaetano C, Pramstaller PP, Capogrossi MC, Recchia FA, Pompilio G, Rivolta I, and Rossini A
- Subjects
- Acetylation drug effects, Anacardic Acids administration & dosage, Animals, Connexin 43 genetics, Dogs, Electric Stimulation, Gap Junctions pathology, Heart Ventricles pathology, Histone Acetyltransferases antagonists & inhibitors, Histone Acetyltransferases metabolism, Histone Deacetylase 1 antagonists & inhibitors, Histone Deacetylase 1 metabolism, Humans, Mice, Myocytes, Cardiac pathology, RNA, Messenger biosynthesis, Cell Communication genetics, Connexin 43 biosynthesis, Gap Junctions genetics, Heart Ventricles metabolism, Myocytes, Cardiac metabolism
- Abstract
Communication between cardiomyocytes depends upon gap junctions (GJ). Previous studies have demonstrated that electrical stimulation induces GJ remodeling and modifies histone acetylase (HAT) and deacetylase (HDAC) activities, although these two results have not been linked. The aim of this work was to establish whether electrical stimulation modulates GJ-mediated cardiac cell-cell communication by acetylation-dependent mechanisms. Field stimulation of HL-1 cardiomyocytes at 0.5 Hz for 24 h significantly reduced connexin43 (Cx43) expression and cell-cell communication. HDAC activity was down-regulated whereas HAT activity was not modified resulting in increased acetylation of Cx43. Consistent with a post-translational mechanism, we did not observe a reduction in Cx43 mRNA in electrically stimulated cells, while the proteasomal inhibitor MG132 maintained Cx43 expression. Further, the treatment of paced cells with the HAT inhibitor Anacardic Acid maintained both the levels of Cx43 and cell-cell communication. Finally, we observed increased acetylation of Cx43 in the left ventricles of dogs subjected to chronic tachypacing as a model of abnormal ventricular activation. In conclusion, our findings suggest that altered electrical activity can regulate cardiomyocyte communication by influencing the acetylation status of Cx43., (Copyright © 2015 Elsevier Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
47. Generation of Induced Pluripotent Stem Cells from Frozen Buffy Coats using Non-integrating Episomal Plasmids.
- Author
-
Meraviglia V, Zanon A, Lavdas AA, Schwienbacher C, Silipigni R, Di Segni M, Chen HS, Pramstaller PP, Hicks AA, and Rossini A
- Subjects
- Cellular Reprogramming genetics, Centrifugation methods, Cryopreservation methods, Humans, Induced Pluripotent Stem Cells cytology, Leukocytes, Mononuclear cytology, Leukocytes, Mononuclear physiology, Induced Pluripotent Stem Cells physiology, Plasmids genetics
- Abstract
Somatic cells can be reprogrammed into induced pluripotent stem cells (iPSCs) by forcing the expression of four transcription factors (Oct-4, Sox-2, Klf-4, and c-Myc), typically expressed by human embryonic stem cells (hESCs). Due to their similarity with hESCs, iPSCs have become an important tool for potential patient-specific regenerative medicine, avoiding ethical issues associated with hESCs. In order to obtain cells suitable for clinical application, transgene-free iPSCs need to be generated to avoid transgene reactivation, altered gene expression and misguided differentiation. Moreover, a highly efficient and inexpensive reprogramming method is necessary to derive sufficient iPSCs for therapeutic purposes. Given this need, an efficient non-integrating episomal plasmid approach is the preferable choice for iPSC derivation. Currently the most common cell type used for reprogramming purposes are fibroblasts, the isolation of which requires tissue biopsy, an invasive surgical procedure for the patient. Therefore, human peripheral blood represents the most accessible and least invasive tissue for iPSC generation. In this study, a cost-effective and viral-free protocol using non-integrating episomal plasmids is reported for the generation of iPSCs from human peripheral blood mononuclear cells (PBMNCs) obtained from frozen buffy coats after whole blood centrifugation and without density gradient separation.
- Published
- 2015
- Full Text
- View/download PDF
48. Syngeneic cardiac and bone marrow stromal cells display tissue-specific microRNA signatures and microRNA subsets restricted to diverse differentiation processes.
- Author
-
Meraviglia V, Azzimato V, Piacentini L, Chiesa M, Kesharwani RK, Frati C, Capogrossi MC, Gaetano C, Pompilio G, Colombo GI, and Rossini A
- Subjects
- Cell Differentiation genetics, Gene Expression Profiling, Humans, Organ Specificity genetics, Bone Marrow Cells cytology, MicroRNAs genetics, Myocardium cytology, Stromal Cells cytology
- Abstract
MicroRNAs are key modulators at molecular level in different biological processes, including determination of cell fate and differentiation. Herein, microRNA expression profiling experiments were performed on syngeneic cardiac (CStC) and bone marrow (BMStC) mesenchymal stromal cells cultured in standard growth medium and then in vitro exposed to adipogenic, osteogenic, cardiomyogenic and endothelial differentiation media. Analysis identified a tissue-specific microRNA signature composed of 16 microRNAs that univocally discriminated cell type of origin and that were completely unaffected by in vitro differentiation media: 4 microRNAs were over-expressed in cardiac stromal cells, and 12 were overexpressed or present only in bone marrow stromal cells. Further, results revealed microRNA subsets specifically modulated by each differentiation medium, irrespective of the cell type of origin, and a subset of 7 microRNAs that were down-regulated by all media with respect to growth medium. Finally, we identified 16 microRNAs that were differentially modulated by the media when comparing the two tissues of origin. The existence of a tissue-specific microRNA signature surviving to any differentiation stimuli, strongly support the role if microRNAs determining cell identity related to tissue origin. Moreover, we identified microRNA subsets modulated by different culture conditions in a tissue-specific manner, pointing out their importance during differentiation processes.
- Published
- 2014
- Full Text
- View/download PDF
49. microRNAs and Cardiac Cell Fate.
- Author
-
Piubelli C, Meraviglia V, Pompilio G, D'Alessandra Y, Colombo GI, and Rossini A
- Abstract
The role of small, non-coding microRNAs (miRNAs) has recently emerged as fundamental in the regulation of the physiology of the cardiovascular system. Several specific miRNAs were found to be expressed in embryonic, postnatal, and adult cardiac tissues. In the present review, we will provide an overview about their role in controlling the different pathways regulating cell identity and fate determination. In particular, we will focus on the involvement of miRNAs in pluripotency determination and reprogramming, and specifically on cardiac lineage commitment and cell direct transdifferentiation into cardiomyocytes. The identification of cardiac-specific miRNAs and their targets provide new promising insights into the mechanisms that regulate cardiac development, function and dysfunction. Furthermore, due to their contribution in reprogramming, they could offer new opportunities for developing safe and efficient cell-based therapies for cardiovascular disorders.
- Published
- 2014
- Full Text
- View/download PDF
50. Human chorionic villus mesenchymal stromal cells reveal strong endothelial conversion properties.
- Author
-
Meraviglia V, Vecellio M, Grasselli A, Baccarin M, Farsetti A, Capogrossi MC, Pompilio G, Coviello DA, Gaetano C, Di Segni M, and Rossini A
- Subjects
- Bone Marrow growth & development, Cell Proliferation, Chorionic Villi growth & development, Endothelium cytology, Genomic Instability, Homeodomain Proteins metabolism, Humans, Nanog Homeobox Protein, Octamer Transcription Factor-3 metabolism, SOXB1 Transcription Factors metabolism, Telomerase metabolism, Cell Differentiation, Cell Lineage, Culture Media, Endothelium growth & development, Mesenchymal Stem Cells cytology
- Abstract
Chorion, amnion and villi are reservoirs of mesenchymal stromal cells (StC) and the hypothesis that StC from fetal tissues retain higher plasticity compared to adult StC has been suggested. Aimed at investigating this aspect, a series of in vitro experiments were performed with StC isolated from first trimester human chorionic villi (CVStC). CVStC were cultured in: (i) standard mesenchymal medium (MM) and (ii) AmniomaxII® (AM), specifically designed to grow amnion-derived cells in prenatal diagnostic procedures. Cells were then exposed to distinct differentiation treatments and distinguished according to morphology, immunophenotype and molecular markers. Human StC obtained from adult bone marrow (BMStC) were used as control. CVStC cultured either in MM or AM presented stromal morphology and immunophenotype, were negative for pluripotency factors (Nanog, Oct-4 and Sox-2), lacked detectable telomerase activity and retained high genomic stability. In AM, however, CVStC exhibited a faster proliferation rate compared to BMStC or CVStC kept in MM. During differentiation, CVStC were less efficient than BMStC in acquiring adipocytes and osteocytes features; the cardiomyogenic conversion occurred at low efficiency in both cell types. Remarkably, in the presence of pro-angiogenic factors, CVStC reprogrammed toward an endothelial-like phenotype at significantly higher efficiency than BMStC. This effect was particularly evident in CVStC expanded in AM. Mechanistically, the reduced CVStC expression of anti-angiogenic microRNA could support this process. The present study demonstrates that, despite of fetal origin, CVStC exhibit restricted plasticity, distinct from that of BMStC and predominantly directed toward the endothelial lineage., (Copyright © 2012 International Society of Differentiation. Published by Elsevier B.V. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.