Skip to main content
Top
Published in: Angiogenesis 3/2013

01-07-2013 | Original Paper

Scleroderma Mesenchymal Stem Cells display a different phenotype from healthy controls; implications for regenerative medicine

Authors: Paola Cipriani, Alessandra Marrelli, Paola Di Benedetto, Vasiliki Liakouli, Francesco Carubbi, Piero Ruscitti, Saverio Alvaro, Ilenia Pantano, Antonio Francesco Campese, Paola Grazioli, Isabella Screpanti, Roberto Giacomelli

Published in: Angiogenesis | Issue 3/2013

Login to get access

Abstract

Introduction

Vascular involvement is a key feature of Systemic sclerosis (SSc). Although the pericytes/endothelial cells (ECs) cross-talk regulates vessels formation, no evidences about the pericytes contribution to ineffective angiogenesis in SSc are available. Recent findings showed similarities between pericytes and Bone Marrow Mesenchymal Stem Cells (BM-MSCs). Due to difficulties in pericytes isolation, this work explores the possibility to use BM-MSCs as pericytes surrogate, clarifying their role in supporting neo-angiogenesis during SSc.

Methods

To demonstrate their potential to normally differentiate into pericytes, both SSc and healthy controls (HC) BM-MSCs were treated with TGF-β and PDGF-BB. The expression of pericytes specific markers (α-SMA, NG2, RGS5 and desmin) was assessed by qPCR, western blot, and immunofluorescence; chemioinvasion and capillary morphogenesis were also performed. Cell-sorting of BM-MSCs co-cultured with HC-ECs was used to identify a possible change in contractile proteins genes expression.

Results

We showed that BM-MSCs isolated from SSc patients displayed an up-regulation of α-SMA and SM22α genes and a reduced proliferative activity. Moreover during SSc, both TGF-β and PDGF-BB can specifically modulate BM-MSCs toward pericytes. TGF-β was found interfering with the PDGF-BB effects. Using BM-MSCs/MVECs co-culture system we observed that SSc BM-MSCs improve ECs tube formation in stressed condition, and BM-MSCs, sorted after co-culture, showed a reduced α-SMA and SM22α gene expression.

Conclusions

BM-MSCs from SSc patients behave as pericytes. They display a more mature and myofibroblast-like phenotype, probably related to microenvironmental cues operating during the disease. After their co-culture with HC-MVECs, SSc BM-MSCs underwent to a phenotypic modulation which re-programs these cells toward a pro-angiogenic behaviour.
Literature
1.
2.
go back to reference Kahaleh MB (2004) Vascular involvement in systemic sclerosis (SSc). Clin Exp Rheumatol 22(3 Suppl 33): S19–S23 Kahaleh MB (2004) Vascular involvement in systemic sclerosis (SSc). Clin Exp Rheumatol 22(3 Suppl 33): S19–S23
3.
go back to reference Fleming JN, Nash RA, Mahoney WM Jr, Schwartz SM (2009) Is scleroderma a vasculopathy? Curr Rheumatol Rep 11:103–110PubMedCrossRef Fleming JN, Nash RA, Mahoney WM Jr, Schwartz SM (2009) Is scleroderma a vasculopathy? Curr Rheumatol Rep 11:103–110PubMedCrossRef
4.
go back to reference Cipriani P, Guiducci S, Miniati I, Cinelli M, Urbani S et al (2007) Impairment of endothelial cell differentiation from bone marrow-derived mesenchymal stem cells:new insight into the pathogenesis of systemic sclerosis. Arthritis Rheum 56:1994–2004PubMedCrossRef Cipriani P, Guiducci S, Miniati I, Cinelli M, Urbani S et al (2007) Impairment of endothelial cell differentiation from bone marrow-derived mesenchymal stem cells:new insight into the pathogenesis of systemic sclerosis. Arthritis Rheum 56:1994–2004PubMedCrossRef
5.
go back to reference Manetti M, Ibba-Manneschi L, Liakouli V, Guiducci S, Milia AF et al (2010) The IL1-like cytokine IL33 and its receptor ST2 are abnormally expressed in the affected skin and visceral organs of patients with systemic sclerosis. Ann Rheum Dis 69:598–605PubMedCrossRef Manetti M, Ibba-Manneschi L, Liakouli V, Guiducci S, Milia AF et al (2010) The IL1-like cytokine IL33 and its receptor ST2 are abnormally expressed in the affected skin and visceral organs of patients with systemic sclerosis. Ann Rheum Dis 69:598–605PubMedCrossRef
6.
go back to reference Cipriani P, Franca Milia A, Liakouli V, Pacini A, Manetti M et al (2006) Differential expression of stromal cell-derived factor 1 and its receptor CXCR4 in the skin and endothelial cells of systemic sclerosis patients: pathogenetic implications. Arthritis Rheum 54:3022–3033PubMedCrossRef Cipriani P, Franca Milia A, Liakouli V, Pacini A, Manetti M et al (2006) Differential expression of stromal cell-derived factor 1 and its receptor CXCR4 in the skin and endothelial cells of systemic sclerosis patients: pathogenetic implications. Arthritis Rheum 54:3022–3033PubMedCrossRef
7.
go back to reference Cipriani P, Marrelli A, Liakouli V, Di Benedetto P, Giacomelli R (2011) Cellular players in angiogenesis during the course of systemic sclerosis. Autoimmun Rev 10:641–646PubMedCrossRef Cipriani P, Marrelli A, Liakouli V, Di Benedetto P, Giacomelli R (2011) Cellular players in angiogenesis during the course of systemic sclerosis. Autoimmun Rev 10:641–646PubMedCrossRef
8.
go back to reference Liakouli V, Cipriani P, Marrelli A, Alvaro S, Ruscitti P et al (2011) Angiogenic cytokines and growth factors in systemic sclerosis. Autoimmun Rev 10:590–594PubMedCrossRef Liakouli V, Cipriani P, Marrelli A, Alvaro S, Ruscitti P et al (2011) Angiogenic cytokines and growth factors in systemic sclerosis. Autoimmun Rev 10:590–594PubMedCrossRef
11.
go back to reference Hirschi KK, D’Amore PA (1996) Pericytes in the microvasculature. Cardiovasc Res 32:687–698PubMed Hirschi KK, D’Amore PA (1996) Pericytes in the microvasculature. Cardiovasc Res 32:687–698PubMed
12.
go back to reference Antonelli-Orlidge A, Smith SR, D’Amore PA (1989) Influence of pericytes on capillary endothelial cell growth. Am Rev Respir Dis 140:1129–1131PubMedCrossRef Antonelli-Orlidge A, Smith SR, D’Amore PA (1989) Influence of pericytes on capillary endothelial cell growth. Am Rev Respir Dis 140:1129–1131PubMedCrossRef
13.
14.
go back to reference Armulik A, Abramsson A, Betsholtz C (2005) Endothelial/pericyte interactions. Circ Res 97:512–523PubMedCrossRef Armulik A, Abramsson A, Betsholtz C (2005) Endothelial/pericyte interactions. Circ Res 97:512–523PubMedCrossRef
15.
go back to reference Takakura N (2011) Role of intimate interactions between endothelial cells and the surrounding accessory cells in the maturation of blood vessels. J Thromb Haemost 9:144–150PubMedCrossRef Takakura N (2011) Role of intimate interactions between endothelial cells and the surrounding accessory cells in the maturation of blood vessels. J Thromb Haemost 9:144–150PubMedCrossRef
16.
go back to reference Hirschi KK, Rohovsky SA, D’Amore PA (1998) PDGF, TGF-beta, and heterotypic cell–cell interactions mediate endothelial cell-induced recruitment of 10T1/2 cells and their differentiation to a smooth muscle fate. J Cell Biol 141:805–814PubMedCrossRef Hirschi KK, Rohovsky SA, D’Amore PA (1998) PDGF, TGF-beta, and heterotypic cell–cell interactions mediate endothelial cell-induced recruitment of 10T1/2 cells and their differentiation to a smooth muscle fate. J Cell Biol 141:805–814PubMedCrossRef
18.
go back to reference Darland DC, D’Amore PA (2001) TGF beta is required for the formation of capillary-like structures in three-dimensional cocultures of 10T1/2 and endothelial cells. Angiogenesis 4:11–20PubMedCrossRef Darland DC, D’Amore PA (2001) TGF beta is required for the formation of capillary-like structures in three-dimensional cocultures of 10T1/2 and endothelial cells. Angiogenesis 4:11–20PubMedCrossRef
19.
go back to reference Crisan M, Yap S, Casteilla L, Chen CW, Corselli M et al (2008) A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell 11:301–313CrossRef Crisan M, Yap S, Casteilla L, Chen CW, Corselli M et al (2008) A perivascular origin for mesenchymal stem cells in multiple human organs. Cell Stem Cell 11:301–313CrossRef
20.
go back to reference da Silva Meirelles L, Caplan AI, Nardi NB (2008) In search of the in vivo identity of mesenchymal stem cells. Stem Cells 26:2287–2299PubMedCrossRef da Silva Meirelles L, Caplan AI, Nardi NB (2008) In search of the in vivo identity of mesenchymal stem cells. Stem Cells 26:2287–2299PubMedCrossRef
21.
go back to reference Cai X, Lin Y, Friedrich CC, Neville C, Pomerantseva I et al (2009) Bone marrow derived pluripotent cells are pericytes which contribute to vascularization. Stem Cell Rev 5:437–445PubMedCrossRef Cai X, Lin Y, Friedrich CC, Neville C, Pomerantseva I et al (2009) Bone marrow derived pluripotent cells are pericytes which contribute to vascularization. Stem Cell Rev 5:437–445PubMedCrossRef
22.
go back to reference Bryan BA, D’Amore PA (2008) Pericyte isolation and use in endothelial/pericyte coculture models. Methods Enzymol 443:315–331PubMedCrossRef Bryan BA, D’Amore PA (2008) Pericyte isolation and use in endothelial/pericyte coculture models. Methods Enzymol 443:315–331PubMedCrossRef
23.
go back to reference Sacchetti B, Funari A, Michienzi S, Di Cesare S, Piersanti S et al (2007) Self-renewing osteoprogenitors in bone marrow sinusoids can organize a hematopoietic microenvironment. Cell 19:324–336CrossRef Sacchetti B, Funari A, Michienzi S, Di Cesare S, Piersanti S et al (2007) Self-renewing osteoprogenitors in bone marrow sinusoids can organize a hematopoietic microenvironment. Cell 19:324–336CrossRef
24.
go back to reference Tormin A, Li O, Brune JC, Walsh S, Schütz B et al (2011) CD146 expression on primary nonhematopoietic bone marrow stem cells is correlated with in situ localization. Blood 12:5067–5077CrossRef Tormin A, Li O, Brune JC, Walsh S, Schütz B et al (2011) CD146 expression on primary nonhematopoietic bone marrow stem cells is correlated with in situ localization. Blood 12:5067–5077CrossRef
25.
go back to reference da Silva Meirelles L, Sand TT, Harman RJ, Lennon DP, Caplan AI (2009) MSC frequency correlates with blood vessel in equine adipose tissue. Tissue Eng Part A 15:221–229PubMedCrossRef da Silva Meirelles L, Sand TT, Harman RJ, Lennon DP, Caplan AI (2009) MSC frequency correlates with blood vessel in equine adipose tissue. Tissue Eng Part A 15:221–229PubMedCrossRef
26.
go back to reference Toma JG, Akhavan M, Fernandes KJ, Barnabé-Heider F, Sadikot A et al (2001) Isolation of multipotent adult stem cells from the dermis of mammalian skin. Nat Cell Biol 3:778–784PubMedCrossRef Toma JG, Akhavan M, Fernandes KJ, Barnabé-Heider F, Sadikot A et al (2001) Isolation of multipotent adult stem cells from the dermis of mammalian skin. Nat Cell Biol 3:778–784PubMedCrossRef
27.
go back to reference Gronthos S, Mankani M, Brahim J, Robey PG, Shi S (2000) Postnatal human dental pulp stem cells (DPSCs) in vitro and in vivo. Proc Natl Acad Sci USA 97:13625–13630PubMedCrossRef Gronthos S, Mankani M, Brahim J, Robey PG, Shi S (2000) Postnatal human dental pulp stem cells (DPSCs) in vitro and in vivo. Proc Natl Acad Sci USA 97:13625–13630PubMedCrossRef
28.
go back to reference Díaz-Flores L Jr, Madrid JF, Gutiérrez R, Varela H, Valladares F (2006) Adult stem and transit-amplifying cell location. Histol Histopathol 21:995–1027PubMed Díaz-Flores L Jr, Madrid JF, Gutiérrez R, Varela H, Valladares F (2006) Adult stem and transit-amplifying cell location. Histol Histopathol 21:995–1027PubMed
29.
go back to reference Hoofnagle MH, Thomas JA, Wamhoff BR, Owens GK (2006) Origin of neointimal smooth muscle: we’ve come full circle. Arterioscler Thromb Vasc Biol 26:2579–2581PubMedCrossRef Hoofnagle MH, Thomas JA, Wamhoff BR, Owens GK (2006) Origin of neointimal smooth muscle: we’ve come full circle. Arterioscler Thromb Vasc Biol 26:2579–2581PubMedCrossRef
30.
go back to reference Owens GK (1995) Regulation of differentiation of vascular smooth muscle cells. Physiol Rev 75:487–517PubMed Owens GK (1995) Regulation of differentiation of vascular smooth muscle cells. Physiol Rev 75:487–517PubMed
31.
go back to reference Mack CP (2011) Signaling mechanisms that regulate smooth muscle cell differentiation. Arterioscler Thromb Vasc Biol 31:1495–1505PubMedCrossRef Mack CP (2011) Signaling mechanisms that regulate smooth muscle cell differentiation. Arterioscler Thromb Vasc Biol 31:1495–1505PubMedCrossRef
32.
go back to reference Challier JC, Kacemi A, Olive G (1995) Mixed culture of pericytes and endothelial cells from fetal microvessels of the human placenta. Cell Mol Biol 41:233–241PubMed Challier JC, Kacemi A, Olive G (1995) Mixed culture of pericytes and endothelial cells from fetal microvessels of the human placenta. Cell Mol Biol 41:233–241PubMed
33.
go back to reference Helmbold P, Nayak RC, Marsch WC, Herman IM (2001) Isolation and in vitro characterization of human dermal microvascular pericytes. Microvasc Res 61:160–165PubMedCrossRef Helmbold P, Nayak RC, Marsch WC, Herman IM (2001) Isolation and in vitro characterization of human dermal microvascular pericytes. Microvasc Res 61:160–165PubMedCrossRef
34.
go back to reference Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F et al (2006) Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 8:315–317PubMedCrossRef Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F et al (2006) Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 8:315–317PubMedCrossRef
35.
go back to reference Hungerford JE, Little CD (1999) Developmental biology of the vascular smooth muscle cell. J Vasc Res 36:2–27PubMedCrossRef Hungerford JE, Little CD (1999) Developmental biology of the vascular smooth muscle cell. J Vasc Res 36:2–27PubMedCrossRef
36.
go back to reference Owens GK, Kumar MS, Wamhoff BR (2004) Molecular regulation of vascular smooth muscle cell differentiation in development and disease. Physiol Rev 84:767–801PubMedCrossRef Owens GK, Kumar MS, Wamhoff BR (2004) Molecular regulation of vascular smooth muscle cell differentiation in development and disease. Physiol Rev 84:767–801PubMedCrossRef
37.
go back to reference Aikawa M, Sakomura Y, Ueda M, Kimura K, Manabe I et al (1997) Differentiation of smooth muscle cells after coronary angioplasty determined via myosin heavy chain expression. Circulation 96:82–90PubMedCrossRef Aikawa M, Sakomura Y, Ueda M, Kimura K, Manabe I et al (1997) Differentiation of smooth muscle cells after coronary angioplasty determined via myosin heavy chain expression. Circulation 96:82–90PubMedCrossRef
38.
go back to reference Mulvany MJ, Baumbach GL, Aalkjaer C, Heagerty AM, Korsgaard N et al (1996) Vascular remodelling. Hypertension 28(3):505–506PubMed Mulvany MJ, Baumbach GL, Aalkjaer C, Heagerty AM, Korsgaard N et al (1996) Vascular remodelling. Hypertension 28(3):505–506PubMed
39.
go back to reference Helmbold P, Fiedler E, Fischer M, Marsch WCh (2004) Hyperplasia of dermal microvascular pericytes in scleroderma. J Cutan Pathol 31:431–440PubMedCrossRef Helmbold P, Fiedler E, Fischer M, Marsch WCh (2004) Hyperplasia of dermal microvascular pericytes in scleroderma. J Cutan Pathol 31:431–440PubMedCrossRef
40.
go back to reference Rajkumar VS, Sundberg C, Abraham DJ, Rubin K, Black CM (1999) Activation of microvascular pericytes in autoimmune Raynaud’s phenomenon and systemic sclerosis. Arthritis Rheum 42:930–941PubMedCrossRef Rajkumar VS, Sundberg C, Abraham DJ, Rubin K, Black CM (1999) Activation of microvascular pericytes in autoimmune Raynaud’s phenomenon and systemic sclerosis. Arthritis Rheum 42:930–941PubMedCrossRef
41.
go back to reference Fleming JN, Nash RA, McLeod DO, Fiorentino DF, Shulman HM, et al (2008) Capillary regeneration in scleroderma: stem cell therapy reverses phenotype? PLoS One 3(1):e 14–52. doi: 10.1371/journal.pone.0001452 Fleming JN, Nash RA, McLeod DO, Fiorentino DF, Shulman HM, et al (2008) Capillary regeneration in scleroderma: stem cell therapy reverses phenotype? PLoS One 3(1):e 14–52. doi: 10.​1371/​journal.​pone.​0001452
42.
go back to reference Witmer AN, van Blijswijk BC, van Noorden CJF, Vrensen GFJM, Schlingemann RO (2004) In vivo angiogenic phenotype of endothelial cells and pericyte induced by vascular endothelial growth factor-A. J Histochem Cytochem 52:39–52PubMedCrossRef Witmer AN, van Blijswijk BC, van Noorden CJF, Vrensen GFJM, Schlingemann RO (2004) In vivo angiogenic phenotype of endothelial cells and pericyte induced by vascular endothelial growth factor-A. J Histochem Cytochem 52:39–52PubMedCrossRef
43.
go back to reference Palumbo R, Gaetano C, Melillo G, Toschi E, Remuzzi A et al (2000) Shear stress downregulation of platelet-derived growth factor receptor-beta and matrix metalloprotease-2 is associated with inhibition of smooth muscle cell invasion and migration. Circulation 102:225–230PubMedCrossRef Palumbo R, Gaetano C, Melillo G, Toschi E, Remuzzi A et al (2000) Shear stress downregulation of platelet-derived growth factor receptor-beta and matrix metalloprotease-2 is associated with inhibition of smooth muscle cell invasion and migration. Circulation 102:225–230PubMedCrossRef
44.
go back to reference Newby AC (2006) Matrix metalloproteinases regulate migration, proliferation, and death of vascular smooth muscle cells by degrading matrix and non-matrix substrates. Cardiovasc Res 69:614–624PubMedCrossRef Newby AC (2006) Matrix metalloproteinases regulate migration, proliferation, and death of vascular smooth muscle cells by degrading matrix and non-matrix substrates. Cardiovasc Res 69:614–624PubMedCrossRef
45.
go back to reference Risinger GM Jr, Hunt TS, Updike DL, Bullen EC, Howard EW (2006) Matrix metalloproteinase-2 expression by vascular smooth muscle cells is mediated by both stimulatory and inhibitory signals in response to growth factors. J Biol Chem 281:25915–25925PubMedCrossRef Risinger GM Jr, Hunt TS, Updike DL, Bullen EC, Howard EW (2006) Matrix metalloproteinase-2 expression by vascular smooth muscle cells is mediated by both stimulatory and inhibitory signals in response to growth factors. J Biol Chem 281:25915–25925PubMedCrossRef
46.
go back to reference Tokunaga A, Oya T, Ishii Y, Motomura H, Nakamura C et al (2008) PDGF receptor beta is a potent regulator of mesenchymal stromal cell function. J Bone Miner Res 23:1519–1528PubMedCrossRef Tokunaga A, Oya T, Ishii Y, Motomura H, Nakamura C et al (2008) PDGF receptor beta is a potent regulator of mesenchymal stromal cell function. J Bone Miner Res 23:1519–1528PubMedCrossRef
47.
go back to reference Varga J, Pasche B (2009) Transforming growth factor beta as a therapeutic target in systemic sclerosis. Nat Rev Rheumatol 5(4):200–206PubMedCrossRef Varga J, Pasche B (2009) Transforming growth factor beta as a therapeutic target in systemic sclerosis. Nat Rev Rheumatol 5(4):200–206PubMedCrossRef
48.
go back to reference Hunzelmann N, Brinckmann J (2010) What are the new milestones in the pathogenesis of Systemic Sclerosis? Ann Rheum Dis 69:52–56CrossRef Hunzelmann N, Brinckmann J (2010) What are the new milestones in the pathogenesis of Systemic Sclerosis? Ann Rheum Dis 69:52–56CrossRef
49.
go back to reference von Tell D, Armulik A, Betsholtz C (2006) Pericytes and vascular stability. Exp Cell Res 312(5):623–629CrossRef von Tell D, Armulik A, Betsholtz C (2006) Pericytes and vascular stability. Exp Cell Res 312(5):623–629CrossRef
50.
51.
go back to reference Rouwkema J, de Boer J, Van Blitterswijk CA (2006) Endothelial cells assemble into a 3-dimensional provascular network in a bone tissue engineering construct. Tissue Eng 12(9):2685–2693PubMedCrossRef Rouwkema J, de Boer J, Van Blitterswijk CA (2006) Endothelial cells assemble into a 3-dimensional provascular network in a bone tissue engineering construct. Tissue Eng 12(9):2685–2693PubMedCrossRef
52.
go back to reference Forte A, Della Corte A, De Feo M, Cerasuolo F, Cipollaro M (2010) Role of myofibroblasts in vascular remodelling: focus on restenosis and aneurysm. Cardiovasc Res 88:395–405PubMedCrossRef Forte A, Della Corte A, De Feo M, Cerasuolo F, Cipollaro M (2010) Role of myofibroblasts in vascular remodelling: focus on restenosis and aneurysm. Cardiovasc Res 88:395–405PubMedCrossRef
Metadata
Title
Scleroderma Mesenchymal Stem Cells display a different phenotype from healthy controls; implications for regenerative medicine
Authors
Paola Cipriani
Alessandra Marrelli
Paola Di Benedetto
Vasiliki Liakouli
Francesco Carubbi
Piero Ruscitti
Saverio Alvaro
Ilenia Pantano
Antonio Francesco Campese
Paola Grazioli
Isabella Screpanti
Roberto Giacomelli
Publication date
01-07-2013
Publisher
Springer Netherlands
Published in
Angiogenesis / Issue 3/2013
Print ISSN: 0969-6970
Electronic ISSN: 1573-7209
DOI
https://doi.org/10.1007/s10456-013-9338-9

Other articles of this Issue 3/2013

Angiogenesis 3/2013 Go to the issue
Live Webinar | 27-06-2024 | 18:00 (CEST)

Keynote webinar | Spotlight on medication adherence

Live: Thursday 27th June 2024, 18:00-19:30 (CEST)

WHO estimates that half of all patients worldwide are non-adherent to their prescribed medication. The consequences of poor adherence can be catastrophic, on both the individual and population level.

Join our expert panel to discover why you need to understand the drivers of non-adherence in your patients, and how you can optimize medication adherence in your clinics to drastically improve patient outcomes.

Prof. Kevin Dolgin
Prof. Florian Limbourg
Prof. Anoop Chauhan
Developed by: Springer Medicine
Obesity Clinical Trial Summary

At a glance: The STEP trials

A round-up of the STEP phase 3 clinical trials evaluating semaglutide for weight loss in people with overweight or obesity.

Developed by: Springer Medicine

Highlights from the ACC 2024 Congress

Year in Review: Pediatric cardiology

Watch Dr. Anne Marie Valente present the last year's highlights in pediatric and congenital heart disease in the official ACC.24 Year in Review session.

Year in Review: Pulmonary vascular disease

The last year's highlights in pulmonary vascular disease are presented by Dr. Jane Leopold in this official video from ACC.24.

Year in Review: Valvular heart disease

Watch Prof. William Zoghbi present the last year's highlights in valvular heart disease from the official ACC.24 Year in Review session.

Year in Review: Heart failure and cardiomyopathies

Watch this official video from ACC.24. Dr. Biykem Bozkurt discusses last year's major advances in heart failure and cardiomyopathies.