93
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

      Modulation of human embryonic stem cell-derived cardiomyocyte growth: A testbed for studying human cardiac hypertrophy?

      research-article
      a , b , * , a , a , a , a , b , a , a , a , 1 , a , 1
      Journal of Molecular and Cellular Cardiology
      Academic Press
      ANF, atrial natriuretic factor, bFGF, basic human fibroblast growth factor, CaMK II, Ca2+/calmodulin-dependent kinase II, EB, embryoid body, ERK, extracellular signal-regulated kinases, GSK3, glycogen synthase kinase 3, HDACII, histone deacetylase, FKBP, FK506 binding protein, hESC, human embryonic stem cells, hESC-CM, human embryonic stem cell-derived cardiomyocytes, JNK, c-Jun N-terminal kinases, MAP2K4 and MAP2K3, MAPK kinase 4 and 3, respectively, MEF, mouse embryonic fibroblast, MHC, myosin heavy chains, MOI, multiplicity of infection, mTOR, mammalian target of rapamycin, p38–MAPK, p38 mitogen-activated protein kinase, PKG, protein kinase G, Ryr2, cardiac ryanodine receptor 2, and SERCA2, sarco/endoplasmic reticulum Ca2±-ATPase., Embryonic stem cells, Cardiomyocytes, Human, Protein kinases, Hypertrophy

      Read this article at

      ScienceOpenPublisherPMC
      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Human embryonic stem cell-derived cardiomyocytes (hESC-CM) are being developed for tissue repair and as a model system for cardiac physiology and pathophysiology. However, the signaling requirements of their growth have not yet been fully characterized. We showed that hESC-CM retain their capacity for increase in size in long-term culture. Exposing hESC-CM to hypertrophic stimuli such as equiaxial cyclic stretch, angiotensin II, and phenylephrine (PE) increased cell size and volume, percentage of hESC-CM with organized sarcomeres, levels of ANF, and cytoskeletal assembly. PE effects on cell size were separable from those on cell cycle. Changes in cell size by PE were completely inhibited by p38–MAPK, calcineurin/FKBP, and mTOR blockers. p38–MAPK and calcineurin were also implicated in basal cell growth. Inhibitors of ERK, JNK, and CaMK II partially reduced PE effects; PKG or GSK3β inhibitors had no effect. The role of p38–MAPK was confirmed by an additional pharmacological inhibitor and adenoviral infection of hESC-CM with a dominant-inhibitory form of p38–MAPK. Infection of hESC-CM with constitutively active upstream MAP2K3b resulted in an increased cell size, sarcomere and cytoskeletal assembly, elongation of the cells, and induction of ANF mRNA levels. siRNA knockdown of p38–MAPK inhibited PE-induced effects on cell size. These results reveal an important role for active protein kinase signaling in hESC-CM growth and hypertrophy, with potential implications for hESC-CM as a novel in vitro test system. This article is part of a special issue entitled, "Cardiovascular Stem Cells Revisited".

          Research Highlights

          ► Human embryonic stem cell-derived cardiomyocytes increase in size. ► Their growth is stimulated by classical physiological and pathological hypertrophic agents. ► A number of hypertrophic pathways act in this process, including p38–MAPK, calcineurin, FKBP, mTOR, HDAC II, ERK, JNK, and CAMK II. ► The regulation of cell growth and cell cycle progression are separable processes. ► The development of the cells can be a tool for the cardiac researcher or pharmaceutical industry.

          Related collections

          Most cited references30

          • Record: found
          • Abstract: found
          • Article: not found

          Feeder-free growth of undifferentiated human embryonic stem cells.

          Previous studies have shown that maintenance of undifferentiated human embryonic stem (hES) cells requires culture on mouse embryonic fibroblast (MEF) feeders. Here we demonstrate a successful feeder-free hES culture system in which undifferentiated cells can be maintained for at least 130 population doublings. In this system, hES cells are cultured on Matrigel or laminin in medium conditioned by MEF. The hES cells maintained on feeders or off feeders express integrin alpha6 and beta1, which may form a laminin-specific receptor. The hES cell populations in feeder-free conditions maintained a normal karyotype, stable proliferation rate, and high telomerase activity. Similar to cells cultured on feeders, hES cells maintained under feeder-free conditions expressed OCT-4, hTERT, alkaline phosphatase, and surface markers including SSEA-4, Tra 1-60, and Tra 1-81. In addition, hES cells maintained without direct feeder contact formed teratomas in SCID/beige mice and differentiated in vitro into cells from all three germ layers. Thus, the cells retain fundamental characteristics of hES cells in this culture system and are suitable for scaleup production.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Patient-specific induced pluripotent stem cell derived models of LEOPARD syndrome

            Generation of reprogrammed induced pluripotent stem cells (iPSC) from patients with defined genetic disorders promises important avenues to understand the etiologies of complex diseases, and the development of novel therapeutic interventions. We have generated iPSC from patients with LEOPARD syndrome (LS; acronym of its main features: Lentigines, Electrocardiographic abnormalities, Ocular hypertelorism, Pulmonary valve stenosis, Abnormal genitalia, Retardation of growth and Deafness), an autosomal dominant developmental disorder belonging to a relatively prevalent class of inherited RAS-MAPK signaling diseases, which also includes Noonan syndrome (NS), with pleiomorphic effects on several tissues and organ systems1,2. The patient-derived cells have a mutation in the PTPN11 gene, which encodes the SHP2 phosphatase. The iPSC have been extensively characterized and produce multiple differentiated cell lineages. A major disease phenotype in patients with LEOPARD syndrome is hypertrophic cardiomyopathy. We show that in vitro-derived cardiomyocytes from LS-iPSC are larger, have a higher degree of sarcomeric organization and preferential localization of NFATc4 in the nucleus when compared to cardiomyocytes derived from human embryonic stem cells (HESC) or wild type (wt) iPSC derived from a healthy brother of one of the LS patients. These features correlate with a potential hypertrophic state. We also provide molecular insights into signaling pathways that may promote the disease phenotype.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Rapamycin attenuates load-induced cardiac hypertrophy in mice.

              Cardiac hypertrophy, or an increase in heart size, is an important risk factor for cardiac morbidity and mortality. The mammalian target of rapamycin (mTOR) is a component of the insulin-phosphoinositide 3-kinase pathway, which is known to play a critical role in the determination of cell, organ, and body size. To examine the role of mTOR in load-induced cardiac hypertrophy, we administered rapamycin, a specific inhibitor of mTOR, to mice with ascending aortic constriction. Activity of p70 ribosomal S6 kinase 1 (S6K1), an effector of mTOR, was increased by 3.8-fold in the aortic-constricted heart. Pretreatment of mice with 2 mg. kg-1. d-1 of rapamycin completely suppressed S6K1 activation and S6 phosphorylation in response to pressure overload. The heart weight/tibial length ratio of vehicle-treated aortic-banded mice was increased by 34.4+/-3.6% compared with vehicle-treated sham-operated mice. Rapamycin suppressed the load-induced increase in heart weight by 67%. Attenuation of cardiac hypertrophy by rapamycin was associated with attenuation of the increase in myocyte cell size induced by aortic constriction. Rapamycin did not cause loss of body weight, lethality, or left ventricular dysfunction. mTOR or its target(s) seems to play an important role in load-induced cardiac hypertrophy. Because systemic administration of rapamycin has been used successfully for the treatment of transplant rejection in clinical practice, it may be a useful therapeutic modality to suppress cardiac hypertrophy in patients.
                Bookmark

                Author and article information

                Journal
                J Mol Cell Cardiol
                J. Mol. Cell. Cardiol
                Journal of Molecular and Cellular Cardiology
                Academic Press
                0022-2828
                1095-8584
                February 2011
                February 2011
                : 50
                : 2-4
                : 367-376
                Affiliations
                [a ]National Heart and Lung Institute, Imperial College London, UK
                [b ]Heart Center, Semmelweis University, Budapest, Hungary
                Author notes
                [* ]Corresponding author. National Heart and Lung Institute, Imperial College London, Flowers Building, Armstrong Road, London, UK, SW7 2AZ. Tel.: +44 207 594 3009; fax: +44 20 7823 3392. g.foldes@ 123456imperial.ac.uk
                [1]

                These authors contributed equally.

                Article
                YJMCC6950
                10.1016/j.yjmcc.2010.10.029
                3034871
                21047517
                1a426dac-cf31-48f5-8461-baabf86734ec
                © 2011 Elsevier Ltd.

                This document may be redistributed and reused, subject to certain conditions.

                History
                : 29 June 2010
                : 12 October 2010
                : 26 October 2010
                Categories
                Original Article

                Cardiovascular Medicine
                human,pkg, protein kinase g,hypertrophy,camk ii, ca2+/calmodulin-dependent kinase ii,map2k4 and map2k3, mapk kinase 4 and 3, respectively,mef, mouse embryonic fibroblast,embryonic stem cells,hdacii, histone deacetylase,hesc-cm, human embryonic stem cell-derived cardiomyocytes,cardiomyocytes,protein kinases,erk, extracellular signal-regulated kinases,moi, multiplicity of infection,mtor, mammalian target of rapamycin,bfgf, basic human fibroblast growth factor,jnk, c-jun n-terminal kinases,p38–mapk, p38 mitogen-activated protein kinase,ryr2, cardiac ryanodine receptor 2,mhc, myosin heavy chains,gsk3, glycogen synthase kinase 3,eb, embryoid body,and serca2, sarco/endoplasmic reticulum ca2±-atpase.,hesc, human embryonic stem cells,fkbp, fk506 binding protein,anf, atrial natriuretic factor

                Comments

                Comment on this article