Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Invest Dermatol ; 137(10): 2197-2207, 2017 10.
Article in English | MEDLINE | ID: mdl-28647344

ABSTRACT

On acquisition of an oncogenic mutation, primary human and mouse cells can enter oncogene-induced senescence (OIS). OIS is characterized by a stable proliferation arrest and secretion of proinflammatory cytokines and chemokines, the senescence-associated secretory phenotype. Proliferation arrest and the senescence-associated secretory phenotype collaborate to enact tumor suppression, the former by blocking cell proliferation and the latter by recruiting immune cells to clear damaged cells. However, the interactions of OIS cells with the immune system are still poorly defined. Here, we show that engagement of OIS in primary human melanocytes, specifically by melanoma driver mutations NRASQ61K and BRAFV600E, causes expression of the major histocompatibility class II antigen presentation apparatus, via secreted IL-1ß signaling and expression of CIITA, a master regulator of major histocompatibility class II gene transcription. In vitro, OIS melanocytes activate T-cell proliferation. In vivo, nonproliferating oncogene-expressing melanocytes localize to skin-draining lymph nodes, where they induce T-cell proliferation and an antigen presentation gene expression signature. In patients, expression of major histocompatibility class II in melanoma is linked to favorable disease outcome. We propose that OIS in melanocytes is accompanied by an antigen presentation phenotype, likely to promote tumor suppression via activation of the adaptive immune system.


Subject(s)
Genes, MHC Class II/genetics , Melanocytes/metabolism , Melanoma/genetics , Oncogenes/genetics , Up-Regulation , Animals , Cell Line, Tumor , Cell Proliferation , Cellular Senescence , Humans , Melanocytes/pathology , Melanoma/metabolism , Melanoma/pathology , Mice , Signal Transduction
2.
Cell Rep ; 12(9): 1483-96, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26299965

ABSTRACT

Oncogene-induced senescence (OIS) is a tumor suppression mechanism that blocks cell proliferation in response to oncogenic signaling. OIS is frequently accompanied by multinucleation; however, the origin of this is unknown. Here, we show that multinucleate OIS cells originate mostly from failed mitosis. Prior to senescence, mutant H-RasV12 activation in primary human fibroblasts compromised mitosis, concordant with abnormal expression of mitotic genes functionally linked to the observed mitotic spindle and chromatin defects. Simultaneously, H-RasV12 activation enhanced survival of cells with damaged mitoses, culminating in extended mitotic arrest and aberrant exit from mitosis via mitotic slippage. ERK-dependent transcriptional upregulation of Mcl1 was, at least in part, responsible for enhanced survival and slippage of cells with mitotic defects. Importantly, mitotic slippage and oncogene signaling cooperatively induced senescence and key senescence effectors p21 and p16. In summary, activated Ras coordinately triggers mitotic disruption and enhanced cell survival to promote formation of multinucleate senescent cells.


Subject(s)
Cellular Senescence , Giant Cells/cytology , Mitosis , ras Proteins/metabolism , Cell Line , Cells, Cultured , Giant Cells/metabolism , Humans , Mutation , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , ras Proteins/genetics
3.
Genes Dev ; 28(24): 2712-25, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25512559

ABSTRACT

Cellular senescence is a stable proliferation arrest that suppresses tumorigenesis. Cellular senescence and associated tumor suppression depend on control of chromatin. Histone chaperone HIRA deposits variant histone H3.3 and histone H4 into chromatin in a DNA replication-independent manner. Appropriately for a DNA replication-independent chaperone, HIRA is involved in control of chromatin in nonproliferating senescent cells, although its role is poorly defined. Here, we show that nonproliferating senescent cells express and incorporate histone H3.3 and other canonical core histones into a dynamic chromatin landscape. Expression of canonical histones is linked to alternative mRNA splicing to eliminate signals that confer mRNA instability in nonproliferating cells. Deposition of newly synthesized histones H3.3 and H4 into chromatin of senescent cells depends on HIRA. HIRA and newly deposited H3.3 colocalize at promoters of expressed genes, partially redistributing between proliferating and senescent cells to parallel changes in expression. In senescent cells, but not proliferating cells, promoters of active genes are exceptionally enriched in H4K16ac, and HIRA is required for retention of H4K16ac. HIRA is also required for retention of H4K16ac in vivo and suppression of oncogene-induced neoplasia. These results show that HIRA controls a specialized, dynamic H4K16ac-decorated chromatin landscape in senescent cells and enforces tumor suppression.


Subject(s)
Cell Cycle Proteins/metabolism , Cellular Senescence/physiology , Histone Chaperones/metabolism , Transcription Factors/metabolism , Animals , Antineoplastic Agents, Hormonal/pharmacology , Carcinogenesis/drug effects , Carcinogenesis/genetics , Cell Cycle Proteins/genetics , Cell Line , Cell Proliferation , Cellular Senescence/genetics , Chromatin/metabolism , Female , Gene Expression Regulation/drug effects , Genetic Markers , Histone Chaperones/genetics , Histones/genetics , Histones/metabolism , Humans , Male , Mice , Papilloma/pathology , Skin Neoplasms/pathology , Tamoxifen/pharmacology , Transcription Factors/genetics
4.
Nat Cell Biol ; 15(12): 1495-506, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24270890

ABSTRACT

Altered DNA methylation and associated destabilization of genome integrity and function is a hallmark of cancer. Replicative senescence is a tumour suppressor process that imposes a limit on the proliferative potential of normal cells that all cancer cells must bypass. Here we show by whole-genome single-nucleotide bisulfite sequencing that replicative senescent human cells exhibit widespread DNA hypomethylation and focal hypermethylation. Hypomethylation occurs preferentially at gene-poor, late-replicating, lamin-associated domains and is linked to mislocalization of the maintenance DNA methyltransferase (DNMT1) in cells approaching senescence. Low-level gains of methylation are enriched in CpG islands, including at genes whose methylation and silencing is thought to promote cancer. Gains and losses of methylation in replicative senescence are thus qualitatively similar to those in cancer, and this 'reprogrammed' methylation landscape is largely retained when cells bypass senescence. Consequently, the DNA methylome of senescent cells might promote malignancy, if these cells escape the proliferative barrier.


Subject(s)
Cellular Senescence/genetics , Epigenesis, Genetic , Neoplasms/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , CpG Islands , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation , Gene Expression , Genome, Human , Humans , Nerve Tissue Proteins/genetics , Promoter Regions, Genetic , Protein Transport
5.
Proc Natl Acad Sci U S A ; 110(40): 16009-14, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24043806

ABSTRACT

Cellular senescence is a stable proliferation arrest associated with an altered secretory pathway (senescence-associated secretory phenotype). Cellular senescence is also a tumor suppressor mechanism, to which both proliferation arrest and senescence-associated secretory phenotype are thought to contribute. The melanocytes within benign human nevi are a paradigm for tumor-suppressive senescent cells in a premalignant neoplasm. Here a comparison of proliferating and senescent melanocytes and melanoma cell lines by RNA sequencing emphasizes the importance of senescence-associated proliferation arrest in suppression of transformation. Previous studies showed that activation of the Wnt signaling pathway can delay or bypass senescence. Consistent with this, we present evidence that repression of Wnt signaling contributes to melanocyte senescence in vitro. Surprisingly, Wnt signaling is active in many senescent human melanocytes in nevi, and this is linked to histological indicators of higher proliferative and malignant potential. In a mouse, activated Wnt signaling delays senescence-associated proliferation arrest to expand the population of senescent oncogene-expressing melanocytes. These results suggest that Wnt signaling can potentiate nevogenesis in vivo by delaying senescence. Further, we suggest that activated Wnt signaling in human nevi undermines senescence-mediated tumor suppression and enhances the probability of malignancy.


Subject(s)
Cellular Senescence/physiology , Melanocytes/physiology , Melanoma/etiology , Nevus/physiopathology , Wnt Signaling Pathway/physiology , Animals , Cell Line, Tumor , DNA Primers/genetics , HEK293 Cells , Humans , Immunoblotting , Immunohistochemistry , Melanocytes/cytology , Mice , Microarray Analysis , Nevus/metabolism , Real-Time Polymerase Chain Reaction , Sequence Analysis, RNA
6.
J Cell Biol ; 202(1): 129-43, 2013 Jul 08.
Article in English | MEDLINE | ID: mdl-23816621

ABSTRACT

Cellular senescence is a stable proliferation arrest, a potent tumor suppressor mechanism, and a likely contributor to tissue aging. Cellular senescence involves extensive cellular remodeling, including of chromatin structure. Autophagy and lysosomes are important for recycling of cellular constituents and cell remodeling. Here we show that an autophagy/lysosomal pathway processes chromatin in senescent cells. In senescent cells, lamin A/C-negative, but strongly γ-H2AX-positive and H3K27me3-positive, cytoplasmic chromatin fragments (CCFs) budded off nuclei, and this was associated with lamin B1 down-regulation and the loss of nuclear envelope integrity. In the cytoplasm, CCFs were targeted by the autophagy machinery. Senescent cells exhibited markers of lysosomal-mediated proteolytic processing of histones and were progressively depleted of total histone content in a lysosome-dependent manner. In vivo, depletion of histones correlated with nevus maturation, an established histopathologic parameter associated with proliferation arrest and clinical benignancy. We conclude that senescent cells process their chromatin via an autophagy/lysosomal pathway and that this might contribute to stability of senescence and tumor suppression.


Subject(s)
Cellular Senescence , Chromatin/metabolism , Lysosomes/metabolism , Autophagy , Biological Transport , Cell Membrane Permeability , Cell Nucleus/metabolism , Cells, Cultured , Chromatin/genetics , Chromatin Assembly and Disassembly , Cytoplasm/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Histones/metabolism , Humans , Laminin/metabolism , Nuclear Envelope/metabolism , Proteolysis , Time-Lapse Imaging
7.
Cell Rep ; 3(4): 1012-9, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23602572

ABSTRACT

The HIRA chaperone complex, comprised of HIRA, UBN1, and CABIN1, collaborates with histone-binding protein ASF1a to incorporate histone variant H3.3 into chromatin in a DNA replication-independent manner. To better understand HIRA's function and mechanism, we integrated HIRA, UBN1, ASF1a, and histone H3.3 chromatin immunoprecipitation sequencing and gene expression analyses. Most HIRA-binding sites colocalize with UBN1, ASF1a, and H3.3 at active promoters and active and weak/poised enhancers. At promoters, binding of HIRA/UBN1/ASF1a correlates with the level of gene expression. HIRA is required for deposition of histone H3.3 at its binding sites. There are marked differences in nucleosome and coregulator composition at different classes of HIRA-bound regulatory sites. Underscoring this, we report physical interactions between the HIRA complex and transcription factors, a chromatin insulator and an ATP-dependent chromatin-remodeling complex. Our results map the distribution of the HIRA chaperone across the chromatin landscape and point to different interacting partners at functionally distinct regulatory sites.


Subject(s)
Cell Cycle Proteins/metabolism , Chromatin/metabolism , Histone Chaperones/metabolism , Histones/metabolism , Transcription Factors/metabolism , Binding Sites , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Chromatin Immunoprecipitation , Cluster Analysis , Genetic Loci , HeLa Cells , Histone Chaperones/antagonists & inhibitors , Histone Chaperones/genetics , Histones/genetics , Humans , Molecular Chaperones , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic , RNA Interference , RNA, Small Interfering/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics
8.
Nat Cell Biol ; 14(4): 339-41, 2012 Apr 02.
Article in English | MEDLINE | ID: mdl-22469830

ABSTRACT

Cellular senescence is a stable proliferation arrest induced by triggers such as short telomeres, activated oncogenes and genotoxic stress. Two studies show that cellular senescence induced by genotoxic stress depends on chronic DNA-damage signalling from irreparable damage to telomeres. Hence, dysfunctional or damaged telomeres are the initiators of multiple modes of senescence.


Subject(s)
DNA Damage , Telomere/metabolism , Animals , Humans
9.
J Cell Mol Med ; 16(7): 1508-21, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22003890

ABSTRACT

We previously showed that human cardiomyocyte progenitor cells (hCMPCs) injected after myocardial infarction (MI) had differentiated into cardiomyocytes in vivo 3 months after MI. Here, we investigated the short-term (2 weeks) effects of hCMPCs on the infarcted mouse myocardium. MI was induced in immunocompromised (NOD/scid) mice, immediately followed by intramyocardial injection of hCMPCs labelled with enhanced green fluorescent protein (hCMPC group) or vehicle only (control group). Sham-operated mice served as reference. Cardiac performance was measured 2 and 14 days after MI by magnetic resonance imaging at 9.4 T. Left ventricular (LV) pressure-volume measurements were performed at day 15 followed by extensive immunohistological analysis. Animals injected with hCMPCs demonstrated a higher LV ejection fraction, lower LV end-systolic volume and smaller relaxation time constant than control animals 14 days after MI. hCMPCs engrafted in the infarcted myocardium, did not differentiate into cardiomyocytes, but increased vascular density and proliferation rate in the infarcted and border zone area of the hCMPC group. Injected hCMPCs engraft into murine infarcted myocardium where they improve LV systolic function and attenuate the ventricular remodelling process 2 weeks after MI. Since no cardiac differentiation of hCMPCs was evident after 2 weeks, the observed beneficial effects were most likely mediated by paracrine factors, targeting amongst others vascular homeostasis. These results demonstrate that hCMPCs can be applied to repair infarcted myocardium without the need to undergo differentiation into cardiomyocytes.


Subject(s)
Cell Differentiation , Ischemia/therapy , Myoblasts/transplantation , Myocardial Infarction/therapy , Myocytes, Cardiac/cytology , Animals , Cells, Cultured , Green Fluorescent Proteins/metabolism , Humans , Ischemia/pathology , Magnetic Resonance Imaging , Male , Mice , Mice, Inbred NOD , Mice, SCID , Myoblasts/cytology , Myocardial Infarction/pathology , Ventricular Function, Left , Ventricular Remodeling
10.
Mol Cell Biol ; 31(19): 4107-18, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21807893

ABSTRACT

The mammalian HIRA/UBN1/ASF1a complex is a histone chaperone complex that is conserved from yeast (Saccharomyces cerevisiae) to humans. This complex preferentially deposits the histone variant H3.3 into chromatin in a DNA replication-independent manner and is implicated in diverse chromatin regulatory events from gene activation to heterochromatinization. In yeast, the orthologous complex consists of three Hir proteins (Hir1p, Hir2p, and Hir3p), Hpc2p, and Asf1p. Yeast Hir3p has weak homology to CABIN1, a fourth member of the human complex, suggesting that Hir3p and CABIN1 may be orthologs. Here we show that HIRA and CABIN1 interact at ectopic and endogenous levels of expression in cells, and we isolate the quaternary HIRA/UBN1/CABIN1/ASF1a (HUCA) complex, assembled from recombinant proteins. Mutational analyses support the view that HIRA acts as a scaffold to bring together UBN1, ASF1a, and CABIN1 into a quaternary complex. We show that, like HIRA, UBN1, and ASF1a, CABIN1 is involved in heterochromatinization of the genome of senescent human cells. Moreover, in proliferating cells, HIRA and CABIN1 regulate overlapping sets of genes, and these genes are enriched in the histone variant H3.3. In sum, these data demonstrate that CABIN1 is a functional member of the human HUCA complex and so is the likely ortholog of yeast Hir3p.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , Histone Chaperones/metabolism , Histones/metabolism , Molecular Chaperones/metabolism , Multiprotein Complexes/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Cycle Proteins/genetics , Cell Line , Cellular Senescence , Histone Chaperones/genetics , Histones/genetics , Humans , Molecular Chaperones/genetics , Nuclear Proteins/genetics , Protein Binding , Transcription Factors/genetics
11.
Basic Res Cardiol ; 106(5): 829-47, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21516490

ABSTRACT

Adult epicardial cells are required for endogenous cardiac repair. After myocardial injury, they are reactivated, undergo epithelial-to-mesenchymal transformation (EMT) and migrate into the injured myocardium where they generate various cell types, including coronary smooth muscle cells and cardiac interstitial fibroblasts, which contribute to cardiac repair. To understand what drives epicardial EMT, we used an in vitro model for human adult epicardial cells. These cells have an epithelium-like morphology and markedly express the cell surface marker vascular cell adhesion marker (VCAM-1). In culture, epicardial cells spontaneously undergo EMT after which the spindle-shaped cells now express endoglin. Both epicardial cells before and after EMT express the epicardial marker, Wilms tumor 1 (WT1). Adding transforming growth factor beta (TGFß) induces loss of epithelial character and initiates the onset of mesenchymal differentiation in human adult epicardial cells. In this study, we show that TGFß-induced EMT is dependent on type-1 TGFß receptor activity and can be inhibited by soluble VCAM-1. We also show that epicardial-specific knockdown of Wilms tumor-1 (WT1) induces the process of EMT in human adult epicardial cells, through transcriptional regulation of platelet-derived growth factor receptor alpha (Pdgfrα), Snai1 and VCAM-1. These data provide new insights into the process of EMT in human adult epicardial cells, which might provide opportunities to develop new strategies for endogenous cell-based cardiac repair.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , Pericardium/cytology , Pericardium/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta/metabolism , WT1 Proteins/metabolism , Antigens, CD/metabolism , Cell Differentiation/physiology , Cells, Cultured , Endoglin , Humans , In Vitro Techniques , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Receptors, Cell Surface/metabolism , Snail Family Transcription Factors , Transcription Factors/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
12.
J Cell Mol Med ; 15(12): 2675-83, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21251220

ABSTRACT

The myocardium of the developing heart tube is covered by epicardium. These epicardial cells undergo a process of epithelial-to-mesenchymal transformation (EMT) and develop into epicardium-derived cells (EPDCs). The ingrowing EPDCs differentiate into several celltypes of which the cardiac fibroblasts form the main group. Disturbance of EMT of the epicardium leads to serious hypoplasia of the myocardium, abnormal coronary artery differentiation and Purkinje fibre paucity. Interestingly, the electrophysiological properties of epicardial cells and whether EMT influences electrical conductivity of epicardial cells is not yet known. We studied the electrophysiological aspects of epicardial cells before and after EMT in a dedicated in vitro model, using micro-electrode arrays to investigate electrical conduction across epicardial cells. Therefore, human adult epicardial cells were placed between two neonatal rat cardiomyocyte populations. Before EMT the epicardial cells have a cobblestone (epithelium-like) phenotype that was confirmed by staining for the cell-adhesion molecule ß-catenin. After spontaneous EMT in vitro the EPDCs acquired a spindle-shaped morphology confirmed by vimentin staining. When comparing both types we observed that the electrical conduction is influenced by EMT, resulting in significantly reduced conductivity of spindle-shaped EPDCs, associated with a conduction block. Furthermore, the expression of both gap junction (connexins 40, Cx43 and Cx45) and ion channel proteins (SCN5a, CACNA1C and Kir2.1) was down-regulated after EMT. This study shows for the first time the conduction differences between epicardial cells before and after EMT. These differences may be of relevance for the role of EPDCs in cardiac development, and in EMT-related cardiac dysfunction.


Subject(s)
Cell Differentiation , Electric Conductivity , Epithelial-Mesenchymal Transition/physiology , Pericardium/cytology , Pericardium/metabolism , Animals , Blotting, Western , Cells, Cultured , Humans , Male , Microscopy, Fluorescence , Myocardium/cytology , Myocardium/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , RNA, Messenger/genetics , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
13.
Circ Res ; 103(2): 167-76, 2008 Jul 18.
Article in English | MEDLINE | ID: mdl-18556577

ABSTRACT

Alignment of cardiomyocytes (CMCs) contributes to the anisotropic (direction-related) tissue structure of the heart, thereby facilitating efficient electrical and mechanical activation of the ventricles. This study aimed to investigate the effects of forced alignment of stem cells during cardiomyogenic differentiation on their functional integration with CMC cultures. Labeled neonatal rat (nr) mesenchymal stem cells (nrMSCs) were allowed to differentiate into functional heart muscle cells in different cell-alignment patterns during 10 days of coculture with nrCMCs. Development of functional cellular properties was assessed by measuring impulse transmission across these stem cells between 2 adjacent nrCMC fields, cultured onto microelectrode arrays and previously separated by a laser-dissected channel (230+/-10 microm) for nrMSC transplantation. Coatings in these channels were microabraded in a direction (1) parallel or (2) perpendicular to the channel or were (3) left unabraded to establish different cell patterns. Application of cells onto microabraded coatings resulted in anisotropic cell alignment within the channel. Application on unabraded coatings resulted in isotropic (random) alignment. After coculture, conduction across seeded nrMSCs occurred from day 1 (perpendicular and isotropic) or day 6 (parallel) onward. Conduction velocity across nrMSCs at day 10 was highest in the perpendicular (11+/-0.9 cm/sec; n=12), intermediate in the isotropic (7.1+/-1 cm/sec; n=11) and lowest in the parallel configuration (4.9+/-1 cm/sec; n=11) (P<0.01). nrCMCs and fibroblasts served as positive and negative control, respectively. Also, immunocytochemical analysis showed alignment-dependent increases in connexin 43 expression. In conclusion, forced alignment of nrMSCs undergoing cardiomyogenic differentiation affects the time course and degree of functional integration with surrounding cardiac tissue.


Subject(s)
Cell Communication/physiology , Cell Differentiation/physiology , Heart Conduction System/physiology , Mesenchymal Stem Cells/cytology , Myocytes, Cardiac/cytology , Animals , Cells, Cultured , Coculture Techniques , Connexin 43/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Male , Mesenchymal Stem Cells/metabolism , Microelectrodes , Models, Animal , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Patch-Clamp Techniques , Rats , Rats, Wistar
14.
Stem Cells ; 26(4): 1083-93, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18203678

ABSTRACT

Human mesenchymal stem cells (hMSCs) have only a limited differentiation potential toward cardiomyocytes. Forced expression of the cardiomyogenic transcription factor myocardin may stimulate hMSCs to acquire a cardiomyogenic phenotype, thereby improving their possible therapeutic potential. hMSCs were transduced with green fluorescent protein (GFP) and myocardin (hMSC(myoc)) or GFP and empty vector (hMSC). After coronary ligation in immune-compromised NOD/scid mice, hMSC(myoc) (n = 10), hMSC (n = 10), or medium only (n = 12) was injected into the infarct area. Sham-operated mice (n = 12) were used to determine baseline characteristics. Left ventricular (LV) volumes and ejection fraction (EF) were serially (days 2 and 14) assessed using 9.4-T magnetic resonance imaging. LV pressure-volume measurements were performed at day 15, followed by histological evaluation. At day 2, no differences in infarct size, LV volumes, or EF were observed among the myocardial infarction groups. At day 14, left ventricular ejection fraction in both cell-treated groups was preserved compared with the nontreated group; in addition, hMSC(myoc) injection also reduced LV volumes compared with medium injection (p < .05). Furthermore, pressure-volume measurements revealed a significantly better LV function after hMSC(myoc) injection compared with hMSC treatment. Immunohistochemistry at day 15 demonstrated that the engraftment rate was higher in the hMSC(myoc) group compared with the hMSC group (p < .05). Furthermore, these cells expressed a number of cardiomyocyte-specific markers not observed in the hMSC group. After myocardial infarction, injection of hMSC(myoc) improved LV function and limited LV remodeling, effects not observed after injection of hMSC. Furthermore, forced myocardin expression improved engraftment and induced a cardiomyocyte-like phenotype hMSC differentiation.


Subject(s)
Mesenchymal Stem Cell Transplantation/methods , Myocardial Ischemia/metabolism , Myocardial Ischemia/surgery , Nuclear Proteins/biosynthesis , Trans-Activators/biosynthesis , Adult , Animals , Cell Differentiation/physiology , Cells, Cultured , Gene Expression Regulation/physiology , Humans , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred NOD , Mice, SCID , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Phenotype , Trans-Activators/genetics , Trans-Activators/physiology
15.
Circulation ; 116(18): 2018-28, 2007 Oct 30.
Article in English | MEDLINE | ID: mdl-17938287

ABSTRACT

BACKGROUND: Nonresponse to cardiac resynchronization therapy is associated with the presence of slow or nonconducting scar tissue. Genetic modification of scar tissue, aimed at improving conduction, may be a novel approach to achieve effective resynchronization. Therefore, the feasibility of resynchronization with genetically modified human ventricular scar fibroblasts was studied in a coculture model. METHODS AND RESULTS: An in vitro model was used to study the effects of forced expression of the myocardin (MyoC) gene in human ventricular scar fibroblasts (hVSFs) on resynchronization of 2 rat cardiomyocyte fields separated by a strip of hVSFs. Furthermore, the effects of MyoC expression on the capacity of hVSFs to serve as pacing sites were studied. MyoC-dependent gene activation in hVSFs was examined by gene and immunocytochemical analysis. Forced MyoC expression in hVSFs decreased dyssynchrony, expressed as the activation delay between 2 cardiomyocyte fields (control hVSFs 27.6+/-0.2 ms [n=11] versus MyoC-hVSFs 3.6+/-0.3 ms [n=11] at day 8, P<0.01). Also, MyoC-hVSFs could be stimulated electrically, which resulted in simultaneous activation of the 2 adjacent cardiomyocyte fields. Forced MyoC expression in hVSFs led to the expression of various connexin and cardiac ion channel genes. Intracellular measurements of MyoC-hVSFs coupled to surrounding cardiomyocytes showed strongly improved action potential conduction. CONCLUSIONS: Forced MyoC gene expression in hVSFs allowed electrical stimulation of these cells and conferred the ability to conduct an electrical impulse at high velocity, which resulted in resynchronization of 2 separated cardiomyocyte fields. Both phenomena appear mediated mainly by MyoC-dependent activation of genes that encode connexins, strongly enforcing intercellular electrical coupling.


Subject(s)
Action Potentials/physiology , Cicatrix/physiopathology , Fibroblasts/physiology , Myocytes, Cardiac/physiology , Animals , Cells, Cultured , Cicatrix/metabolism , Cicatrix/pathology , Coculture Techniques/methods , Connexins/biosynthesis , Connexins/genetics , Electric Stimulation/methods , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation/physiology , Heart Ventricles/cytology , Heart Ventricles/metabolism , Humans , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Nuclear Proteins/physiology , Rats , Rats, Wistar , Trans-Activators/physiology , Transcriptional Activation , Ventricular Function
16.
Am J Physiol Heart Circ Physiol ; 293(4): H2438-47, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17644573

ABSTRACT

Mesenchymal stem cells (MSCs) from healthy donors improve cardiac function in experimental acute myocardial infarction (AMI) models. However, little is known about the therapeutic capacity of human MSCs (hMSCs) from patients with ischemic heart disease (IHD). Therefore, the behavior of hMSCs from IHD patients in an immune-compromised mouse AMI model was studied. Enhanced green fluorescent protein-labeled hMSCs from IHD patients (hMSC group: 2 x 10(5) cells in 20 microl, n = 12) or vehicle only (medium group: n = 14) were injected into infarcted myocardium of NOD/scid mice. Sham-operated mice were used as the control (n = 10). Cardiac anatomy and function were serially assessed using 9.4-T magnetic resonance imaging (MRI); 2 wk after cell transplantation, immunohistological analysis was performed. At day 2, delayed-enhancement MRI showed no difference in myocardial infarction (MI) size between the hMSC and medium groups (33 +/- 2% vs. 36 +/- 2%; P = not significant). A comparable increase in left ventricular (LV) volume and decrease in ejection fraction (EF) was observed in both MI groups. However, at day 14, EF was higher in the hMSC than in the medium group (24 +/- 3% vs. 16 +/- 2%; P < 0.05). This was accompanied by increased vascularity and reduced thinning of the infarct scar. Engrafted hMSCs (4.1 +/- 0.3% of injected cells) expressed von Willebrand factor (16.9 +/- 2.7%) but no stringent cardiac or smooth muscle markers. hMSCs from patients with IHD engraft in infarcted mouse myocardium and preserve LV function 2 wk after AMI, potentially through an enhancement of scar vascularity and a reduction of wall thinning.


Subject(s)
Adult Stem Cells/pathology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/pathology , Myocardial Infarction/surgery , Myocardial Ischemia/pathology , Myocardium/pathology , Ventricular Function, Left , Adult Stem Cells/metabolism , Animals , Body Weight , Cell Differentiation , Cell Survival , Cells, Cultured , Coronary Vessels/pathology , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , Lung/pathology , Magnetic Resonance Imaging, Cine , Male , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/metabolism , Organ Size , Research Design , Stroke Volume , Time Factors , Transduction, Genetic , Ventricular Remodeling , von Willebrand Factor/metabolism
17.
FASEB J ; 21(12): 3369-79, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17579192

ABSTRACT

Myocardial scar formation impairs heart function by inducing cardiac remodeling, decreasing myocardial compliance, and compromising normal electrical conduction. Conversion of myocardial scar fibroblasts (MSFs) into (functional) cardiomyocytes may be an effective alternative treatment to limit loss of cardiac performance after myocardial injury. In this study, we investigated whether the phenotype of MSFs can be modified by gene transfer into cells with properties of cardiomyocytes. To this end, fibroblasts from postmyocardial infarction scars of human left ventricles were isolated and characterized by cell biological, immunological, and molecular biological assays. Cultured human MSFs express GATA4 and connexin 43 and display adipogenic differentiation potential. Infection of human MSFs with a lentivirus vector encoding the potent cardiogenic transcription factor myocardin renders them positive for a wide variety of cardiomyocyte-specific proteins, including sarcomeric components, transcription factors, and ion channels, and induces the expression of several smooth muscle marker genes. Forced myocardin expression also endowed human MSFs with the ability to transmit an action potential and to repair an artificially created conduction block in cardiomyocyte cultures. These finding indicate that in vivo myocardin gene transfer may potentially limit cardiomyocyte loss, myocardial fibrosis, and disturbances in electrical conduction caused by myocardial infarction.


Subject(s)
Cell Differentiation/physiology , Cicatrix , Fibroblasts/physiology , Myocardial Infarction/pathology , Myocardium , Myocytes, Cardiac/physiology , Nuclear Proteins/genetics , Trans-Activators/genetics , Adipogenesis , Animals , Biomarkers/metabolism , Cells, Cultured , Electric Conductivity , Fibroblasts/cytology , Gene Expression Regulation , Genetic Vectors/genetics , Genetic Vectors/metabolism , HeLa Cells , Heart Conduction System/physiology , Heart Ventricles/cytology , Heart Ventricles/pathology , Humans , Lentivirus/genetics , Lentivirus/metabolism , Myocardium/cytology , Myocardium/pathology , Myocytes, Cardiac/cytology , Nuclear Proteins/metabolism , Osteogenesis , Phenotype , Promoter Regions, Genetic , Rats , Trans-Activators/metabolism , Transcription Factors/metabolism , Transduction, Genetic
18.
Stem Cells ; 25(2): 271-8, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16990583

ABSTRACT

Myocardial and coronary development are both critically dependent on epicardial cells. During cardiomorphogenesis, a subset of epicardial cells undergoes an epithelial-to-mesenchymal transition (EMT) and invades the myocardium to differentiate into various cell types, including coronary smooth muscle cells and perivascular and cardiac interstitial fibroblasts. Our current knowledge of epicardial EMT and the ensuing epicardium-derived cells (EPDCs) comes primarily from studies of chick and mouse embryonic development. Due to the absence of an in vitro culture system, very little is known about human EPDCs. Here, we report for the first time the establishment of cultures of primary epicardial cells from human adults and describe their immunophenotype, transcriptome, transducibility, and differentiation potential in vitro. Changes in morphology and beta-catenin staining pattern indicated that human epicardial cells spontaneously undergo EMT early during ex vivo culture. The surface antigen profile of the cells after EMT closely resembles that of subepithelial fibroblasts; however, only EPDCs express the cardiac marker genes GATA4 and cardiac troponin T. After infection with an adenovirus vector encoding the transcription factor myocardin or after treatment with transforming growth factor-beta1 or bone morphogenetic protein-2, EPDCs obtain characteristics of smooth muscle cells. Moreover, EPDCs can undergo osteogenesis but fail to form adipocytes or endothelial cells in vitro. Cultured epicardial cells from human adults recapitulate at least part of the differentiation potential of their embryonic counterparts and represent an excellent model system to explore the biological properties and therapeutic potential of these cells.


Subject(s)
Cell Differentiation , Epithelial Cells/cytology , Mesoderm/cytology , Myocytes, Smooth Muscle/cytology , Pericardium/cytology , Adenoviridae , Adult , Biomarkers/metabolism , Cell Separation , Cells, Cultured , Epithelial Cells/metabolism , Fibroblasts/cytology , Gene Expression Regulation , Genetic Vectors , Humans , Immunophenotyping , Mesenchymal Stem Cells/cytology , Mesoderm/metabolism , Myocytes, Smooth Muscle/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Osteogenesis , Pericardium/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Transduction, Genetic
19.
Cardiovasc Res ; 72(2): 282-91, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-16956599

ABSTRACT

OBJECTIVE: The purpose of the study was to investigate the development of electrical transmission across human adult bone marrow-derived mesenchymal stem cells (hMSCs) during long-term co-incubation with cardiomyocytes (CMCs). METHODS: Neonatal rat CMCs were cultured in multi-electrode array dishes. A conduction block was induced by creating a central acellular channel, yielding two asynchronously beating CMC fields. Enhanced green fluorescent protein (eGFP)-labeled hMSCs from ischemic heart disease patients (n=8), eGFP-labeled hMSCs having RNA interference-mediated connexin43 (Cx43) knockdown (n=6), 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine (Dil)-labeled CMCs (n=6), or no cells (n=9) were seeded in the channel. Assessment of conduction velocity (CV), Cx expression and localization, gap junctional coupling, and intracellular electrical recordings were performed for up to 14 days. RESULTS: Resynchronization of the two CMC fields occurred within 24 h after seeding of hMSCs. CV across hMSCs increased from 1.4+/-0.4 cm/s at day 7 to 3.5+/-0.1 cm/s (p<0.05) at day 14. CV across seeded CMCs was 16.8+/-0.2 cm/s throughout this period. No resynchronization occurred in the absence of seeded cells. Knockdown of Cx43 in hMSCs abolished conduction across the channel completely. Time-dependent increase of CV across hMSCs was associated with increased Cx43 mRNA and protein expression resulting in increased gap junctional coupling. Intracellular recordings in coupled hMSCs showed increased conducted action potential (AP) amplitude, lower resting membrane potential, and decreased duration of conducted AP (p<0.05, day 14 versus day 1). CONCLUSIONS: CV across hMSCs increases progressively after 7 days of co-incubation with CMCs, most likely via improved electrotonic interaction. This is associated with increased Cx43 expression, increased functional gap junctional coupling, and enhanced intercellular electrical coupling between hMSCs and CMCs.


Subject(s)
Action Potentials/physiology , Gap Junctions/physiology , Heart Block/physiopathology , Mesenchymal Stem Cells/physiology , Action Potentials/drug effects , Aged , Analysis of Variance , Animals , Animals, Newborn , Carbenoxolone/pharmacology , Chi-Square Distribution , Coculture Techniques , Connexin 43/genetics , Connexin 43/metabolism , Diuretics/pharmacology , Dose-Response Relationship, Drug , Fluorescent Antibody Technique , Gap Junctions/drug effects , Humans , Male , Membrane Potentials/drug effects , Middle Aged , Patch-Clamp Techniques , RNA Interference , RNA, Small Interfering/pharmacology , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Time Factors
20.
J Am Coll Cardiol ; 46(10): 1943-52, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16286184

ABSTRACT

OBJECTIVES: We evaluated whether human adult bone marrow-derived mesenchymal stem cells (hMSCs) could repair an experimentally induced conduction block in cardiomyocyte cultures. BACKGROUND: Autologous stem cell therapy is a novel treatment option for patients with heart disease. However, detailed electrophysiological characterization of hMSCs is still lacking. METHODS: Neonatal rat cardiomyocytes were seeded on multi-electrode arrays. After 48 h, abrasion of a 200- to 450-microm-wide channel caused conduction block. Next, we applied adult hMSCs (hMSC group, n = 8), human skeletal myoblasts (myoblast group, n = 7), rat cardiac fibroblasts (fibroblast group, n = 7), or no cells (control group, n = 7) in a channel-crossing pattern. Cross-channel electrical conduction was analyzed after 24 and 48 h. Intracellular action potentials of hMSCs and cardiomyocytes were recorded. Immunostaining for connexins and intercellular dye transfer (calcein) assessed the presence of functional gap junctions. RESULTS: After creation of conduction block, two asynchronously beating fields of cardiomyocytes were present. Application of hMSCs restored synchronization between the two fields in five of eight cultures after 24 h. Conduction velocity across hMSCs (0.9 +/- 0.4 cm/s) was approximately 11-fold slower than across cardiomyocytes (10.4 +/- 5.8 cm/s). No resynchronization occurred in the myoblast, fibroblast, or control group. Intracellular action potential recordings indicated that conduction across the channel presumably occurred by electrotonic impulse propagation. Connexin-43 was present along regions of hMSC-to-cardiomyocyte contact, but not along regions of cardiomyocyte-to-myoblast or cardiomyocyte-to-fibroblast contact. Calcein transfer from cardiomyocytes to hMSCs was observed within 24 h after co-culture initiation. CONCLUSIONS: Human mesenchymal stem cells are able to repair conduction block in cardiomyocyte cultures, probably through connexin-mediated coupling.


Subject(s)
Bone Marrow Transplantation , Heart Block/surgery , Mesenchymal Stem Cell Transplantation , Myocytes, Cardiac , Animals , Cells, Cultured , Electrophysiology , Heart Block/physiopathology , Humans , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...