Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Front Physiol ; 11: 590, 2020.
Article in English | MEDLINE | ID: mdl-32612537

ABSTRACT

Background: Myocardial infarction (MI) is caused by occlusion of the coronary artery and induces ischemia in the myocardium and eventually a massive loss in cardiomyocytes. Studies have shown many factors or treatments that can affect the healing and remodeling of the heart upon infarction, leading to better cardiac performance and clinical outcome. Previously, miR-132/212 has been shown to play an important role in arteriogenesis in a mouse model of hindlimb ischemia and in the regulation of cardiac contractility in hypertrophic cardiomyopathy in mice. In this study, we explored the role of miR-132/212 during ischemia in a murine MI model. Methods and Results: miR-132/212 knockout mice show enhanced cardiac contractile function at baseline compared to wild-type controls, as assessed by echocardiography. One day after induction of MI by permanent occlusion, miR-132/212 knockout mice display similar levels of cardiac damage as wild-type controls, as demonstrated by infarction size quantification and LDH release, although a trend toward more cardiomyocyte cell death was observed in the knockout mice as shown by TUNEL staining. Four weeks after MI, miR-132/212 knockout mice show no differences in terms of cardiac function, expression of cardiac stress markers, and fibrotic remodeling, although vascularization was reduced. In line with these in vivo observation, overexpression of miR-132 or miR-212 in neonatal rat cardiomyocyte suppress hypoxia induced cardiomyocyte cell death. Conclusion: Although we previously observed a role in collateral formation and myocardial contractility, the absence of miR-132/212 did not affect the overall myocardial performance upon a permanent occlusion of the coronary artery. This suggests an interplay of different roles of this miR-132/212 before and during MI, including an inhibitory effect on cell death and angiogenesis, and a positive effect on cardiac contractility and autophagic response. Thus, spatial or tissue specific manipulation of this microRNA family may be essential to fully understand the roles and to develop interventions to reduce infarct size.

2.
Biotechnol Rep (Amst) ; 18: e00255, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29876304

ABSTRACT

To understand and assess the roles of miRNAs, visualization of the expression patterns of specific miRNAs is needed at the cellular level in a wide variety of different tissue types. Although miRNA in situ hybridization techniques have been greatly improved in recent years, they remain difficult to routinely perform due to the complexity of the procedure. In addition, as it is crucial to define which tissues or cells are expressing a particular miRNA in order to elucidate the biological function of the miRNA, incorporation of additional stainings for different cellular markers is necessary. Here, we describe a robust and flexible multicolor miRNA in situ hybridization (MMISH) technique for paraffin embedded sections. We show that the miRNA in situ protocol is sensitive and highly specific and can successfully be combined with both immunohistochemical and immunofluorescent stainings.

3.
Adv Healthc Mater ; 5(19): 2555-2565, 2016 10.
Article in English | MEDLINE | ID: mdl-27570124

ABSTRACT

To date, cellular transplantation therapy has not yet fulfilled its high expectations for cardiac repair. A major limiting factor is lack of long-term engraftment of the transplanted cells. Interestingly, transplanted cells can positively affect their environment via secreted paracrine factors, among which are extracellular vesicles, including exosomes: small bi-lipid-layered vesicles containing proteins, mRNAs, and miRNAs. An exosome-based therapy will therefore relay a plethora of effects, without some of the limiting factors of cell therapy. Since cardiomyocyte progenitor cells (CMPC) and mesenchymal stem cells (MSC) induce vessel formation and are frequently investigated for cardiac-related therapies, the pro-angiogenic properties of CMPC and MSC-derived exosome-like vesicles are investigated. Both cell types secrete exosome-like vesicles, which are efficiently taken up by endothelial cells. Endothelial cell migration and vessel formation are stimulated by these exosomes in in vitro models, mediated via ERK/Akt-signaling. Additionally, these exosomes stimulated blood vessel formation into matrigel plugs. Analysis of pro-angiogenic factors revealed high levels of extracellular matrix metalloproteinase inducer (EMMPRIN). Knockdown of EMMPRIN on CMPCs leads to a diminished pro-angiogenic effect, both in vitro and in vivo. Therefore, CMPC and MSC exosomes have powerful pro-angiogenic effects, and this effect is largely mediated via the presence of EMMPRIN on exosomes.


Subject(s)
Basigin/metabolism , Exosomes/metabolism , Mesenchymal Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Neovascularization, Pathologic/metabolism , Stem Cells/metabolism , Animals , Cell Movement/physiology , Cell- and Tissue-Based Therapy/methods , Cells, Cultured , Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells , Humans , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Signal Transduction/physiology
4.
J Cardiovasc Transl Res ; 9(4): 291-301, 2016 08.
Article in English | MEDLINE | ID: mdl-27383837

ABSTRACT

Plasma-circulating microRNAs have been implicated as novel early biomarkers for myocardial infarction (MI) due to their high specificity for cardiac injury. For swift clinical translation of this potential biomarker, it is important to understand their temporal and spatial characteristics upon MI. Therefore, we studied the temporal release, potential source, and transportation of circulating miRNAs in different models of ischemia reperfusion (I/R) injury. We demonstrated that extracellular vesicles are released from the ischemic myocardium upon I/R injury. Moreover, we provided evidence that cardiac and muscle-specific miRNAs are transported by extracellular vesicles and are rapidly detectable in plasma. Since these vesicles are enriched for the released miRNAs and their detection precedes traditional damage markers, they hold great potential as specific early biomarkers for MI.


Subject(s)
Extracellular Vesicles/metabolism , MicroRNAs/genetics , Myocardial Infarction/genetics , Myocardial Reperfusion Injury/genetics , Animals , Disease Models, Animal , Female , Genetic Markers , Isolated Heart Preparation , Male , Mice, Inbred C57BL , MicroRNAs/blood , Myocardial Infarction/blood , Myocardial Infarction/diagnosis , Myocardial Reperfusion Injury/blood , Myocardial Reperfusion Injury/diagnosis , Sus scrofa , Time Factors
5.
J Control Release ; 222: 18-31, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26616760

ABSTRACT

Recent developments in microRNA (miRNA) research have identified these as important mediators in the pathophysiological response upon myocardial infarction (MI). Specific miRNAs can inhibit the translation of entire groups of mRNAs, which are involved in specific processes in the pathophysiology after MI, e.g. the fibrotic, apoptotic or angiogenic response. By modulating miRNAs in the heart, these processes can be tuned to improve cardiac function. Antagomirs are effective miRNA-inhibitors, but have a low myocardial specificity and cardiac antagomir treatment therefore requires high doses, which causes side effects. In the present study, ultrasound-triggered microbubble destruction (UTMD) was studied to increase specific delivery of antagomir to the myocardium. Healthy control mice were treated with UTMD and sacrificed at 30min, 24h and 48h, after which antagomir delivery in the heart was analyzed, both qualitatively and quantitatively. Additionally, potential harmful effects of treatment were analyzed by monitoring ECG, analyzing neutrophil invasion and cell death in the heart, and measuring troponin I after treatment. Finally, UTMD was tested for delivery of antagomir in a model of ischemia-reperfusion (I/R) injury. We found that UTMD can significantly increase local antagomir delivery to the non-ischemic heart with modest side-effects like neutrophil invasion without causing apoptosis. Delivered antagomirs enter cardiomyocytes within 30min after treatment and remains there for at least 48h. Interestingly, after I/R injury antagomir already readily enters the infarcted zone and we observed no additional benefit of UTMD for antagomir delivery. This study is the first to explore cardiac antagomir delivery using UTMD. In addition, it is the first to study tissue distribution of short RNA based therapeutics (~22 base pairs) at both the cellular and organ levels after UTMD to the heart in general. In summary, UTMD provides a myocardial delivery strategy for non-vascular permeable cardiac conditions later in the I/R response or chronic conditions like cardiac hypertrophy.


Subject(s)
Microbubbles , Myocardium/metabolism , Oligonucleotides/administration & dosage , Ultrasonic Waves , Animals , Drug Delivery Systems , Male , Mice, Inbred C57BL , MicroRNAs/antagonists & inhibitors , Oligonucleotides/pharmacokinetics , Reperfusion Injury/metabolism
6.
J Mol Med (Berl) ; 91(8): 1001-12, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23625462

ABSTRACT

Instigated by the discovery of adult cardiac progenitor cells, cell replacement therapy has become a promising option for myocardial repair in the past decade. We have previously shown that human-derived cardiomyocyte progenitor cells (hCMPCs) can differentiate into cardiomyocyte-, endothelial-, and smooth muscle-like cells in vitro, and in vivo after transplantation in a mouse model of myocardial infarction, resulting in preservation of cardiac function. However, to allow successful repopulation of the injured myocardium, it is of key importance to restore myocardial perfusion by the formation of new vasculature. Several studies have shown that microRNAs regulate vascular differentiation of different stem/progenitor cells. Here, we show that miR-1 is upregulated in hCMPCs during angiogenic differentiation. Upregulation of miR-1 enhanced the formation of vascular tubes on Matrigel and within a collagen matrix, and also increased hCMPC motility, as shown by planar and transwell migration assays. By western blot, qRT-PCR and luciferase reporter assays, miR-1 was found to directly target and inhibit the expression of sprouty-related EVH1 domain-containing protein 1 (Spred1). Knocking down Spred1 phenocopies the functional effect seen for miR-1 upregulation. Using a systems biology approach, we found that in hCMPCs, miR-1 is proposed to control a network of genes predominantly involved in angiogenesis-related processes, including the Spred1 pathway. Our data shows that by upregulation of miR-1, the angiogenic differentiation of hCMPCs can be enhanced, which may be used as a new therapeutic approach to improve the efficiency of cell-based therapy for cardiac regeneration by enhancing the formation of new vasculature.


Subject(s)
Cell Differentiation/physiology , MicroRNAs/physiology , Neovascularization, Physiologic/physiology , Stem Cells/physiology , Adaptor Proteins, Signal Transducing , Cell Movement , Cells, Cultured , Humans , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Myocytes, Cardiac/cytology , Stem Cells/cytology
7.
J Cell Mol Med ; 16(10): 2379-86, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22348515

ABSTRACT

Undesired cell migration after targeted cell transplantation potentially limits beneficial effects for cardiac regeneration. MicroRNAs are known to be involved in several cellular processes, including cell migration. Here, we attempt to reduce human cardiomyocyte progenitor cell (hCMPC) migration via increasing microRNA-155 (miR-155) levels, and investigate the underlying mechanism. Human cardiomyocyte progenitor cells (hCMPCs) were transfected with pre-miR-155, anti-miR-155 or control-miR (ctrl-miR), followed by scratch- and transwell-assays. These functional assays displayed that miR-155 over-expression efficiently inhibited cell migration by 38 ± 3.6% and 59 ± 3.7% respectively. Conditioned medium from miR-155 transfected cells was collected and zymography analysis showed a significant decrease in MMP-2 and MMP-9 activities. The predicted 3'-UTR of MMP-16, an activator of MMP-2 and -9, was cloned into the pMIR-REPORT vector and luciferase assays were performed. Introduction of miR-155 significantly reduced luciferase activity which could be abolished by cotransfection with anti-miR-155 or target site mutagenesis. By using MMP-16 siRNA to reduce MMP-16 levels or by using an MMP-16 blocking antibody, hCMPC migration could be blocked as well. By directly targeting MMP-16, miR-155 efficiently inhibits cell migration via a reduction in MMP-2 and -9 activities. Our study shows that miR-155 might be used to improve local retention of hCMPCs after intramyocardial delivery.


Subject(s)
Cell Movement , Matrix Metalloproteinase 16/metabolism , MicroRNAs/metabolism , Myocytes, Cardiac/cytology , Stem Cells/metabolism , Blotting, Western , Cell Proliferation , Cells, Cultured , Cloning, Molecular , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Immunohistochemistry , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , MicroRNAs/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transfection
8.
Biomaterials ; 33(6): 1782-90, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22136718

ABSTRACT

Tissue engineering is emerging as a potential therapeutic approach to overcome limitations of cell therapy, like cell retention and survival, as well as to mechanically support the ventricular wall and thereby prevent dilation. Tissue printing technology (TP) offers the possibility to deliver, in a defined and organized manner, scaffolding materials and living cells. The aim of our study was to evaluate the combination of TP, human cardiac-derived cardiomyocyte progenitor cells (hCMPCs) and biomaterials to obtain a construct with cardiogenic potential for in vitro use or in vivo application. With this approach, we were able to generate an in vitro tissue with homogenous distribution of cells in the scaffold. Cell viability was determined after printing and showed that 92% and 89% of cells were viable at 1 and 7 days of culturing, respectively. Moreover, we demonstrated that printed hCMPCs retained their commitment for the cardiac lineage. In particular, we showed that 3D culture enhanced gene expression of the early cardiac transcription factors Nkx2.5, Gata-4 and Mef-2c as well as the sarcomeric protein TroponinT. Printed cells were also able to migrate from the alginate matrix and colonize a matrigel layer, thereby forming tubular-like structures. This indicated that printing can be used for defined cell delivery, while retaining functional properties.


Subject(s)
Myocardium/metabolism , Tissue Engineering/methods , Alginates/chemistry , Biocompatible Materials/chemistry , Biotechnology/methods , Cell Movement , Cell Proliferation , Cell Survival , DNA, Complementary/metabolism , Glucuronic Acid/chemistry , Heart/physiology , Hexuronic Acids/chemistry , Humans , Microscopy, Fluorescence/methods , Myocytes, Cardiac/cytology , Oligopeptides/chemistry , Stem Cells/cytology , Time Factors , Transcription Factors/metabolism , Troponin T/chemistry
9.
J Cell Mol Med ; 15(7): 1474-82, 2011 Jul.
Article in English | MEDLINE | ID: mdl-20550618

ABSTRACT

To improve regeneration of the injured myocardium, cardiomyocyte progenitor cells (CMPCs) have been put forward as a potential cell source for transplantation therapy. Although cell transplantation therapy displayed promising results, many issues need to be addressed before fully appreciating their impact. One of the hurdles is poor graft-cell survival upon injection, thereby limiting potential beneficial effects. Here, we attempt to improve CMPCs survival by increasing microRNA-155 (miR-155) levels, potentially to improve engraftment upon transplantation. Using quantitative PCR, we observed a 4-fold increase of miR-155 when CMPCs were exposed to hydrogen-peroxide stimulation. Flow cytometric analysis of cell viability, apoptosis and necrosis showed that necrosis is the main cause of cell death. Overexpressing miR-155 in CMPCs revealed that miR-155 attenuated necrotic cell death by 40 ± 2.3%via targeting receptor interacting protein 1 (RIP1). In addition, inhibiting RIP1, either by pre-incubating the cells with a RIP1 specific inhibitor, Necrostatin-1 or siRNA mediated knockdown, reduced necrosis by 38 ± 2.5% and 33 ± 1.9%, respectively. Interestingly, analysing gene expression using a PCR-array showed that increased miR-155 levels did not change cell survival and apoptotic related gene expression. By targeting RIP1, miR-155 repressed necrotic cell death of CMPCs, independent of activation of Akt pro-survival pathway. MiR-155 provides the opportunity to block necrosis, a conventionally thought non-regulated process, and might be a potential novel approach to improve cell engraftment for cell therapy.


Subject(s)
Cell Death/physiology , MicroRNAs/metabolism , Myocytes, Cardiac/physiology , Necrosis/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Stem Cells/physiology , Cell Survival , Cells, Cultured , Humans , Imidazoles/metabolism , Indoles/metabolism , MicroRNAs/genetics , Myocytes, Cardiac/cytology , Oxidative Stress , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Signal Transduction/physiology , Stem Cells/cytology
10.
J Cell Mol Med ; 14(4): 861-70, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20219011

ABSTRACT

In the past years, cardiovascular progenitor cells have been isolated from the human heart and characterized. Up to date, no studies have been reported in which the developmental potential of foetal and adult cardiovascular progenitors was tested simultaneously. However, intrinsic differences will likely affect interpretations regarding progenitor cell potential and application for regenerative medicine. Here we report a direct comparison between human foetal and adult heart-derived cardiomyocyte progenitor cells (CMPCs). We show that foetal and adult CMPCs have distinct preferences to differentiate into mesodermal lineages. Under pro-angiogenic conditions, foetal CMPCs form more endothelial but less smooth muscle cells than adult CMPCs. Foetal CMPCs can also develop towards adipocytes, whereas neither foetal nor adult CMPCs show significant osteogenic differentiation. Interestingly, although both cell types differentiate into heart muscle cells, adult CMPCs give rise to electrophysiologically more mature cardiomyocytes than foetal CMPCs. Taken together, foetal CMPCs are suitable for molecular cell biology and developmental studies. The potential of adult CMPCs to form mature cardiomyocytes and smooth muscle cells may be essential for cardiac repair after transplantation into the injured heart.


Subject(s)
Adult Stem Cells/cytology , Fetus/cytology , Myocytes, Cardiac/cytology , Adipogenesis , Adult , Cell Proliferation , Humans , Membrane Potentials/physiology , Neovascularization, Physiologic , Osteogenesis
11.
Arterioscler Thromb Vasc Biol ; 30(4): 859-68, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20081117

ABSTRACT

OBJECTIVE: To improve regeneration of the injured myocardium, it is necessary to enhance the intrinsic capacity of the heart to regenerate itself and/or replace the damaged tissue by cell transplantation. Cardiomyocyte progenitor cells (CMPCs) are a promising cell population, easily expanded and efficiently differentiated into beating cardiomyocytes. Recently, several studies have demonstrated that microRNAs (miRNAs) are important for stem cell maintenance and differentiation via translational repression. We hypothesize that miRNAs are also involved in proliferation/differentiation of the human CMPCs in vitro. METHODS AND RESULTS: Human fetal CMPCs were isolated, cultured, and efficiently differentiated into beating cardiomyocytes. miRNA expression profiling demonstrated that muscle-specific miR-1 and miR-499 were highly upregulated in differentiated cells. Transient transfection of miR-1 and -499 in CMPC reduced proliferation rate by 25% and 15%, respectively, and enhanced differentiation into cardiomyocytes in human CMPCs and embryonic stem cells, likely via the repression of histone deacetylase 4 or Sox6. Histone deacetylase 4 and Sox6 protein levels were reduced, and small interference RNA (siRNA)-mediated knockdown of Sox6 strongly induced myogenic differentiation. CONCLUSIONS: miRNAs regulate the proliferation of human CMPC and their differentiation into cardiomyocytes. By modulating miR-1 and -499 expression levels, human CMPC function can be altered and differentiation directed, thereby enhancing cardiomyogenic differentiation.


Subject(s)
Cell Differentiation/genetics , Cell Proliferation , Fetal Stem Cells/metabolism , Gene Expression Regulation, Developmental , MicroRNAs/metabolism , Muscle Development/genetics , Myocytes, Cardiac/metabolism , Cells, Cultured , Gene Expression Profiling/methods , Gestational Age , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Oligonucleotide Array Sequence Analysis , RNA Interference , Repressor Proteins/genetics , Repressor Proteins/metabolism , SOXD Transcription Factors/genetics , SOXD Transcription Factors/metabolism , Transcription, Genetic , Transfection , Up-Regulation
12.
J Mol Cell Cardiol ; 48(1): 254-60, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19460390

ABSTRACT

Cardiomyocyte progenitor cells (CMPCs) can be isolated from the human heart and differentiated into cardiomyocytes in vitro. A comprehensive assessment of their electrical phenotype upon differentiation is essential to predict potential future applications of this cell source. CMPCs isolated from human fetal heart were differentiated in vitro and examined using immunohistochemistry, Western blotting, RT-PCR, voltage clamp and current clamp techniques. Differentiated cultures presented up to 95% alpha-actinin positive cardiomyocytes. Adherens junction and desmosomal proteins beta-catenin, N-cadherin, desmin and plakophilin2 were upregulated. Expression levels of cardiac connexins were not affected by differentiation, however Cx43 phosphorylation was increased upon differentiation, accompanied by translocation of connexins to the cell border. RT-PCR analysis demonstrated upregulation of all major cardiac ion channel constituents during differentiation. Patch clamp experiments showed that cardiomyocytes had a stable resting membrane potential of -73.4+/-1.8 mV. Infusion of 1 mM BaCl(2) resulted in depolarization to -59.9+/-2.8 mV, indicating I(K1) channel activity. Subsequent voltage clamp experiments confirmed presence of near mature I(Na), I(Ca,L) and I(K1) current densities. Infusion of the I(Kr) blocker Almokalant caused prolongation of the action potential by 40%. Differentiated monolayers were not spontaneously contracting in the absence of serum, but responded to field stimulation, displaying adult ventricular-like action potentials. Human fetal CMPC-derived cardiomyocytes have a homogenous and rather mature electrical phenotype that benefits to in vitro physiology and pharmacology. In the context of cardiac repair, their properties may translate into a reduced pro-arrhythmic risk and enhanced electrical integration upon transplantation.


Subject(s)
Electrophysiology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Stem Cells/cytology , Anti-Arrhythmia Agents/pharmacology , Cell Differentiation/physiology , Cells, Cultured , Heart/drug effects , Heart/physiology , Humans , Ion Channels/metabolism , Membrane Potentials/drug effects , Membrane Potentials/physiology , Myocytes, Cardiac/drug effects , Potassium Channels, Inwardly Rectifying/metabolism , Propanolamines/pharmacology
13.
Stem Cell Res ; 2(2): 108-12, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19383416

ABSTRACT

Development involves an interplay between various cell types from their birth to their disappearance by differentiation, migration, or death. Analyzing these interactions provides insights into their roles during the formation of a new organism. As a study tool for these interactions, we have created a model based on embryoid bodies (EBs) generated from mouse embryonic stem (mES) cells, which can be used to visualize the differentiation of mES cells into specific cell types while at the same time allowing controlled removal of this same cell population using an enzyme-prodrug approach. Cell-specific expression of Cre induces a switch of EGFP expression to LacZ. Furthermore, it leads to the expression of nitroreductase (NTR), which in combination with the prodrug CB1954 induces apoptosis. Here, we validate this model by showing expression of LacZ and NTR after Cre-mediated recombination. Additionally we show, as an example, that we can target the endothelial cells in EBs using the Tie-2 promoter driving Cre. Ablating Cre-expressing cells by adding CB1954 to the culture led to an abrogated vascular formation. This system can easily be adapted to determine the fate and interaction of many different cell types provided that there is a cell-type-specific promoter available.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , Genes, Transgenic, Suicide/genetics , Models, Biological , Animals , Apoptosis/drug effects , Aziridines/administration & dosage , Cell Communication , Cell Culture Techniques/methods , Endothelial Cells/cytology , Genetic Vectors , Green Fluorescent Proteins/genetics , Integrases/genetics , Lac Operon , Mice , Nitroreductases/genetics , Promoter Regions, Genetic , Transfection
14.
Nat Protoc ; 4(2): 232-43, 2009.
Article in English | MEDLINE | ID: mdl-19197267

ABSTRACT

To date, there is no suitable in vitro model to study human adult cardiac cell biology. Although embryonic stem cells are able to differentiate into cardiomyocytes in vitro, the efficiency of this process is very low. Other methods to differentiate progenitor cells into beating cardiomyocytes rely on coculturing with rat neonatal cardiomyocytes, making it difficult to study human cardiomyocyte differentiation and (patho)physiology. Here we have developed a method for efficient isolation and expansion of human cardiomyocyte progenitor cells (CMPCs) from cardiac surgical waste or alternatively from fetal heart tissue. Furthermore, we provide a detailed in vitro protocol for efficient differentiation of CMPCs into cardiomyocytes with great efficiency (80-90% of differentiation). Once CMPCs are rapidly dividing ( approximately 1 month after isolation), differentiation can be achieved in 3-4 weeks.


Subject(s)
Cell Culture Techniques/methods , Heart/physiology , Heart/physiopathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Stem Cells/cytology , Humans , Stem Cells/physiology
15.
J Biol Chem ; 283(31): 21714-24, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18539601

ABSTRACT

The parvulin peptidyl-prolyl isomerase Pin1 catalyzes cis-trans isomerization of p(S/T)-P bonds and might alter conformation and function of client proteins. Since the trans conformation of p(S/T)-P bonds is preferred by protein phosphatase 2A (PP2A), Pin1 may facilitate PP2A-mediated dephosphorylation. Juglone irreversibly inhibits parvulins and is often used to study the function of Pin1 in vivo. The drug prevents dephosphorylation of mitotic phosphoproteins, perhaps because they bind Pin1 and are dephosphorylated by PP2A. We show here however that juglone inhibited post-mitotic dephosphorylation and the exit of mitosis, independent of Pin1. This effect involved covalent modification of sulfhydryl groups in proteins essential for metaphase/anaphase transition. Particularly cytoplasmic proteins with a high cysteine content were vulnerable to the drug. Alkylation of sulfhydryl groups altered the conformation of such proteins, as evidenced by the disappearance of antibody epitopes on tubulin and the mitotic checkpoint component BubR1. The latter activates the anaphase-promoting complex/cyclosome, which degrades regulatory proteins, such as cyclin B1 and securins, and is required for mitotic exit. Indeed, juglone-treated cells failed to assemble a mitotic spindle, which correlated with perturbed microtubule dynamics, loss of immunodetectable tubulin, and formation of tubulin aggregates. Juglone also prevented degradation of cyclin B1, independently of the Mps1-controlled mitotic spindle checkpoint. Since juglone affected cell cycle progression at several levels, more specific drugs need to be developed for studies of Pin1 function in vivo.


Subject(s)
Cysteine/chemistry , Mitosis/drug effects , Naphthoquinones/pharmacology , Animals , CHO Cells , Cattle , Cell Cycle , Cricetinae , Cricetulus , Cyclin B/chemistry , Cyclin B1 , Cytoplasm/metabolism , Enzyme Inhibitors/pharmacology , Fibroblasts/metabolism , Mice , Microtubules/metabolism
16.
Stem Cell Res ; 1(2): 138-49, 2007 Nov.
Article in English | MEDLINE | ID: mdl-19383394

ABSTRACT

The adult mammalian heart has limited regenerative capacity and was generally considered to contain no dividing cells. Recently, however, a resident population of progenitor cells has been identified, which could represent a new source of cardiomyocytes. Here, we describe the efficient isolation and propagation of human cardiomyocyte progenitor cells (hCMPCs) from fetal heart and patient biopsies. Establishment of hCMPC cultures was remarkably reproducible, with over 70% of adult atrial biopsies resulting in robustly expanding cell populations. Following the addition of transforming growth factor beta, almost all cells differentiated into spontaneously beating myocytes with characteristic cross striations. hCMPC-derived cardiomyocytes showed gap-junctional communication and action potentials of maturing cardiomyocytes. These are the first cells isolated from human heart that proliferate and form functional cardiomyocytes without requiring coculture with neonatal myocytes. Their scalability and homogeneity are unique and provide an excellent basis for developing physiological, pharmacological, and toxicological assays on human heart cells in vitro.


Subject(s)
Cell Differentiation/drug effects , Myocytes, Cardiac/cytology , Stem Cells/cytology , Transforming Growth Factor beta1/pharmacology , Cell Culture Techniques , Cell Proliferation , Cell Separation , Humans
17.
Mol Biol Cell ; 16(2): 731-41, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15548590

ABSTRACT

Griscelli syndrome type 2 (GS2) is a genetic disorder in which patients exhibit life-threatening defects of cytotoxic T lymphocytes (CTLs) whose lytic granules fail to dock on the plasma membrane and therefore do not release their contents. The disease is caused by the absence of functional rab27a, but how rab27a controls secretion of lytic granule contents remains elusive. Mutations in Munc13-4 cause familial hemophagocytic lymphohistiocytosis subtype 3 (FHL3), a disease phenotypically related to GS2. We show that Munc13-4 is a direct partner of rab27a. The two proteins are highly expressed in CTLs and mast cells where they colocalize on secretory lysosomes. The region comprising the Munc13 homology domains is essential for the localization of Munc13-4 to secretory lysosomes. The GS2 mutant rab27aW73G strongly reduced binding to Munc13-4, whereas the FHL3 mutant Munc13-4Delta608-611 failed to bind rab27a. Overexpression of Munc13-4 enhanced degranulation of secretory lysosomes in mast cells, showing that it has a positive regulatory role in secretory lysosome fusion. We suggest that the secretion defects seen in GS2 and FHL3 have a common origin, and we propose that the rab27a/Munc13-4 complex is an essential regulator of secretory granule fusion with the plasma membrane in hematopoietic cells. Mutations in either of the two genes prevent formation of this complex and abolish secretion.


Subject(s)
Lysosomes/metabolism , Mast Cells/cytology , Mast Cells/metabolism , Proteins/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Blotting, Western , Cell Line , Glutathione Transferase/metabolism , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Immunohistochemistry , Jurkat Cells , K562 Cells , Mast Cells/ultrastructure , Microscopy, Immunoelectron , Mutation , Protein Binding , Protein Structure, Tertiary , Proteins/chemistry , Proteins/genetics , Proteins/ultrastructure , Rats , Recombinant Proteins/metabolism , Sulfur Radioisotopes/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Transfection , U937 Cells , rab GTP-Binding Proteins/chemistry , rab GTP-Binding Proteins/genetics , rab27 GTP-Binding Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...