Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Control Release ; 369: 734-745, 2024 May.
Article in English | MEDLINE | ID: mdl-38604385

ABSTRACT

Despite research efforts being made towards preserving (or even regenerating) heart tissue after an ischemic event, there is a lack of resources in current clinical treatment modalities for patients with acute myocardial infarction that specifically address cardiac tissue impairment. Modified messenger RNA (modRNA) presents compelling properties that could allow new therapeutic strategies to tackle the underlying molecular pathways that ultimately lead to development of chronic heart failure. However, clinical application of modRNA for the heart is challenged by the lack of effective and safe delivery systems. Lipid nanoparticles (LNPs) represent a well characterized class of RNA delivery systems, which were recently approved for clinical usage in mRNA-based COVID-19 vaccines. In this study, we evaluated the potential of LNPs for cardiac delivery of modRNA. We tested how variations in C12-200 modRNA-LNP composition affect transfection levels and biodistribution after intramyocardial administration in both healthy and myocardial-infarcted mice, and determined the targeted cardiac cell types. Our data revealed that LNP-mediated modRNA delivery outperforms the current state of the art (modRNA in citrate buffer) upon intramyocardial administration in mice, with only minor differences among the formulations tested. Furthermore, we determined both in vitro and in vivo that the cardiac cells targeted by modRNA-LNPs include fibroblasts, endothelial cells and epicardial cells, suggesting that these cell types could represent targets for therapeutic interference with these LNP formulations. These outcomes may serve as a starting point for LNP development specifically for therapeutic mRNA cardiac delivery applications.


Subject(s)
Mice, Inbred C57BL , Myocardial Infarction , Myocardium , Nanoparticles , RNA, Messenger , Animals , RNA, Messenger/administration & dosage , Tissue Distribution , Myocardial Infarction/therapy , Myocardium/metabolism , Lipids/chemistry , Mice , Humans , Male , Gene Transfer Techniques , Transfection/methods , Liposomes
2.
Eur J Pharm Biopharm ; 170: 59-69, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34864197

ABSTRACT

Extracellular vesicles (EVs) are phospholipid bilayer enclosed vesicles which play an important role in intercellular communication. To date, many studies have focused on therapeutic application of EVs. However, to progress EV applications faster towards the clinic, more information about the physical stability and scalable production of EVs is needed. The goal of this study was to evaluate EV recovery and function after varying several conditions in the isolation process or during storage. Physical stability and recovery rates of EVs were evaluated by measuring EV size, particle and protein yields using nanoparticle tracking analysis, microBCA protein quantification assay and transmission electron microscopy. Western blot analyses of specific EV markers were performed to determine EV yields and purity. EV functionality was tested in an endothelial cell wound healing assay. Higher EV recovery rates were found when using HEPES buffered saline (HBS) as buffer compared to phosphate buffered saline (PBS) during EV isolation. When concentrating EVs, 15 ml spinfilters with a 10 kDa membrane cutoff gave the highest EV recovery. Next, EV storage in polypropylene tubes was shown to be superior compared to glass tubes. The use of protective excipients during EV storage, i.e. bovine serum albumin (BSA) and Tween 20, improved EV preservation without influencing their functionality. Finally, it was shown that both 4 °C and -80 °C are suitable for short term storage of EVs. Together, our results indicate that optimizing buffer compositions, concentrating steps, protective excipients and storage properties may collectively increase EV recovery rates significantly while preserving their functional properties, which accelerates translation of EV-based therapeutics towards clinical application.


Subject(s)
Extracellular Vesicles , Organ Preservation/methods , Cell Tracking , Cells, Cultured , Excipients/pharmacology , Nanoparticles , Organ Preservation Solutions/pharmacology , Temperature
3.
Neth Heart J ; 29(7-8): 409-414, 2021 Jul.
Article in English | MEDLINE | ID: mdl-34114177

ABSTRACT

AIMS: The CardioMEMS HF system is used to measure pulmonary artery (PA) pressures of patients with heart failure (HF). The goal of this study was to determine the impact of time in the daily PA pressure measurements, considering variance and influence of circadian rhythms on cardiovascular pathophysiology. METHODS AND RESULTS: The study included 10 patients with HF with reduced ejection fraction (LVEF < 40%; New York Heart Association class III). Individual daily PA pressures were obtained by CardioMEMS sensors, per protocol, measured up to six times throughout the day, for a period of 5 days. Differences between variation of morning versus evening PA pressures were compared with Wilcoxon signed-rank test. Mean PA pressures (mPAP) increased from a morning value of 19.1 ± 2 mm Hg (8 am; mean ± standard error of the mean [SEM]) to 21.3 ± 2 mm Hg late in the evening (11 pm; mean ± SEM). Over the course of 5 days, evening mPAP exhibited a significantly higher median coefficient of variation than morning mPAP (14.9 (interquartile range [IQR] 7.6-21.0) and 7.0 (IQR 5.0-12.8) respectively; p = 0.01). The same daily pattern of pressure variability was observed in diastolic (p = 0.01) and systolic (p = 0.04) pressures, with diastolic pressures being more variable than systolic at all time points. CONCLUSIONS: Morning PA pressure measurements yield more stable values for observing PA trends. Patients should thus be advised to consistently perform their daily PA pressure measurements early in the morning. This will improve reliability and interpretation of the CardioMEMS management, indicating true alterations in the patient's health status, rather than time-of-day-dependent variations.

4.
Heliyon ; 4(6): e00642, 2018 Jun.
Article in English | MEDLINE | ID: mdl-30003150

ABSTRACT

Adverse remodeling after myocardial infarction (MI) is strongly influenced by T cells. Stem cell therapy after MI, using mesenchymal stem cells (MSC) or cardiomyocyte progenitor cells (CMPC), improved cardiac function, despite low cell retention and limited differentiation. As MSC secrete many factors affecting T cell proliferation and function, we hypothesized the immune response could be affected as one of the targets of stem cell therapy. Therefore, we studied the immunosuppressive properties of human BM-MSC and CMPC and their extracellular vesicles (EVs) in co-culture with activated T cells. Proliferation of T cells, measured by carboxyfluorescein succinimidyl ester dilution, was significantly reduced in the presence of BM-MSC and CMPC. The inflammatory cytokine panel of the T cells in co-culture, measured by Luminex assay, changed, with strong downregulation of IFN-gamma and TNF-alpha. The effect on proliferation was observed in both direct cell contact and transwell co-culture systems. Transfer of conditioned medium to unrelated T cells abrogated proliferation in these cells. EVs isolated from the conditioned medium of BM-MSC and CMPC prevented T cell proliferation in a dose-dependent fashion. Progenitor cells presence induces up- and downregulation of multiple previously unreported pathways in T cells. In conclusion, both BM-MSC and CMPC have a strong capacity for in vitro immunosuppression. This effect is mediated by paracrine factors, such as extracellular vesicles. Besides proliferation, many additional pathways are influenced by both BM-MSC and CMPC.

5.
Sci Rep ; 8(1): 6047, 2018 Apr 11.
Article in English | MEDLINE | ID: mdl-29643426

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

6.
Sci Rep ; 7(1): 14218, 2017 10 27.
Article in English | MEDLINE | ID: mdl-29079786

ABSTRACT

Large animal models are essential for the development of novel therapeutics for myocardial infarction. To optimize translation, we need to assess the effect of experimental design on disease outcome and model experimental design to resemble the clinical course of MI. The aim of this study is therefore to systematically investigate how experimental decisions affect outcome measurements in large animal MI models. We used control animal-data from two independent meta-analyses of large animal MI models. All variables of interest were pre-defined. We performed univariable and multivariable meta-regression to analyze whether these variables influenced infarct size and ejection fraction. Our analyses incorporated 246 relevant studies. Multivariable meta-regression revealed that infarct size and cardiac function were influenced independently by choice of species, sex, co-medication, occlusion type, occluded vessel, quantification method, ischemia duration and follow-up duration. We provide strong systematic evidence that commonly used endpoints significantly depend on study design and biological variation. This makes direct comparison of different study-results difficult and calls for standardized models. Researchers should take this into account when designing large animal studies to most closely mimic the clinical course of MI and enable translational success.


Subject(s)
Disease Models, Animal , Myocardial Infarction , Animals , Myocardial Infarction/mortality , Regression Analysis
7.
Adv Exp Med Biol ; 998: 207-219, 2017.
Article in English | MEDLINE | ID: mdl-28936742

ABSTRACT

Cardiac progenitor cells (CPCs) have emerged as potential therapy to improve cardiac repair and prevent damage in cardiac diseases. CPCs are a promising cell source for cardiac therapy as they can generate all cardiovascular lineages in vitro and in vivo. Originating from the heart itself, CPCs may be destined to activate endogenous repair mechanisms. These CPCs release paracrine molecules that are able to stimulate cardiac repair mechanisms, including stimulation of vessel formation and inhibition of cardiomyocyte apoptosis. In addition to proteins and growth factors, CPCs release extracellular membrane vesicles, such as exosomes, which have gained increasing interest in recent years. Exosomal-derived miRNAs have been indicated to play an important role in these processes. Hereby, CPC exosomes can be considered as potential off-the-shelf therapeutics, as they are able to stimulate the regenerative capacity of the heart by increasing vessel density and lowering apoptosis of cardiomyocytes.


Subject(s)
Cardiovascular Diseases/surgery , Cell-Free System/transplantation , Exosomes/transplantation , Myocardium/pathology , Myocytes, Cardiac/pathology , Regeneration , Stem Cells/pathology , Animals , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Cell-Free System/metabolism , Cell-Free System/pathology , Exosomes/genetics , Exosomes/metabolism , Exosomes/pathology , Gene Expression Regulation , Humans , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Paracrine Communication , Recovery of Function , Signal Transduction , Stem Cells/metabolism
8.
J Cardiovasc Transl Res ; 8(7): 393-403, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26382088

ABSTRACT

Cardiac cell therapy is a strategy to treat patients with chronic myocardial infarction (MI). No consensus exists regarding the optimal cell type. First, a comparison between autologous bone marrow-derived mononuclear cells (BMMNC) and mesenchymal stem cells (MSC) on therapeutic efficacy after MI was performed. Next, the effect of repetitive, NOGA-guided transendocardial injection was determined via a crossover design. Nineteen pigs were allocated in three groups: (1) placebo (at 4 and 8 weeks), (2) MSC (followed by placebo at 8 weeks), or (3) BMMNC (followed by MSC at 8 weeks) delivery including a priming strategy to enhance MSC effect. At 4 weeks, ejection fraction (EF) was significantly improved after MSC injection and not by BMMNC injection. After 8 weeks, no difference was observed in EF between cell-treated groups demonstrating the positive systolic effect of MSC. This study showed that MSC rather than BMMNC injection improves systolic function in chronic MI.


Subject(s)
Bone Marrow Transplantation , Mesenchymal Stem Cell Transplantation , Myocardial Infarction/surgery , Anesthesia, Intravenous , Animals , Bone Marrow Transplantation/methods , Cells, Cultured , Chronic Disease , Disease Models, Animal , Echocardiography , Female , Mesenchymal Stem Cell Transplantation/methods , Myocardial Infarction/physiopathology , Premedication , Stroke Volume , Swine , Systole/physiology , Transplantation, Autologous
9.
Curr Genomics ; 16(2): 88-94, 2015 Apr.
Article in English | MEDLINE | ID: mdl-26085807

ABSTRACT

Myocarditis is an inflammatory disease of the heart, which can persist over a long time. During this time, known as the chronic phase of myocarditis, ongoing inflammation damages the cardiomyocytes. The loss of cardiac cells culminates in the development of dilated cardiomyopathy, often followed by non-ischemic heart failure due to diminished cardiac function. During the course of the disease, expression levels of non-coding small RNAs, called microRNAs (miRNAs), change. Although mainly studied in the acute setting, some of these changes in expression level appear to persist in the chronic phase. In addition to being a much-needed diagnostic tool, these miRNA could provide new treatment options. miRNA-based intervention strategies already showed promising results in the treatment of ischemic cardiovascular diseases in preclinical animal models. By implementing more knowledge on the role of miRNAs in the progression towards heart failure, this can potentially be used in the development of miRNA-based therapeutic interventions in the treatment of myocarditis and thereby preventing the progression towards heart failure. The first part of this review will focus on the natural course of myocarditis and the progression towards heart failure. Secondly, we will discuss the current knowledge on alterations of miRNA expression patterns, and suggest some possible future interventions.

10.
Neth Heart J ; 22(11): 493-500, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25331760

ABSTRACT

BACKGROUND: Intramyocardial cell injections in the context of cardiac regenerative therapy can currently be performed using electromechanical mapping (EMM) provided by the NOGA®XP catheter injection system. The gold standard technique to determine infarct size and location, however, is late gadolinium enhanced magnetic resonance imaging (LGE-MRI). In this article we describe a practical and accurate technique to co-register LGE-MRI and NOGA®XP datasets during the injection procedures to ultimately perform image-guided injections to the border zone of the infarct determined by LGE-MRI. MATERIALS AND METHODS: LGE-MRI and EMM were obtained in three pigs with chronic myocardial infarction. MRI and EMM datasets were registered using the in-house developed 3D CartBox image registration toolbox consisting of three steps: 1) landmark registration, 2) surface registration, and 3) manual optimization. The apex and the coronary ostia were used as landmarks. RESULTS: Image registration was successful in all datasets, and resulted in a mean registration error of 3.22 ± 1.86 mm between the MRI surface mesh and EMM points. Visual assessment revealed that the locations and the transmural extent of the infarctions measured by LGE-MRI only partly overlap with the infarct areas identified by the EMM parameters. CONCLUSIONS: The 3D CartBox image registration toolbox enables registration of EMM on pre-procedurally acquired MRI during the catheter injection procedure. This allows the operator to perform real-time image-guided cell injections into the border zone of the infarct as assessed by LGE-MRI. The 3D CartBox thereby enables, for the first time, standardisation of the injection location for cardiac regenerative therapy.

11.
J Cardiovasc Transl Res ; 6(6): 884-98, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23897095

ABSTRACT

Small non-coding microRNAs (miRNAs) are important physiological regulators of post-transcriptional gene expression. miRNAs not only reside in the cytoplasm but are also stably present in several extracellular compartments, including the circulation. For that reason, miRNAs are proposed as diagnostic biomarkers for various diseases. Early diagnosis of acute coronary syndrome (ACS), especially non-ST elevated myocardial infarction and unstable angina pectoris, is essential for optimal treatment outcome, and due to the ongoing need for additional identifiers, miRNAs are of special interest as biomarkers for ACS. This review highlights the nature and cellular release mechanisms of circulating miRNAs and therefore their potential role in the diagnosis of myocardial infarction. We will give an update of clinical studies addressing the role of circulating miRNA expression after myocardial infarction and explore the diagnostic value of this potential biomarker.


Subject(s)
Acute Coronary Syndrome/diagnosis , MicroRNAs/blood , Myocardial Infarction/diagnosis , Acute Coronary Syndrome/blood , Acute Coronary Syndrome/genetics , Animals , Early Diagnosis , Gene Expression Regulation , Genetic Markers , Humans , Myocardial Infarction/blood , Myocardial Infarction/genetics , Myocardium/metabolism , Predictive Value of Tests , Prognosis
12.
Biochim Biophys Acta ; 1830(2): 2449-58, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22975401

ABSTRACT

BACKGROUND: After myocardial infarction (MI) a local inflammatory reaction clears the damaged myocardium from dead cells and matrix debris at the onset of scar formation. The intensity and duration of this inflammatory reaction are intimately linked to post-infarct remodeling and cardiac dysfunction. Strikingly, treatment with standard anti-inflammatory drugs worsens clinical outcome, suggesting a dual role of inflammation in the cardiac response to injury. Cardiac stem cell therapy with different stem or progenitor cells, e.g. mesenchymal stem cells (MSC), was recently found to have beneficial effects, mostly related to paracrine actions. One of the suggested paracrine effects of cell therapy is modulation of the immune system. SCOPE OF REVIEW: MSC are reported to interact with several cells of the immune system and could therefore be an excellent means to reduce detrimental inflammatory reactions and promote the switch to the healing phase upon cardiac injury. This review focuses on the potential use of MSC therapy for post-MI inflammation. To understand the effects MSC might have on the post-MI heart the cellular and molecular changes in the myocardium after MI need to be understood. MAJOR CONCLUSIONS: By studying the general pathways involved in immunomodulation, and examining the interactions with cell types important for post-MI inflammation, it becomes clear that MSC treatment might provide a new therapeutic opportunity to improve cardiac outcome after acute injury. GENERAL SIGNIFICANCE: Using stem cells to target the post-MI inflammation is a novel therapy which could have considerable clinical implications. This article is part of a Special Issue entitled Biochemistry of Stem Cells.


Subject(s)
Inflammation/therapy , Mesenchymal Stem Cell Transplantation , Myocardial Infarction/therapy , Humans
13.
Mediators Inflamm ; 2013: 181020, 2013.
Article in English | MEDLINE | ID: mdl-24391353

ABSTRACT

BACKGROUND: After myocardial infarction (MI), the inflammatory response is indispensable for initiating reparatory processes. However, the intensity and duration of the inflammation cause additional damage to the already injured myocardium. Treatment with mesenchymal stem cells (MSC) upon MI positively affects cardiac function. This happens likely via a paracrine mechanism. As MSC are potent modulators of the immune system, this could influence this postinfarct immune response. Since MSC express toll-like receptors (TLR), danger signal (DAMP) produced after MI could influence their immunomodulatory properties. SCOPE OF REVIEW: Not much is known about the direct immunomodulatory efficiency of MSC when injected in a strong inflammatory environment. This review focuses first on the interactions between MSC and the immune system. Subsequently, an overview is provided of the effects of DAMP-associated TLR activation on MSC and their immunomodulative properties after myocardial infarction. MAJOR CONCLUSIONS: MSC can strongly influence most cell types of the immune system. TLR signaling can increase and decrease this immunomodulatory potential, depending on the available ligands. Although reports are inconsistent, TLR3 activation may boost immunomodulation by MSC, while TLR4 activation suppresses it. GENERAL SIGNIFICANCE: Elucidating the effects of TLR activation on MSC could identify new preconditioning strategies which might improve their immunomodulative properties.


Subject(s)
Inflammation/metabolism , Mesenchymal Stem Cells/cytology , Myocardium/metabolism , Stem Cell Transplantation , Toll-Like Receptors/metabolism , Animals , B-Lymphocytes/cytology , Cell Differentiation , Cell Movement , Cell Proliferation , Humans , Immunomodulation , Ligands , Macrophages/cytology , Mice , Monocytes/cytology , Myocardial Infarction/pathology , Neutrophils/cytology , Signal Transduction , T-Lymphocytes/cytology , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4/metabolism
14.
J Cell Mol Med ; 16(8): 1827-39, 2012 Aug.
Article in English | MEDLINE | ID: mdl-21973026

ABSTRACT

Although mesenchymal stromal cells (MSCs) have been applied clinically to treat cardiac diseases, it is unclear how and to which extent transplanted MSCs exert their beneficial effects. To address these questions, pre-clinical MSC administrations are needed for which pigs appear to be the species of choice. This requires the use of porcine cells to prevent immune rejection. However, it is currently unknown to what extent porcine MSCs (pMSCs) resemble human MSCs (hMSCs). Aim of this study was to compare MSC from porcine bone marrow (BM) with human cells for phenotype, multi-lineage differentiation potential, immune-modulatory capacity and the effect on cardiac function after transplantation in a mouse model of myocardial infarction. Flow cytometric analysis revealed that pMSC expressed surface antigens also found on hMSC, including CD90, MSCA-1 (TNAP/W8B2 antigen), CD44, CD29 and SLA class I. Clonogenic outgrowth was significantly enriched following selection of CD271+ cells from BM of human and pig (129 ± 29 and 1961 ± 485 fold, respectively). hMSC and pMSC differentiated comparably into the adipogenic, osteogenic or chondrogenic lineages, although pMSC formed fat much faster than hMSC. Immuno-modulation, an important feature of hMSC, was clearly demonstrated for pMSC when co-cultured with porcine peripheral blood cells stimulated with PMA and pIL-2. Finally, pMSC transplantation after myocardial infarction attenuated adverse remodelling to a similar extent as hMSC when compared to control saline injection. These findings demonstrate that pMSCs have comparable characteristics and functionality with hMSCs, making reliable extrapolation of pre-clinical pMSC studies into a clinical setting very well possible.


Subject(s)
Cell Differentiation , Immunomodulation/immunology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Myocardium/pathology , Adipocytes/cytology , Adipocytes/metabolism , Adipogenesis , Animals , Antigens, CD/metabolism , Bone Marrow Cells/cytology , Cell Proliferation , Cell Separation , Chondrogenesis , Flow Cytometry , Heart Function Tests , Humans , Immunophenotyping , Male , Mice , Mice, SCID , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis , Phenotype , Sus scrofa , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
15.
J Cell Mol Med ; 14(5): 1064-70, 2010 May.
Article in English | MEDLINE | ID: mdl-20465578

ABSTRACT

Patients suffering from heart failure as a result of myocardial infarction are in need of heart transplantation. Unfortunately the number of donor hearts is very low and therefore new therapies are subject of investigation. Cell transplantation therapy upon myocardial infarction is a very promising strategy to replace the dead myocardium with viable cardiomyocytes, smooth muscle cells and endothelial cells, thereby reducing scarring and improving cardiac performance. Despite promising results, resulting in reduced infarct size and improved cardiac function on short term, only a few cells survive the ischemic milieu and are retained in the heart, thereby minimizing long-term effects. Although new capillaries and cardiomyocytes are formed around the infarcted area, only a small percentage of the transplanted cells can be detected months after myocardial infarction. This suggests the stimulation of an endogenous regenerative capacity of the heart upon cell transplantation, resulting from release of growth factor, cytokine and other paracrine molecules by the progenitor cells--the so-called paracrine hypothesis. Here, we focus on a relative new component of paracrine signalling, i.e. exosomes. We are interested in the release and function of exosomes derived from cardiac progenitor cells and studied their effects on the migratory capacity of endothelial cells.


Subject(s)
Cell Movement , Endothelial Cells/cytology , Exosomes/metabolism , Myocytes, Cardiac/cytology , Stem Cells/metabolism , Basigin/metabolism , Cell Movement/drug effects , Culture Media, Conditioned/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Exosomes/drug effects , Humans , Matrix Metalloproteinases/metabolism , Models, Biological , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Signal Transduction/drug effects , Stem Cell Transplantation , Stem Cells/cytology , Stem Cells/drug effects
16.
Panminerva Med ; 52(1): 27-40, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20228724

ABSTRACT

Following myocardial infarction, damage due to ischemia potentially leads to heart failure. Stem cell transplantation has emerged as a potential treatment to repair the injured heart, due to the inherent characteristics of stem cells such as self-renewal, unlimited capacity for proliferation and ability to differentiate to various cell lineages. Most promising results have been reported thus far on mesenchymal stem cells (MSC). Following transplantation in the heart, stem cells are expected to 1) reduce the damage; 2) activate the endogenous regenerative potential of the heart; and 3) participate in the regeneration of the tissue. Until now, the results of intervention with stem cells in animals were promising, but clinical studies have failed to live up to those expectations. Current problems limiting the efficacy of cellular therapy are: 1) limited knowledge on the time and mode of administration; 2) loss of homing receptors on culture-expanded cells as a consequence of the culture conditions; 3) massive cell death in the transplanted graft in the damaged heart, due to the hostile environment, 4) lack of knowledge on MSC behaviour in the heart. Since generally only 1-5% of delivered cells were found to actually engraft within the infarct zone, there is an urgent need for improvement. In animal models, strategies to precondition MSC before transplantation to survive in the damaged heart were applied successfully. These include exposure of cells to physical treatments (hypoxia and heat shock), pharmacological agents, "priming" of cells with growth factors, and genetic modification by over-expression of anti-apoptotic proteins, growth factors or pro-survival genes. To develop the strategy with maximal engraftment, survival and function of cells in the heart is the ultimate challenge for years to come.


Subject(s)
Cardiovascular Diseases/therapy , Mesenchymal Stem Cells/cytology , Stromal Cells/cytology , Cell Differentiation , Cell Transplantation , Humans , Regenerative Medicine , Survival Rate
17.
Curr Opin Organ Transplant ; 14(5): 560-5, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19623073

ABSTRACT

PURPOSE OF REVIEW: For heart failure patients, the urgent need for heart transplantation exceeds the availability of donor hearts. Therefore, cell transplantation has emerged as an interesting and potential solution. This review will focus on the capability of different types of stem cells to regenerate the heart. Moreover, the mechanism for success will be addressed, focusing on the specific (and indispensable?) role of the cells. RECENT FINDINGS: In recent years, many types of stem cells have been described as a possible source for cell transplantation in failing hearts, with mixed outcomes. Cell transplantation is hampered by suboptimal delivery techniques, limited survival of cells, and reduced proliferation and differentiation rates in vivo. Interestingly, the number of injected cells that engrafted the heart successfully cannot explain the observed beneficial effects and, therefore, paracrine effects are suggested for the success in cell therapy. SUMMARY: This review summarizes the current types of stem or progenitor cells used in cardiac cell therapy and beneficial effects on heart function and morphology in preclinical studies. Currently, the observed effects suggest that paracrine effects might be responsible, thereby triggering mobilization and activation of resident (stem) cells, which challenges the classical concept and true regenerative capacity of cell therapy at this point.


Subject(s)
Heart Failure/surgery , Stem Cell Transplantation/methods , Humans , Treatment Outcome
18.
Neth Heart J ; 16(5): 163-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18566670

ABSTRACT

BACKGROUND: In recent years, resident cardiac progenitor cells have been identified in, and isolated from the rodent heart. These cells show the potential to form cardiomyocytes, smooth muscle cells, and endothelial cells in vitro and in vivo and could potentially be used as a source for cardiac repair. However, previously described cardiac progenitor cell populations show immature development and need co-culture with neonatal rat cardiomyocytes in order to differentiate in vitro. Here we describe the localisation, isolation, characterisation, and differentiation of cardiomyocyte progenitor cells (CMPCs) isolated from the human heart. METHODS: hCMPCs were identified in human hearts based on Sca-1 expression. These cells were isolated, and FACS, RT-PCR and immunocytochemistry were used to determine their baseline characteristics. Cardiomyogenic differentiation was induced by stimulation with 5-azacytidine. RESULTS: hCMPCs were localised within the atria, atrioventricular region, and epicardial layer of the foetal and adult human heart. In vitro, hCMPCs could be induced to differentiate into cardiomyocytes and formed spontaneously beating aggregates, without the need for co-culture with neonatal cardiomyocytes. CONCLUSION: The human heart harbours a pool of resident cardiomyocyte progenitor cells, which can be expanded and differentiated in vitro. These cells may provide a suitable source for cardiac regeneration cell therapy. (Neth Heart J 2008;16:163-9.).

19.
Panminerva Med ; 50(1): 19-30, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18427385

ABSTRACT

Cell-based cardiac repair has the ambitious aim to replace the malfunctioning cardiac muscle developed after myocardial infarction, with new contractile cardiomyocytes and vessels. Different stem cell populations have been intensively studied in the last decade as a potential source of new cardiomyocytes to ameliorate the injured myocardium, compensate for the loss of ventricular mass and contractility and eventually restore cardiac function. An array of cell types has been explored in this respect, including skeletal muscle, bone marrow derived stem cells, embryonic stem cells (ESC) and more recently cardiac progenitor cells. The best-studied cell types are mouse and human ESC cells, which have undisputedly been demonstrated to differentiate into cardiomyocyte and vascular lineages and have been of great help to understand the differentiation process of pluripotent cells. However, due to their immunogenicity, risk of tumor development and the ethical challenge arising from their embryonic origin, they do not provide a suitable cell source for a regenerative therapy approach. A better option, overcoming ethical and allogenicity problems, seems to be provided by bone marrow derived cells and by the recently identified cardiac precursors. This report will overview current knowledge on these different cell types and their application in cardiac regeneration and address issues like implementation of delivery methods, including tissue engineering approaches that need to be developed alongside.


Subject(s)
Heart/physiology , Myocardial Infarction/therapy , Regeneration , Stem Cell Transplantation/methods , Animals , Bone Marrow Cells/physiology , Clinical Trials as Topic , Cytokines/therapeutic use , Embryonic Stem Cells/transplantation , Humans , Myocytes, Cardiac/cytology , Myocytes, Cardiac/transplantation , Pluripotent Stem Cells/transplantation , Tissue Engineering
20.
Atherosclerosis ; 197(1): 95-104, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17888930

ABSTRACT

BACKGROUND: Toll like receptors (TLR) have been recognized for their role in atherosclerotic lesion development and progression. Endogenous TLR ligands that are also expressed in atherosclerotic tissues have been shown to promote atherosclerosis in mice. Since repetitive stimulation of TLR induces an attenuated inflammatory response, we hypothesized that the TLR response is altered during atherosclerosis development, due to chronic exposure to endogenous ligands. METHODS AND RESULTS: We examined five groups of both ApoE-/- and C57Bl/6 mice aged 5, 10, 15, 25 and 40 weeks. In ApoE-/- mice with advanced stages of atherosclerosis, levels of mRNA encoding TLR2 and TLR4, the endogenous TLR ligands EDA and hsp60 as well as intracellular TLR-regulating mediators, like IRAK-M, were increased. Systemic TLR cell surface expression on circulating monocytes and EDA plasma levels were significantly increased in ApoE-/- mice with advanced atherosclerosis. We also observed that the endogenous TLR ligand EDA was capable of activating the TLR-signaling pathway in white blood cells. During the plaque progression stage however, stimulation of TLR2 and TLR4 in blood samples attenuated MIP-1 alpha and RANTES release in atherosclerotic mice. CONCLUSION: During atherosclerotic lesion development, TLR2 and TLR4 expression increases in atherosclerotic plaques and on circulating blood cells. However, with advanced stages of atherosclerotic disease, circulating blood cells become less responsive to TLR ligation, which may be due to chronic TLR engagement by endogenous EDA.


Subject(s)
Atherosclerosis/metabolism , Atherosclerosis/pathology , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Animals , Aorta, Thoracic/physiology , Apolipoproteins E/genetics , Atherosclerosis/immunology , Disease Progression , Fibronectins/blood , Fibronectins/chemistry , Gene Expression/immunology , Ligands , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Monocytes/metabolism , Protein Structure, Tertiary , RNA, Messenger/metabolism , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics , Vasculitis/immunology , Vasculitis/metabolism , Vasculitis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...