1
|
Neef K, Drey F, Lepperhof V, Wahlers T, Hescheler J, Choi YH, Šarić T. Co-transplantation of Mesenchymal Stromal Cells and Induced Pluripotent Stem Cell-Derived Cardiomyocytes Improves Cardiac Function After Myocardial Damage. Front Cardiovasc Med 2022; 8:794690. [PMID: 35071360 PMCID: PMC8770928 DOI: 10.3389/fcvm.2021.794690] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 11/22/2021] [Indexed: 01/01/2023] Open
Abstract
Induced pluripotent stem cell-derived cardiomyocytes (iPS-CMs) represent an attractive resource for cardiac regeneration. However, survival and functional integration of transplanted iPS-CM is poor and remains a major challenge for the development of effective therapies. We hypothesized that paracrine effects of co-transplanted mesenchymal stromal cells (MSCs) augment the retention and therapeutic efficacy of iPS-CM in a mouse model of myocardial infarction (MI). To test this, either iPS-CM, MSC, or both cell types were transplanted into the cryoinfarction border zone of syngeneic mice immediately after injury. Bioluminescence imaging (BLI) of iPS-CM did not confirm enhanced retention by co-application of MSC during the 28-day follow-up period. However, histological analyses of hearts 28 days after cell transplantation showed that MSC increased the fraction of animals with detectable iPS-CM by 2-fold. Cardiac MRI analyses showed that from day 14 after transplantation on, the animals that have received cells had a significantly higher left ventricular ejection fraction (LVEF) compared to the placebo group. There was no statistically significant difference in LVEF between animals transplanted only with iPS-CM or only with MSC. However, combined iPS-CM and MSC transplantation resulted in higher LVEF compared to transplantation of single-cell populations during the whole observation period. Histological analyses revealed that MSC increased the capillarization in the myocardium when transplanted alone or with iPS-CM and decreased the infarct scar area only when transplanted in combination with iPS-CM. These results indicate that co-transplantation of iPS-CM and MSC improves cardiac regeneration after cardiac damage, demonstrating the potential of combining multiple cell types for increasing the efficacy of future cardiac cell therapies.
Collapse
Affiliation(s)
- Klaus Neef
- Department of Cardiac and Thoracic Surgery, Heart Center, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Florian Drey
- Department of Cardiac and Thoracic Surgery, Heart Center, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Vera Lepperhof
- Institute for Neurophysiology, Center for Physiology and Pathophysiology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Thorsten Wahlers
- Department of Cardiac and Thoracic Surgery, Heart Center, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Jürgen Hescheler
- Institute for Neurophysiology, Center for Physiology and Pathophysiology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Yeong-Hoon Choi
- Department of Cardiac and Thoracic Surgery, Heart Center, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- Clinic for Cardiac Surgery and Surgical Intensive Care Medicine, Kerckhoff Clinic Bad Nauheim, Kerckhoff Campus, Justus Liebig University Giessen, Giessen, Germany
| | - Tomo Šarić
- Institute for Neurophysiology, Center for Physiology and Pathophysiology, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
- *Correspondence: Tomo Šarić
| |
Collapse
|
2
|
Friedrich RP, Cicha I, Alexiou C. Iron Oxide Nanoparticles in Regenerative Medicine and Tissue Engineering. NANOMATERIALS 2021; 11:nano11092337. [PMID: 34578651 PMCID: PMC8466586 DOI: 10.3390/nano11092337] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/03/2021] [Accepted: 09/06/2021] [Indexed: 12/13/2022]
Abstract
In recent years, many promising nanotechnological approaches to biomedical research have been developed in order to increase implementation of regenerative medicine and tissue engineering in clinical practice. In the meantime, the use of nanomaterials for the regeneration of diseased or injured tissues is considered advantageous in most areas of medicine. In particular, for the treatment of cardiovascular, osteochondral and neurological defects, but also for the recovery of functions of other organs such as kidney, liver, pancreas, bladder, urethra and for wound healing, nanomaterials are increasingly being developed that serve as scaffolds, mimic the extracellular matrix and promote adhesion or differentiation of cells. This review focuses on the latest developments in regenerative medicine, in which iron oxide nanoparticles (IONPs) play a crucial role for tissue engineering and cell therapy. IONPs are not only enabling the use of non-invasive observation methods to monitor the therapy, but can also accelerate and enhance regeneration, either thanks to their inherent magnetic properties or by functionalization with bioactive or therapeutic compounds, such as drugs, enzymes and growth factors. In addition, the presence of magnetic fields can direct IONP-labeled cells specifically to the site of action or induce cell differentiation into a specific cell type through mechanotransduction.
Collapse
|
3
|
VEGF Contributes to Mesenchymal Stem Cell-Mediated Reversion of Nor1-Dependent Hypertrophy in iPS Cell-Derived Cardiomyocytes. Stem Cells Int 2021; 2021:8888575. [PMID: 33927770 PMCID: PMC8053052 DOI: 10.1155/2021/8888575] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 03/02/2021] [Accepted: 03/24/2021] [Indexed: 01/19/2023] Open
Abstract
Myocardial hypertrophy is present in many heart diseases, representing a strong predictor of adverse cardiovascular outcomes. Regarding therapeutic intervention, mesenchymal stem cells (MSCs) have been suggested to significantly reduce cardiac hypertrophy and progression to heart failure. Preconditioning of MSCs was previously demonstrated to highly improve their paracrine activity resulting in modulation of immune responses and the progression of diseases. Here, we studied the effects of bone marrow-derived preconditioned MSCs on hypertrophied induced pluripotent stem cell-derived cardiomyocytes (iPS-CM) and also sought to identify MSC-derived antihypertrophic molecules. Phenylephrine (PE) was used to induce hypertrophy in murine iPS-CM, and markers of hypertrophy were identified by microarray analysis. Murine MSCs were treated with IFN-γ and IL-1β to enhance their paracrine activity, and transcriptional profiling was performed by microarray analysis. Hypertrophied iPS-CM were subsequently cocultured with preconditioned MSCs or MSC-conditioned medium (CM), respectively. Effects on hypertrophied iPS-CM were studied by cell area quantification, real-time PCR, and western blot. In some experiments, cells were incubated with fractions of MSC-CM obtained by ultrafiltration or by MSC-CM supplemented with inhibitory antibodies. Intracellular and extracellular levels of vascular endothelial growth factor (VEGF) were evaluated by western blot and ELISA. PE-induced hypertrophy in iPS-CM was associated with an upregulation of neuron-derived orphan receptor (Nor1) expression, activation of Akt, and inhibition of both strongly prevented hypertrophy induction in iPS-CM. VEGF secreted by preconditioned MSCs provoked hypertrophy regression in iPS-CM, and a negative correlation between Nor1 expression and hypertrophic growth could be evidenced. Our results demonstrate that Nor1 expression strongly supports hypertrophy in iPS-CM. Moreover, the secretome of preconditioned MSCs triggered regression of hypertrophy in iPS-CM in a VEGF-dependent manner. We suggest that the delivery of the MSC-derived secretome may represent a therapeutic strategy to limit cardiac hypertrophy. However, additional in vivo studies are needed to prove this hypothesis.
Collapse
|
4
|
Klontzas ME, Kakkos GA, Papadakis GZ, Marias K, Karantanas AH. Advanced clinical imaging for the evaluation of stem cell based therapies. Expert Opin Biol Ther 2021; 21:1253-1264. [PMID: 33576278 DOI: 10.1080/14712598.2021.1890711] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction: As stem cell treatments reach closer to the clinic, the need for appropriate noninvasive imaging for accurate disease diagnosis, treatment planning, follow-up, and early detection of complications, is constantly rising. Clinical radiology affords an extensive arsenal of advanced imaging techniques, to provide anatomical and functional information on the whole spectrum of stem cell treatments from diagnosis to follow-up.Areas covered: This manuscript aims at providing a critical review of major published studies on the utilization of advanced imaging for stem cell treatments. Uses of magnetic resonance imaging (MRI), computed tomography (CT), ultrasound, and positron emission tomography (PET) are reviewed and interrogated for their applicability to stem cell imaging.Expert opinion: A wide spectrum of imaging methods have been utilized for the evaluation of stem cell therapies. The majority of published techniques are not clinically applicable, using methods exclusively applicable to animals or technology irrelevant to current clinical practice. Harmonization of preclinical methods with clinical reality is necessary for the timely translation of stem cell therapies to the clinic. Methods such as diffusion weighted MRI, hybrid imaging, and contrast-enhanced ultrasound hold great promise and should be routinely incorporated in the evaluation of patients receiving stem cell treatments.
Collapse
Affiliation(s)
- Michail E Klontzas
- Department of Medical Imaging, University Hospital of Heraklion, Crete, Greece.,Advanced Hybrid Imaging Systems, Institute of Computer Science, Foundation for Research and Technology (FORTH), Heraklion, Crete, Greece
| | - George A Kakkos
- Department of Medical Imaging, University Hospital of Heraklion, Crete, Greece
| | - Georgios Z Papadakis
- Advanced Hybrid Imaging Systems, Institute of Computer Science, Foundation for Research and Technology (FORTH), Heraklion, Crete, Greece.,Computational Biomedicine Laboratory (CBML), Foundation for Research and Technology Hellas (FORTH), Heraklion, Crete, Greece.,Department of Radiology, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Kostas Marias
- Computational Biomedicine Laboratory (CBML), Foundation for Research and Technology Hellas (FORTH), Heraklion, Crete, Greece.,Department of Electrical and Computer Engineering, Hellenic Mediterranean University, Heraklion, Crete, Greece
| | - Apostolos H Karantanas
- Department of Medical Imaging, University Hospital of Heraklion, Crete, Greece.,Advanced Hybrid Imaging Systems, Institute of Computer Science, Foundation for Research and Technology (FORTH), Heraklion, Crete, Greece.,Computational Biomedicine Laboratory (CBML), Foundation for Research and Technology Hellas (FORTH), Heraklion, Crete, Greece.,Department of Radiology, School of Medicine, University of Crete, Heraklion, Crete, Greece
| |
Collapse
|
5
|
Shelat R, Bhatt LK, Paunipagar B, Kurian T, Khanna A, Chandra S. Regeneration of hyaline cartilage in osteochondral lesion model using L-lysine magnetic nanoparticles labeled mesenchymal stem cells and their in vivo imaging. J Tissue Eng Regen Med 2020; 14:1604-1617. [PMID: 32840054 DOI: 10.1002/term.3120] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/24/2020] [Accepted: 08/04/2020] [Indexed: 12/16/2022]
Abstract
Treatment of osteochondral defects continues to pose a major challenge for patients and orthopedic surgeons due to the limited healing potential of articular cartilage. Mesenchymal stem cells (MSCs) possess therapeutic potential for the treatment of osteochondral pain and pathology. However, it is necessary to use proper labeling and imaging agent of stem cells that can decipher its role posttransplantation. A major limitation of routinely used contrast agents is signal dilution over a period of time which limits its use for further studies. At the same time, regeneration of fibrocartilage over native hyaline cartilage also limits the use of conventional therapies. The present study evaluates the efficacy of bone marrow-derived mesenchymal stem cells (BMSCs) for the treatment of osteochondral defect in rats with the regeneration of hyaline cartilage in situ and in vivo monitoring of the stem cells using L-lysine functionalized magnetic iron oxide nanoparticles (lys-IONPs). L-lysine stabilizes the iron oxide nanoparticles, enhances the biocompatibility, and provides functionalities for efficient stem cell labeling. in vitro toxic effects of lys-IONPs on mitochondrial impairment, morphological alterations, and actin cytoskeleton reveal minimum damage to BM-MSCs. Histological data (H and E, Masson's trichrome and immunohistochemistry) describe the early initiation of healing and regeneration of hyaline-like cartilage over fibrocartilage in stem cell treated groups. MR scans demonstrate generation of hypointense signals in lys-IONPs-BMSCs with improved signal intensity and minimum loss over 28 days revealing its use as a long-term stem cell labeling and imaging agent.
Collapse
Affiliation(s)
- Ruchita Shelat
- Department of Biological Sciences, Sunandan Divatia School of Science, NMIMS University, Mumbai, India
| | - Lokesh Kumar Bhatt
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | | | | | - Aparna Khanna
- Department of Biological Sciences, Sunandan Divatia School of Science, NMIMS University, Mumbai, India.,Computational Biology and Translational Research, Amity University, Mumbai, India
| | - Sudeshna Chandra
- Department of Biological Sciences, Sunandan Divatia School of Science, NMIMS University, Mumbai, India
| |
Collapse
|
6
|
Dou L, Matz EL, Gu X, Shu F, Paxton J, Song J, Yoo J, Atala A, Jackson J, Zhang Y. Non-Invasive Cell Tracking with Brighter and Red-Transferred Luciferase for Potential Application in Stem Cell Therapy. Cell Transplant 2019; 28:1542-1551. [PMID: 31684762 PMCID: PMC6923553 DOI: 10.1177/0963689719885078] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
This study investigated the safety of a novel cell-labeling technology with mKATE and
Renilla reniformis luciferase (mKATE-renLUC) and assessed the efficacy
on tracking implanted human placental stromal cells (PSC) in an erectile dysfunction (ED)
animal model. Human PSC were labeled with mKATE-renLUC using a lentivirus. Cell viability,
apoptosis, proliferation, migration, surface marker expression and differentiation
potential of the labeled PSC were evaluated and compared with non-labeled PSC. The
paracrine profile of labeled cells was examined using an angiogenesis protein array. The
brightness and duration of labeled cells with different densities were evaluated. An ED
rat model was established and labeled PSC were injected into cavernosal tissue of the
penis. The migration and distribution of transplanted PSC were monitored using an IVIS
imaging system in real time. Implanted PSC were identified in isolated tissues via
detection of mKATE fluorescence. The cell viability, morphology, proliferation, migration,
surface marker expression and differentiation potential of mKATE-renLUC-labeled PSC were
similar to those of non-labeled cells in vitro (no statistical difference
p>0.05). Similar expressions of trophic factors were found between
labeled and non-labeled PSC. The migration and distribution of PSC expressing renLUC were
tracked in vivo using IVIS imaging system. mKATE-positive PSC were detected in penile,
kidney, prostate and hepatic tissues using histological methods. This labeling technology
provides a safe and effective cell-tracking approach with a brighter fluorophore and
codon-optimized luciferase.
Collapse
Affiliation(s)
- Lei Dou
- Stomatological Hospital of Chongqing Medical University, Chongqing, China.,Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
| | - Ethan L Matz
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
| | - Xin Gu
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
| | - Fangpeng Shu
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
| | - Jennifer Paxton
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
| | - Jinlin Song
- Stomatological Hospital of Chongqing Medical University, Chongqing, China
| | - James Yoo
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
| | - John Jackson
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
| | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, USA
| |
Collapse
|
7
|
Zeriouh M, Sabashnikov A, Tenbrock A, Neef K, Merkle J, Eghbalzadeh K, Weber C, Liakopoulos OJ, Deppe AC, Stamm C, Cowan DB, Wahlers T, Choi YH. Dysregulation of proangiogeneic factors in pressure-overload left-ventricular hypertrophy results in inadequate capillary growth. Ther Adv Cardiovasc Dis 2019; 13:1753944719841795. [PMID: 31088231 PMCID: PMC6535753 DOI: 10.1177/1753944719841795] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Background: Pressure-overload left-ventricular hypertrophy (LVH) is an increasingly prevalent pathological condition of the myocardial muscle and an independent risk factor for a variety of cardiac diseases. We investigated changes in expression levels of proangiogeneic genes in a small animal model of LVH. Methods: Myocardial hypertrophy was induced by transaortic constriction (TAC) in C57BL/6 mice and compared with sham-operated controls. The myocardial expression levels of vascular endothelial growth factor (VEGF), its receptors (KDR and FLT-1), stromal-cell-derived factor 1 (SDF1) and the transcription factors hypoxia-inducible factor-1 and 2 (HIF1 and HIF2) were analyzed by quantitative polymerase chain reaction over the course of 25 weeks. Histological sections were stained for caveolin-1 to visualize endothelial cells and determine the capillary density. The left-ventricular morphology and function were assessed weekly by electrocardiogram-gated magnetic resonance imaging. Results: The heart weight of TAC animals increased significantly from week 4 to 25 (p = 0.005) compared with sham-treated animals. At 1 day after TAC, the expression of VEGF and SDF1 also increased, but was downregulated again after 1 week. The expression of HIF2 was significantly downregulated after 1 week and remained at a lower level in the subsequent weeks. The expression level of FLT-1 was also significantly decreased 1 week after TAC. HIF-1 and KDR showed similar changes compared with sham-operated animals. However, the expression levels of HIF1 after 4 and 8 weeks were significantly decreased compared with day 1. KDR changes were significantly decreased after 1, 2, 4, 8 and 25 weeks compared with week 3. After 4 weeks post-TAC, the size of the capillary vessels increased (p = 0.005) while the capillary density itself decreased (TAC: 2143 ± 293 /mm2versus sham: 2531 ± 321 /mm2; p = 0.021). Starting from week 4, the left-ventricular ejection fraction decreased compared with controls (p = 0.049). Conclusions: The decrease in capillary density in the hypertrophic myocardium appears to be linked to the dysregulation in the expression of proangiogeneic factors. The results suggest that overcoming this dysregulation may lead to reconstitution of capillary density in the hypertrophic heart, and thus be beneficial for cardiac function and survival.
Collapse
Affiliation(s)
- Mohamed Zeriouh
- Department of Cardiothoracic Surgery, University of Cologne, Cologne, Germany
| | - Anton Sabashnikov
- Department of Cardiothoracic Surgery, University of Cologne, Cologne, Germany
| | - Arne Tenbrock
- Department of Cardiothoracic Surgery, University of Cologne, Cologne, Germany
| | - Klaus Neef
- Department of Cardiothoracic Surgery, University of Cologne, Cologne, Germany
| | - Julia Merkle
- Department of Cardiothoracic Surgery, University of Cologne, Cologne, Germany
| | - Kaveh Eghbalzadeh
- Department of Cardiothoracic Surgery, University of Cologne, Cologne, Germany
| | - Carolyn Weber
- Department of Cardiothoracic Surgery, University of Cologne, Cologne, Germany
| | | | | | - Christof Stamm
- Berlin-Brandenburg Center for Regenerative Therapies, Berlin, Germany
| | - Douglas B Cowan
- Department of Anesthesiology, Perioperative and Pain Medicine, Children's Hospital Boston and Harvard Medical School, Boston, MA, USA
| | - Thorsten Wahlers
- Department of Cardiothoracic Surgery, University of Cologne, Cologne, Germany.,Center of Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Yeong-Hoon Choi
- Center of Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
8
|
Turnbull MT, Zubair AC, Meschia JF, Freeman WD. Mesenchymal stem cells for hemorrhagic stroke: status of preclinical and clinical research. NPJ Regen Med 2019; 4:10. [PMID: 31098299 PMCID: PMC6513857 DOI: 10.1038/s41536-019-0073-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 04/25/2019] [Indexed: 02/06/2023] Open
Abstract
Significant progress has been made during the past few decades in stem cell therapy research for various diseases and injury states; however this has not been overwhelmingly translated into approved therapies, despite much public attention and the rise in unregulated 'regenerative clinics'. In the last decade, preclinical research focusing on mesenchymal stem/stromal cell (MSC) therapy in experimental animal models of hemorrhagic stroke has gained momentum and has led to the development of a small number of human trials. Here we review the current studies focusing on MSC therapy for hemorrhagic stroke in an effort to summarize the status of preclinical and clinical research. Preliminary evidence indicates that MSCs are both safe and tolerable in patients, however future randomized controlled trials are required to translate the promising preclinical research into an effective therapy for hopeful patients.
Collapse
Affiliation(s)
| | - Abba C. Zubair
- Department of Laboratory Medicine and Pathology, Mayo Clinic Florida, Jacksonville, FL USA
| | - James F. Meschia
- Department of Neurology, Mayo Clinic Florida, Jacksonville, FL USA
| | - William D. Freeman
- Department of Neurology, Mayo Clinic Florida, Jacksonville, FL USA
- Department of Neurologic Surgery, Mayo Clinic Florida, Jacksonville, FL USA
- Department of Critical Care Medicine, Mayo Clinic Florida, Jacksonville, FL USA
| |
Collapse
|
9
|
Philipp D, Suhr L, Wahlers T, Choi YH, Paunel-Görgülü A. Preconditioning of bone marrow-derived mesenchymal stem cells highly strengthens their potential to promote IL-6-dependent M2b polarization. Stem Cell Res Ther 2018; 9:286. [PMID: 30359316 PMCID: PMC6202843 DOI: 10.1186/s13287-018-1039-2] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 08/16/2018] [Accepted: 10/07/2018] [Indexed: 12/16/2022] Open
Abstract
Background During the last decade, mesenchymal stem cells (MSCs) have gained much attention in the field of regenerative medicine due to their capacity to differentiate into different cell types and to promote immunosuppressive effects. However, the underlying mechanism of MSC-mediated immunoregulation is not fully understood so far. Macrophages are distinguished in classical activated, pro-inflammatory M1 and alternatively activated M2 cells, which possess different functions and transcriptional profiles with respect to inflammatory responses. As polarization is not fixed, macrophage functional plasticity might be modulated by the microenvironment allowing them to rapidly react to danger signals and maintaining tissue homeostasis. Methods Murine MSCs were preconditioned with IL-1ß and IFN-ɣ to enhance their immunosuppressive capacity regarding macrophage polarization under M1- and M2a-polarizing conditions. Macrophage polarization was analyzed by real-time PCR, flow cytometry, and cytokine detection in culture supernatants. The role of MSC-derived nitric oxide (NO), prostaglandin E2 (PGE2), and IL-6 in this process has been evaluated using siRNA transfection and IL-6 receptor-deficient macrophages, respectively. Results Preconditioned, but not unprimed, MSCs secreted high levels of NO, IL-6, and PGE2. Co-culture with macrophages (M0) in the presence of M1 inducers (LPS + IFN-ɣ) led to significant reduction of CD86 and iNOS protein in macrophages and diminished TNF-α secretion. Additionally, CD86 and iNOS protein expression as well as NO and IL-10 secretion were markedly increased under M2a-polarizing culture conditions (IL-4). MSC-dependent macrophage polarization did not depend on direct cell-cell contact. Co-culturing in the presence of LPS and IFN-ɣ resulted in the upregulation of M2a, M2b, and M2c marker genes, whereas in the presence of IL-4 only M2b markers were significantly increased. In turn, IL-10-producing regulatory M2b cells significantly inhibited IFN-ɣ expression in CD4+ T lymphocytes. Finally, we show that MSC-mediated macrophage polarization strongly depends on IL-6, whereas a minor role for NO and PGE2 was found. Conclusions Preconditioning of MSCs highly strengthens their capacity to regulate macrophage features and to promote immunosuppression. Repression of M1 polarization during inflammation and M2b polarization under anti-inflammatory conditions strongly depend on functional IL-6 signaling in macrophages. The potential benefit of preconditioned MSCs and IL-6 should be considered for future clinical treatment. Electronic supplementary material The online version of this article (10.1186/s13287-018-1039-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Denise Philipp
- Department of Cardiothoracic Surgery, Heart Center of the University of Cologne, Cologne, Germany
| | - Laura Suhr
- Department of Cardiothoracic Surgery, Heart Center of the University of Cologne, Cologne, Germany
| | - Thorsten Wahlers
- Department of Cardiothoracic Surgery, Heart Center of the University of Cologne, Cologne, Germany
| | - Yeong-Hoon Choi
- Department of Cardiothoracic Surgery, Heart Center of the University of Cologne, Cologne, Germany
| | - Adnana Paunel-Görgülü
- Department of Cardiothoracic Surgery, Heart Center of the University of Cologne, Cologne, Germany.
| |
Collapse
|
10
|
Using tyrosinase as a tri-modality reporter gene to monitor transplanted stem cells in acute myocardial infarction. Exp Mol Med 2018; 50:1-10. [PMID: 29700279 PMCID: PMC5938053 DOI: 10.1038/s12276-018-0080-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 02/21/2018] [Accepted: 02/26/2018] [Indexed: 12/24/2022] Open
Abstract
The study aimed to investigate the feasibility of noninvasive monitoring of bone marrow mesenchymal stem cells (MSCs) transduced with the tyrosinase reporter gene for acute myocardial infarction (AMI) with photoacoustic imaging (PAI), magnetic resonance imaging (MRI), and positron emission tomography (PET) in vitro and in vivo. MSCs were transduced with a lentivirus carrying a tyrosinase reporter gene. After transduction, the rate of 18F-5-fluoro-N-(2-[diethylamino]ethyl)picolinamide (18F-5-FPN) uptake was measured. PAI and MRI of stable cell lines expressing tyrosinase (TYR-MSCs) were performed in vitro. An AMI model was induced and verified. TYR-MSCs and MSCs were injected into the margins of the infarcted areas, and PAI, MRI, and PET images were acquired 1, 7, 14, 21, and 28 days after cell injection. Sham-operated models without injection were used as the control group. TYR-MSCs showed noticeably higher uptake of 18F-5-FPN and stronger signals in T1-weighted MRI and PAI than non-transduced MSCs. In vivo studies revealed prominent signals in the injected area of the infarcted myocardium on PAI/MRI/PET images, whereas no signal could be seen in rats injected with non-transduced MSCs or sham-operated rats. The uptake values of 18F-5-FPN in vivo showed a slight decrease over 28 days, whereas MRI and PAI signal intensity decreased dramatically. MSCs stably transduced with the tyrosinase reporter gene could be monitored in vivo in myocardial infarction models by PET, MRI, and PAI, providing a feasible and reliable method for checking the viability, location, and dwell time of transplanted stem cells. Developing stem cell treatments for heart disease could be aided by adding the gene for tyrosinase, an enzyme that can reveal the location and activity of the cells. Transplanting stem cells derived from bone marrow into injured hearts shows promise for repairing the damage caused by heart attacks. Developing the treatment is hampered by limitations of existing methods for monitoring the fate of the transplanted stem cells. Researchers in China led by Xiaoli Lan at Huazhong University of Science and Technology tackled this limitation by adding the gene for tyrosinase into the stem cells. The enzyme produces the pigment melanin which both directly and by binding to other chemicals generates signals which can be identified by photoacoustic imaging, magnetic resonance imaging and positron emission tomography. Tests in cultured cells and rats confirm the procedure’s potential.
Collapse
|
11
|
Dynamic Tracking of Injected Mesenchymal Stem Cells after Myocardial Infarction in Rats: A Serial 7T MRI Study. Stem Cells Int 2016; 2016:4656539. [PMID: 27656215 PMCID: PMC5021478 DOI: 10.1155/2016/4656539] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 07/25/2016] [Indexed: 11/26/2022] Open
Abstract
Purpose. To track the fate of micron-sized particles of iron oxide (MPIO) labeled mesenchymal stem cells (MSCs) in vivo in a rat myocardial infarction model using 7T magnetic resonance imaging (MRI) scanner. Materials and Methods. Male MSCs (2 × 106/50 μL) dual-labeled with MPIO and CM-DiI were injected into the infarct periphery 7 days after myocardial infarction (MI). The control group received cell-free media injection. The temporal stem cell location, signal intensity, and cardiac function were dynamically assessed using a 7T MRI at 24 h before transplantation (baseline), 3 days, 2 weeks, and 4 weeks after transplantation, respectively. Results. MR hypointensities caused by MPIOs were observed on T2⁎-weighted images at all time points after MSCs injection. Cine-MRI showed that MSCs moderated progressive left ventricular remodeling. Double staining for iron and CD68 revealed that most of the iron-positive cells were CD68-positive macrophages. Real-time PCR for rat SRY gene showed the number of survival MSCs considerably decreased after transplantation. MSC-treated hearts had significantly increased capillary density in peri-infarct region and lower cardiomyocytes apoptosis and fibrosis formation. Conclusions. Iron particles are not a reliable marker for in vivo tracking the long-term fate of MSCs engraftment. Despite of poor cell retention, MSCs moderate left ventricular remodeling after MI.
Collapse
|
12
|
Hnatiuk AP, Ong SG, Olea FD, Locatelli P, Riegler J, Lee WH, Jen CH, De Lorenzi A, Giménez CS, Laguens R, Wu JC, Crottogini A. Allogeneic Mesenchymal Stromal Cells Overexpressing Mutant Human Hypoxia-Inducible Factor 1-α (HIF1-α) in an Ovine Model of Acute Myocardial Infarction. J Am Heart Assoc 2016; 5:JAHA.116.003714. [PMID: 27385426 PMCID: PMC5015403 DOI: 10.1161/jaha.116.003714] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background Bone marrow mesenchymal stromal cells (BMMSCs) are cardioprotective in acute myocardial infarction (AMI) because of release of paracrine angiogenic and prosurvival factors. Hypoxia‐inducible factor 1‐α (HIF1‐α), rapidly degraded during normoxia, is stabilized during ischemia and upregulates various cardioprotective genes. We hypothesized that BMMSCs engineered to overexpress mutant, oxygen‐resistant HIF1‐α would confer greater cardioprotection than nontransfected BMMSCs in sheep with AMI. Methods and Results Allogeneic BMMSCs transfected with a minicircle vector encoding mutant HIF1‐α (BMMSC‐HIF) were injected in the peri‐infarct of sheep (n=6) undergoing coronary occlusion. Over 2 months, infarct volume measured by cardiac magnetic resonance (CMR) imaging decreased by 71.7±1.3% (P<0.001), and left ventricular (LV) percent ejection fraction (%EF) increased near 2‐fold (P<0.001) in the presence of markedly decreased end‐systolic volume. Sheep receiving nontransfected BMMSCs (BMMSC; n=6) displayed less infarct size limitation and percent LVEF improvement, whereas in placebo‐treated animals (n=6), neither parameters changed over time. HIF1‐α‐transfected BMMSCs (BMMSC‐HIF) induced angio‐/arteriogenesis and decreased apoptosis by HIF1‐mediated overexpression of erythropoietin, inducible nitrous oxide synthase, vascular endothelial growth factor, and angiopoietin‐1. Cell tracking using paramagnetic iron nanoparticles in 12 additional sheep revealed enhanced long‐term retention of BMMSC‐HIF. Conclusions Intramyocardial delivery of BMMSC‐HIF reduced infarct size and improved LV systolic performance compared to BMMSC, attributed to increased neovascularization and cardioprotective effects induced by HIF1‐mediated overexpression of paracrine factors and enhanced retention of injected cells. Given the safety of the minicircle vector and the feasibility of BMMSCs for allogeneic application, this treatment may be potentially useful in the clinic.
Collapse
Affiliation(s)
- Anna P Hnatiuk
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Universidad Favaloro-CONICET, Buenos Aires, Argentina Departamento de Fisiología, Facultad de Ciencias Médicas, Universidad Favaloro, Buenos Aires, Argentina
| | - Sang-Ging Ong
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA
| | - Fernanda D Olea
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Universidad Favaloro-CONICET, Buenos Aires, Argentina Departamento de Fisiología, Facultad de Ciencias Médicas, Universidad Favaloro, Buenos Aires, Argentina
| | - Paola Locatelli
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Universidad Favaloro-CONICET, Buenos Aires, Argentina Departamento de Fisiología, Facultad de Ciencias Médicas, Universidad Favaloro, Buenos Aires, Argentina
| | - Johannes Riegler
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA
| | - Won Hee Lee
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA
| | | | - Andrea De Lorenzi
- Departmento de Cardiología, Hospital Universitario de la Foundación Favaloro, Buenos Aires, Argentina
| | - Carlos S Giménez
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Universidad Favaloro-CONICET, Buenos Aires, Argentina Departamento de Fisiología, Facultad de Ciencias Médicas, Universidad Favaloro, Buenos Aires, Argentina
| | - Rubén Laguens
- Departmento de Patología, Facultad de Ciencias Médicas, Universidad Favaloro, Buenos Aires, Argentina
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA
| | - Alberto Crottogini
- Instituto de Medicina Traslacional, Trasplante y Bioingeniería (IMETTYB), Universidad Favaloro-CONICET, Buenos Aires, Argentina Departamento de Fisiología, Facultad de Ciencias Médicas, Universidad Favaloro, Buenos Aires, Argentina
| |
Collapse
|
13
|
Li X, Zhao H, Qi C, Zeng Y, Xu F, Du Y. Direct intercellular communications dominate the interaction between adipose-derived MSCs and myofibroblasts against cardiac fibrosis. Protein Cell 2015; 6:735-45. [PMID: 26271509 DOI: 10.1007/s13238-015-0196-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 07/10/2015] [Indexed: 02/07/2023] Open
Abstract
The onset of cardiac fibrosis post myocardial infarction greatly impairs the function of heart. Recent advances of cell transplantation showed great benefits to restore myocardial function, among which the mesenchymal stem cells (MSCs) has gained much attention. However, the underlying cellular mechanisms of MSC therapy are still not fully understood. Although paracrine effects of MSCs on residual cardiomyocytes have been discussed, the amelioration of fibrosis was rarely studied as the hostile environment cannot support the survival of most cell populations and impairs the diffusion of soluble factors. Here in order to decipher the potential mechanism of MSC therapy for cardiac fibrosis, we investigated the interplay between MSCs and cardiac myofibroblasts (mFBs) using interactive co-culture method, with comparison to paracrine approaches, namely treatment by MSC conditioned medium and gap co-culture method. Various fibrotic features of mFBs were analyzed and the most prominent anti-fibrosis effects were always obtained using direct co-culture that allowed cell-to-cell contacts. Hepatocyte growth factor (HGF), a well-known anti-fibrosis factor, was demonstrated to be a major contributor for MSCs' anti-fibrosis function. Moreover, physical contacts and tube-like structures between MSCs and mFBs were observed by live cell imaging and TEM which demonstrate the direct cellular interactions.
Collapse
Affiliation(s)
- Xiaokang Li
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treament of Infectious Diseases, Tsinghua University, Beijing, 100084, China
| | - Hui Zhao
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treament of Infectious Diseases, Tsinghua University, Beijing, 100084, China
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Chunxiao Qi
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treament of Infectious Diseases, Tsinghua University, Beijing, 100084, China
| | - Yang Zeng
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treament of Infectious Diseases, Tsinghua University, Beijing, 100084, China
| | - Feng Xu
- MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiao Tong University, Xi'an, 710049, China
| | - Yanan Du
- Department of Biomedical Engineering, School of Medicine, Collaborative Innovation Center for Diagnosis and Treament of Infectious Diseases, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
14
|
Brodarac A, Šarić T, Oberwallner B, Mahmoodzadeh S, Neef K, Albrecht J, Burkert K, Oliverio M, Nguemo F, Choi YH, Neiss WF, Morano I, Hescheler J, Stamm C. Susceptibility of murine induced pluripotent stem cell-derived cardiomyocytes to hypoxia and nutrient deprivation. Stem Cell Res Ther 2015; 6:83. [PMID: 25900017 PMCID: PMC4445302 DOI: 10.1186/s13287-015-0057-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 05/23/2014] [Accepted: 03/19/2015] [Indexed: 01/06/2023] Open
Abstract
Introduction Induced pluripotent stem cell-derived cardiomyocytes (iPS-CMs) may be suitable for myocardial repair. While their functional and structural properties have been extensively investigated, their response to ischemia-like conditions has not yet been clearly defined. Methods iPS-CMs were differentiated and enriched from murine induced pluripotent stem cells expressing enhanced green fluorescent protein (eGFP) and puromycin resistance genes under the control of an α-myosin heavy chain (α-MHC) promoter. iPS-CMs maturity and function were characterized by microscopy, real-time PCR, calcium transient recordings, electrophysiology, and mitochondrial function assays, and compared to those from neonatal murine cardiomyocytes. iPS-CMs as well as neonatal murine cardiomyocytes were exposed for 3 hours to hypoxia (1% O2) and glucose/serum deprivation, and viability, apoptosis markers, reactive oxygen species, mitochondrial membrane potential and intracellular stress signaling cascades were investigated. Then, the iPS-CMs response to mesenchymal stromal cell-conditioned medium was determined. Results iPS-CMs displayed key morphological and functional properties that were comparable to those of neonatal cardiomyocytes, but several parameters indicated an earlier iPS-CMs maturation stage. During hypoxia and glucose/serum deprivation, iPS-CMs exhibited a significantly higher proportion of poly-caspase-active, 7-aminoactinomycin D-positive and TUNEL-positive cells than neonatal cardiomyocytes. The average mitochondrial membrane potential was reduced in “ischemic” iPS-CMs but remained unchanged in neonatal cardiomyocytes; reactive oxygen species production was only increased in “ischemic” iPS-CMs, and oxidoreductase activity in iPS-CMs dropped more rapidly than in neonatal cardiomyocytes. In iPS-CMs, hypoxia and glucose/serum deprivation led to upregulation of Hsp70 transcripts and decreased STAT3 phosphorylation and total PKCε protein expression. Treatment with mesenchymal stromal cell-conditioned medium preserved oxidoreductase activity and restored pSTAT3 and PKCε levels. Conclusion iPS-CMs appear to be particularly sensitive to hypoxia and nutrient deprivation. Counteracting the ischemic susceptibility of iPS-CMs with mesenchymal stromal cell-conditioned medium may help enhance their survival and efficacy in cell-based approaches for myocardial repair.
Collapse
Affiliation(s)
- Andreja Brodarac
- Berlin-Brandenburg Center for Regenerative Therapies, Föhrer Str.15, Berlin, 13353, Germany.
| | - Tomo Šarić
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany.
| | - Barbara Oberwallner
- Berlin-Brandenburg Center for Regenerative Therapies, Föhrer Str.15, Berlin, 13353, Germany.
| | | | - Klaus Neef
- Department of Cardiothoracic Surgery, Heart Center, University Hospital Cologne, Cologne, Germany.
| | - Julie Albrecht
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany.
| | - Karsten Burkert
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany.
| | - Matteo Oliverio
- Max-Planck-Institute for Metabolism Research, Cologne, Germany.
| | - Filomain Nguemo
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany.
| | - Yeong-Hoon Choi
- Department of Cardiothoracic Surgery, Heart Center, University Hospital Cologne, Cologne, Germany.
| | - Wolfram F Neiss
- Department of Anatomy I, Medical Faculty, University of Cologne, Cologne, Germany.
| | - Ingo Morano
- Max-Delbrueck-Center for Molecular Medicine, Berlin, Germany.
| | - Jürgen Hescheler
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Faculty, University of Cologne, Cologne, Germany.
| | - Christof Stamm
- Berlin-Brandenburg Center for Regenerative Therapies, Föhrer Str.15, Berlin, 13353, Germany. .,Deutsches Herzzentrum Berlin, Berlin, Germany.
| |
Collapse
|
15
|
Bourzac CA, Koenig JB, Link KA, Nykamp SG, Koch TG. Evaluation of ultrasmall superparamagnetic iron oxide contrast agent labeling of equine cord blood and bone marrow mesenchymal stromal cells. Am J Vet Res 2015; 75:1010-7. [PMID: 25350092 DOI: 10.2460/ajvr.75.11.1010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To evaluate the efficacy and effects of labeling equine umbilical cord blood (UCB)- and bone marrow (BM)-derived multipotent mesenchymal stromal cells (MSCs) with an ultrasmall superparamagnetic iron oxide (SPIO) contrast agent and the detection of labeled MSCs by use of MRI. SAMPLE UCB MSCs from placental tissues of 5 foals and BM MSCs from 5 horses. PROCEDURES UCB and BM MSC cultures were seeded in duplicate (5,000 cells/cm(2)). One duplicate was incubated with SPIO (50 μg/mL); the other was processed identically, but without SPIO. Mesenchymal stromal cells were expanded in triplicates for 5 passages and assessed for viability and proliferative capacity, labeling efficacy, and labeled cell proportion. For MRI detection, 5 × 10(6) labeled BM MSCs from passage 1 or 2 were injected into a collagenase-induced superficial digital flexor tendon defect of an equine cadaveric forelimb from 2 horses. RESULTS For passages 1, 2, and 3, labeling efficacy and cell proportion for UCB MSCs (99.6% [range, 98.8% to 99.9%], 16.6% [range, 6.5% to 36.1%], and 1.0% [range, 0.4% to 2.8%], respectively) were significantly higher than for BM MSCs (99.2% [range, 97.8% to 99.7%], 4.5% [range, 1.6% to 11.8%], and 0.2% [range, 0.1% to 0.6%], respectively). Labeling was not detectable after passage 3. Viability of MSCs was not affected, but cell doubling time increased in labeled MSCs, compared with that of unlabeled MSCs. On MRI 3-D T2*-weighted fast gradient echo sequences, decreased signal intensity was observed for BM passage 1 MSCs. CONCLUSIONS AND CLINICAL RELEVANCE Equine UCB and BM MSCs were labeled with SPIO at high efficiencies.
Collapse
Affiliation(s)
- Celine A Bourzac
- Departments of Clinical Studies, Ontario Veterinary College, University of Guelph, Guelph, ON N1G 2W1, Canada
| | | | | | | | | |
Collapse
|
16
|
Geng K, Yang ZX, Huang D, Yi M, Jia Y, Yan G, Cheng X, Wu R. Tracking of mesenchymal stem cells labeled with gadolinium diethylenetriamine pentaacetic acid by 7T magnetic resonance imaging in a model of cerebral ischemia. Mol Med Rep 2014; 11:954-60. [PMID: 25352164 PMCID: PMC4262487 DOI: 10.3892/mmr.2014.2805] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 10/01/2014] [Indexed: 12/11/2022] Open
Abstract
Progress in the development of stem cell and gene therapy requires repeatable and non-invasive techniques to monitor the survival and integration of stem cells in vivo with a high temporal and spatial resolution. The purpose of the present study was to examine the feasibility of using the standard contrast agent gadolinium diethylenetriamine pentaacetic acid (Gd-DTPA) to label rat mesenchymal stem cells (MSCs) for stem cell tracking. MSCs, obtained from the bilateral femora of rats, were cultured and propagated. The non-liposomal lipid transfection reagent effectene was then used to induce the intracellular uptake of Gd-DTPA. Electron microscopy was used to detect the distribution of Gd-DTPA particles in the MSCs. The labeling efficiency of the Gd-DTPA particles in the MSCs was determined using spectrophotometry, and MTT and trypan blue exclusion assays were used to evaluate the viability and proliferation of the labeled MSCs. T1-weighted magnetic resonance imaging (MRI) was used to observe the labeled cells in vitro and in the rat brain. Gd-DTPA particles were detected inside the MSCs using transmission electron microscopy and a high labeling efficiency was observed. No difference was observed in cell viability or proliferation between the labeled and unlabeled MSCs (P>0.05). In the in vitro T1-weighted MRI and in the rat brain, a high signal intensity was observed in the labeled MSCs. The T1-weighted imaging of the labeled cells revealed a significantly higher signal intensity compared with that of the unlabeled cells (P<0.05) and the T1 values were significantly lower. The function of the labeled MSCs demonstrated no change following Gd-DTPA labeling, with no evident adverse effect on cell viability or proliferation. Therefore, a change in MR signal intensity was detected in vitro and in vivo, suggesting Gd-DTPA can be used to label MSCs for MRI tracking.
Collapse
Affiliation(s)
- Kuan Geng
- The Chinese People's Liberation Army 59 Hospital, Yunnan, Kaiyuan, Yunnan 661699, P.R. China
| | - Zhong Xian Yang
- Department of Medical Imaging, The Second Affiliated Hospital, Medical College of Shantou University, Shantou, Guangdong 515041, P.R. China
| | - Dexiao Huang
- Department of Medical Imaging, The Second Affiliated Hospital, Medical College of Shantou University, Shantou, Guangdong 515041, P.R. China
| | - Meizi Yi
- Department of Medical Imaging, The Second Affiliated Hospital, Medical College of Shantou University, Shantou, Guangdong 515041, P.R. China
| | - Yanlong Jia
- Department of Medical Imaging, The Second Affiliated Hospital, Medical College of Shantou University, Shantou, Guangdong 515041, P.R. China
| | - Gen Yan
- Department of Medical Imaging, The Second Affiliated Hospital, Medical College of Shantou University, Shantou, Guangdong 515041, P.R. China
| | - Xiaofang Cheng
- Department of Medical Imaging, The Second Affiliated Hospital, Medical College of Shantou University, Shantou, Guangdong 515041, P.R. China
| | - Renhua Wu
- Department of Medical Imaging, The Second Affiliated Hospital, Medical College of Shantou University, Shantou, Guangdong 515041, P.R. China
| |
Collapse
|
17
|
Treskes P, Neef K, Perumal Srinivasan S, Halbach M, Stamm C, Cowan D, Scherner M, Madershahian N, Wittwer T, Hescheler J, Wahlers T, Choi YH. Preconditioning of skeletal myoblast-based engineered tissue constructs enables functional coupling to myocardium in vivo. J Thorac Cardiovasc Surg 2014; 149:348-56. [PMID: 25439779 DOI: 10.1016/j.jtcvs.2014.09.034] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 09/05/2014] [Accepted: 09/10/2014] [Indexed: 02/01/2023]
Abstract
OBJECTIVE Skeletal myoblasts fuse to form functional syncytial myotubes as an integral part of the skeletal muscle. During this differentiation process, expression of proteins for mechanical and electrical integration is seized, which is a major drawback for the application of skeletal myoblasts in cardiac regenerative cell therapy, because global heart function depends on intercellular communication. METHODS Mechanically preconditioned engineered tissue constructs containing neonatal mouse skeletal myoblasts were transplanted epicardially. A Y-chromosomal specific polymerase chain reaction (PCR) was undertaken up to 10 weeks after transplantation to confirm the presence of grafted cells. Histologic and electrophysiologic analyses were carried out 1 week after transplantation. RESULTS Cells within the grafted construct expressed connexin 43 at the interface to the host myocardium, indicating electrical coupling, confirmed by sharp electrode recordings. Analyses of the maximum stimulation frequency (5.65 ± 0.37 Hz), conduction velocity (0.087 ± 0.011 m/s) and sensitivity for pharmacologic conduction block (0.736 ± 0.080 mM 1-heptanol) revealed effective electrophysiologic coupling between graft and host cells, although significantly less robust than in native myocardial tissue (maximum stimulation frequency, 11.616 ± 0.238 Hz, P < .001; conduction velocity, 0.300 ± 0.057 m/s, P < .01; conduction block, 1.983 ± 0.077 mM 1-heptanol, P < .001). CONCLUSIONS Although untreated skeletal myoblasts cannot couple to cardiomyocytes, we confirm that mechanical preconditioning enables transplanted skeletal myoblasts to functionally interact with cardiomyocytes in vivo and, thus, reinvigorate the concept of skeletal myoblast-based cardiac cell therapy.
Collapse
Affiliation(s)
- Philipp Treskes
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Institute for Neurophysiology, University of Cologne, Cologne, Germany
| | - Klaus Neef
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Sureshkumar Perumal Srinivasan
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany; Institute for Neurophysiology, University of Cologne, Cologne, Germany
| | - Marcel Halbach
- Institute for Neurophysiology, University of Cologne, Cologne, Germany; Department of Internal Medicine III, Heart Center, University of Cologne, Cologne, Germany
| | - Christof Stamm
- Berlin-Brandenburg Center for Regenerative Therapies, Berlin, Germany
| | - Douglas Cowan
- Departments of Anesthesiology and Perioperative and Pain Medicine, Children's Hospital Boston and Harvard Medical School, Boston, Mass
| | - Maximilian Scherner
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany
| | - Navid Madershahian
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany
| | - Thorsten Wittwer
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany
| | - Jürgen Hescheler
- Institute for Neurophysiology, University of Cologne, Cologne, Germany
| | - Thorsten Wahlers
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Yeong-Hoon Choi
- Department of Cardiothoracic Surgery, Heart Center, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany.
| |
Collapse
|
18
|
Oberwallner B, Brodarac A, Anić P, Šarić T, Wassilew K, Neef K, Choi YH, Stamm C. Human cardiac extracellular matrix supports myocardial lineage commitment of pluripotent stem cells. Eur J Cardiothorac Surg 2014; 47:416-25; discussion 425. [PMID: 24778452 DOI: 10.1093/ejcts/ezu163] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVES Cross-talk between organ-specific extracellular matrix (ECM) and stem cells is often assumed but has not been directly demonstrated. We developed a protocol for the preparation of human cardiac ECM (cECM) and studied whether cECM has effects on pluripotent stem cell differentiation that may be useful for future cardiac regeneration strategies in patients with end-stage heart failure. METHODS Of note, 0.3 mm-thick cECM slices were prepared from samples of myocardium from patients with end-stage non-ischaemic dilated cardiomyopathy, using a three-step protocol involving hypotonic lysis buffer, sodium dodecyl sulphate (SDS) and foetal bovine serum (FBS). Murine embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs) and mesenchymal stromal cells (MSCs) were seeded and grown in standard culture, on cECM or on non-specific ECM preparations (Matrigel® or Geltrex®). Cell attachment, apoptosis induction (Caspase 3/7 activity) and metabolic activity (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium conversion) were followed. Transcriptional activation of genes involved in pluripotency; early and late myocardial development; and endothelial, ectodermal or endodermal commitment were monitored by quantitative real-time polymerase chain reaction (rtPCR). Protein expression of selected markers was confirmed by immunohistology. RESULTS cECM supported the proliferation of ESCs and iPSCs, and Caspase 3/7 activity was significantly lower compared with standard culture. Cardiac lineage commitment was favoured when ESCs or iPSCs were grown on cECM, as evidenced by the significantly increased mRNA expression of cardiac alpha myosin heavy polypeptide 6 (Myh6), cardiac troponin T2 (Tnnt2) and NK2 homeobox 5 (Nkx2.5) as well as positive immunohistology for cardiac troponin T and heavy-chain cardiac myosin protein. In contrast, Matrigel or Geltrex did not induce cardiac-specific markers. MSCs showed no evidence of cardiomyocyte differentiation. CONCLUSIONS Human cardiac ECM seems to direct differentiation of pluripotent stem cells towards a cardiomyocyte phenotype. This phenomenon supports the use of cardiac ECM preparations for guided stem cell differentiation and myocardial repair, and may ultimately increase the therapeutic efficacy of cell therapy in heart failure patients.
Collapse
Affiliation(s)
- Barbara Oberwallner
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Campus Virchow-Klinikum, Berlin, Germany
| | - Andreja Brodarac
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Campus Virchow-Klinikum, Berlin, Germany
| | - Petra Anić
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Campus Virchow-Klinikum, Berlin, Germany
| | - Tomo Šarić
- Center for Physiology and Pathophysiology, Institute for Neurophysiology, Medical Center, University of Cologne, Cologne, Germany
| | | | - Klaus Neef
- Department of Cardiac and Thoracic Surgery, Heart Center, University of Cologne, Cologne, Germany
| | - Yeong-Hoon Choi
- Department of Cardiac and Thoracic Surgery, Heart Center, University of Cologne, Cologne, Germany
| | - Christof Stamm
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Campus Virchow-Klinikum, Berlin, Germany Deutsches Herzzentrum Berlin (DHZB), Berlin, Germany
| |
Collapse
|
19
|
Rubach M, Adelmann R, Haustein M, Drey F, Pfannkuche K, Xiao B, Koester A, Udink ten Cate FEA, Choi YH, Neef K, Fatima A, Hannes T, Pillekamp F, Hescheler J, Šarić T, Brockmeier K, Khalil M. Mesenchymal stem cells and their conditioned medium improve integration of purified induced pluripotent stem cell-derived cardiomyocyte clusters into myocardial tissue. Stem Cells Dev 2014; 23:643-53. [PMID: 24219308 DOI: 10.1089/scd.2013.0272] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Induced pluripotent stem cell-derived cardiomyocytes (iPS-CMs) might become therapeutically relevant to regenerate myocardial damage. Purified iPS-CMs exhibit poor functional integration into myocardial tissue. The aim of this study was to investigate whether murine mesenchymal stem cells (MSCs) or their conditioned medium (MScond) improves the integration of murine iPS-CMs into myocardial tissue. Vital or nonvital embryonic murine ventricular tissue slices were cocultured with purified clusters of iPS-CMs in combination with murine embryonic fibroblasts (MEFs), MSCs, or MScond. Morphological integration was assessed by visual scoring and functional integration by isometric force and field potential measurements. We observed a moderate morphological integration of iPS-CM clusters into vital, but a poor integration into nonvital, slices. MEFs and MSCs but not MScond improved morphological integration of CMs into nonvital slices and enabled purified iPS-CMs to confer force. Coculture of vital slices with iPS-CMs and MEFs or MSCs resulted in an improved electrical integration. A comparable improvement of electrical coupling was achieved with the cell-free MScond, indicating that soluble factors secreted by MSCs were involved in electrical coupling. We conclude that cells such as MSCs support the engraftment and adhesion of CMs, and confer force to noncontractile tissue. Furthermore, soluble factors secreted by MSCs mediate electrical coupling of purified iPS-CM clusters to myocardial tissue. These data suggest that MSCs may increase the functional engraftment and therapeutic efficacy of transplanted iPS-CMs into infarcted myocardium.
Collapse
Affiliation(s)
- Martin Rubach
- 1 Department of Pediatric Cardiology, University of Cologne , Cologne, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Khurana A, Chapelin F, Beck G, Lenkov OD, Donig J, Nejadnik H, Messing S, Derugin N, Chan RCF, Gaur A, Sennino B, McDonald DM, Kempen PJ, Tikhomirov GA, Rao J, Daldrup-Link HE. Iron administration before stem cell harvest enables MR imaging tracking after transplantation. Radiology 2013; 269:186-97. [PMID: 23850832 DOI: 10.1148/radiol.13130858] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE To determine whether intravenous ferumoxytol can be used to effectively label mesenchymal stem cells (MSCs) in vivo and can be used for tracking of stem cell transplants. MATERIALS AND METHODS This study was approved by the institutional animal care and use committee. Sprague-Dawley rats (6-8 weeks old) were injected with ferumoxytol 48 hours prior to extraction of MSCs from bone marrow. Ferumoxytol uptake by these MSCs was evaluated with fluorescence, confocal, and electron microscopy and compared with results of traditional ex vivo-labeling procedures. The in vivo-labeled cells were subsequently transplanted in osteochondral defects of 14 knees of seven athymic rats and were evaluated with magnetic resonance (MR) imaging up to 4 weeks after transplantation. T2 relaxation times of in vivo-labeled MSC transplants and unlabeled control transplants were compared by using t tests. MR data were correlated with histopathologic results. RESULTS In vivo-labeled MSCs demonstrated significantly higher ferumoxytol uptake compared with ex vivo-labeled cells. With electron microscopy, iron oxide nanoparticles were localized in secondary lysosomes. In vivo-labeled cells demonstrated significant T2 shortening effects in vitro and in vivo when they were compared with unlabeled control cells (T2 in vivo, 15.4 vs 24.4 msec; P < .05) and could be tracked in osteochondral defects for 4 weeks. Histologic examination confirmed the presence of iron in labeled transplants and defect remodeling. CONCLUSION Intravenous ferumoxytol can be used to effectively label MSCs in vivo and can be used for tracking of stem cell transplants with MR imaging. This method eliminates risks of contamination and biologic alteration of MSCs associated with ex vivo-labeling procedures.
Collapse
Affiliation(s)
- Aman Khurana
- Department of Radiology and Molecular Imaging Program at Stanford, Stanford University School of Medicine, 725 Welch Rd, Room 1665, Stanford, CA 94305-5654; Department of Communication and Statistics and Department of Materials Science and Engineering, Stanford University, Stanford, Calif; Department of Neurology, Comprehensive Cancer Center, Cardiovascular Research Institute and Department of Anatomy, University of California San Francisco, San Francisco, Calif
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Dynamic Imaging of Marrow-Resident Granulocytes Interacting with Human Mesenchymal Stem Cells upon Systemic Lipopolysaccharide Challenge. Stem Cells Int 2013; 2013:656839. [PMID: 23606861 PMCID: PMC3625565 DOI: 10.1155/2013/656839] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2013] [Accepted: 02/27/2013] [Indexed: 12/13/2022] Open
Abstract
Human mesenchymal stem cells (hMSCs) have gained intense research interest due to their immune-modulatory, tissue differentiating, and homing properties to sites of inflammation. Despite evidence demonstrating the biodistribution of infused hMSCs in target organs using static fluorescence imaging or whole-body imaging techniques, surprisingly little is known about how hMSCs behave dynamically within host tissues on a single-cell level in vivo. Here, we infused fluorescently labeled clinical-grade hMSCs into immune-competent mice in which neutrophils and monocytes express a second fluorescent marker under the lysozyme M (LysM) promoter. Using intravital two-photon microscopy (TPM), we were able for the first time to capture dynamic interactions between hMSCs and LysM+ granulocytes in the calvarium bone marrow of recipient mice during systemic LPS challenge in real time. Interestingly, many of the infused hMSCs remained intact despite repeated cellular contacts with host neutrophils. However, we were able to observe the destruction and subsequent phagocytosis of some hMSCs by surrounding granulocytes. Thus, our imaging platform provides opportunities to gain insight into the biology and therapeutic mechanisms of hMSCs in vivo at a single-cell level within live hosts.
Collapse
|