1
|
Alptekin A, Khan MB, Parvin M, Chowdhury H, Kashif S, Selina FA, Bushra A, Kelleher J, Ghosh S, Williams D, Blumling E, Ara R, Bosomtwi A, Frank JA, Dhandapani KM, Arbab AS. Effects of low-intensity pulsed focal ultrasound-mediated delivery of endothelial progenitor-derived exosomes in tMCAo stroke. Front Neurol 2025; 16:1543133. [PMID: 40271117 PMCID: PMC12014438 DOI: 10.3389/fneur.2025.1543133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Accepted: 03/24/2025] [Indexed: 04/25/2025] Open
Abstract
Introduction Exosomes from different sources have been used for therapeutic purposes to target stroke and other disorders. However, exosomes from endothelial progenitor cells (EPCs) have not been tested in any stroke model, and in vivo bio-distribution study is lacking. Targeted delivery of IV-administered exosomes has been a significant challenge. Delivery of exosomes to the brain is a daunting task, and a blood-brain barrier (BBB)-penetrable peptide is being considered. However, the next step in practical treatment will be delivering naïve (unmodified) exosomes to the stroke site without destroying host tissues or disrupting BBB, or the membranes of the delivery vehicles. Low-intensity-pulsed focused ultrasound (LIPFUS) is approved for clinical use in the musculoskeletal, transcranial brain, and physiotherapy clinics. The objectives of the proposed studies were to determine whether LIPFUS-mediated increased delivery of EPC-derived exosomes enhances stroke recovery and functional improvement in mice with transient middle cerebral artery occlusion (tMCAo) stroke. Methods To enhance exosome delivery to the stroke area, we utilized LIPFUS. We evaluated stroke volume using MRI at different time points and conducted behavioral studies parallel to MRI to determine recovery. Ultimately, we studied brain tissue using immunohistochemistry to assess the extent of stroke and tissue regeneration. Results and Discussion In vivo, imaging showed a higher accumulation of EPC exosomes following LIPFUS without any damage to the underlying brain tissues, increased leakage of albumin, or accumulation of CD45+ cells. Groups of mice (14-16 months old) were treated with Vehicle (PBS), LIPFUS only, EPC-exosomes only, and LIPFUS+EPC-exosomes. LIPFUS + EPC exosomes groups showed a significantly decreased stroke volume on day 7, decreased FluoroJade+ cells, and significantly higher numbers of neovascularization in and around the stroke areas compared to that of other groups.
Collapse
Affiliation(s)
- Ahmet Alptekin
- Tumor Angiogenesis Laboratory, GCC, Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Mohammad B. Khan
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Mahrima Parvin
- Tumor Angiogenesis Laboratory, GCC, Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Hasanul Chowdhury
- Tumor Angiogenesis Laboratory, GCC, Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Sawaiz Kashif
- Tumor Angiogenesis Laboratory, GCC, Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Fowzia A. Selina
- Tumor Angiogenesis Laboratory, GCC, Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Anika Bushra
- Tumor Angiogenesis Laboratory, GCC, Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Justin Kelleher
- Tumor Angiogenesis Laboratory, GCC, Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Santu Ghosh
- Department of Biostatistics, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Dylan Williams
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Emily Blumling
- Department of Neurology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Roxan Ara
- Small Animal Imaging Core, GCC, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Asamoah Bosomtwi
- Small Animal Imaging Core, GCC, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Joseph A. Frank
- Laboratory of Diagnostic Radiology Research, Clinical Center, National Institutes of Health, Bethesda, MD, United States
| | - Krishnan M. Dhandapani
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Ali S. Arbab
- Tumor Angiogenesis Laboratory, GCC, Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
2
|
Xia LLCH, Girerd N, Lamiral Z, Duarte K, Merckle L, Leroy C, Nazare JA, Van Den Berghe L, Seconda L, Hoge A, Guillaume M, Laville M, Rossignol P, Boivin JM, Wagner S. Association between ultra-processed food consumption and inflammation: insights from the STANISLAS cohort. Eur J Nutr 2025; 64:94. [PMID: 39960649 DOI: 10.1007/s00394-025-03607-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 01/31/2025] [Indexed: 03/19/2025]
Abstract
PURPOSE High consumption of ultra-processed food (UPF) is associated with an increased risk of developing chronic diseases. Inflammation may be one of the underlying mechanisms behind this association. However, only a limited number of studies have investigated the association between UPF consumption and a few selected inflammation biomarkers, yielding inconsistent results. This study aimed to assess the cross-sectional association between UPF consumption (as a whole and 10 sub-categories), and 78 circulating proteins related to inflammation. METHODS The present study included 1594 adult participants from the STANISLAS cohort. UPF consumption was estimated using the NOVA classification, and linear regression models were used to assess their association with circulating proteins. RESULTS UPFs accounted for 28% of the total energy intake and 5.7 servings on average per day. In the unadjusted model, 15 circulating proteins had a significant association with UPF consumption. After adjustment, only (FGF-19) was significantly associated with UPF consumption (β = - 0.02[- 0.03; - 0.003]). CONCLUSION UPF consumption was negatively associated with Fibroblast Growth Factor 19 (FGF-19) serum levels. When considering UPF sub-categories, no circulating proteins were associated with dairy products and dairy desserts. Of note, circulating proteins were differentially associated depending on the sub-category of UPF. Further studies are needed to better understand the link between UPF and inflammation.
Collapse
Affiliation(s)
- Lea Lin Chun Hu Xia
- Centre d'Investigation Clinique-Plurithématique, Inserm CIC-P 1433, Pierre Drouin, INSERM, CHRU de Nancy Brabois, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Institut Lorrain du Coeur Et Des Vaisseaux Louis Mathieu, Université de Lorraine, 4 Rue du Morvan, 54500, Vandoeuvre-Lès-Nancy, France
| | - Nicolas Girerd
- Centre d'Investigation Clinique-Plurithématique, Inserm CIC-P 1433, Pierre Drouin, INSERM, CHRU de Nancy Brabois, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Institut Lorrain du Coeur Et Des Vaisseaux Louis Mathieu, Université de Lorraine, 4 Rue du Morvan, 54500, Vandoeuvre-Lès-Nancy, France.
| | - Zohra Lamiral
- Centre d'Investigation Clinique-Plurithématique, Inserm CIC-P 1433, Pierre Drouin, INSERM, CHRU de Nancy Brabois, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Institut Lorrain du Coeur Et Des Vaisseaux Louis Mathieu, Université de Lorraine, 4 Rue du Morvan, 54500, Vandoeuvre-Lès-Nancy, France
| | - Kevin Duarte
- Centre d'Investigation Clinique-Plurithématique, Inserm CIC-P 1433, Pierre Drouin, INSERM, CHRU de Nancy Brabois, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Institut Lorrain du Coeur Et Des Vaisseaux Louis Mathieu, Université de Lorraine, 4 Rue du Morvan, 54500, Vandoeuvre-Lès-Nancy, France
| | - Ludovic Merckle
- Centre d'Investigation Clinique-Plurithématique, Inserm CIC-P 1433, Pierre Drouin, INSERM, CHRU de Nancy Brabois, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Institut Lorrain du Coeur Et Des Vaisseaux Louis Mathieu, Université de Lorraine, 4 Rue du Morvan, 54500, Vandoeuvre-Lès-Nancy, France
| | - Celine Leroy
- Centre d'Investigation Clinique-Plurithématique, Inserm CIC-P 1433, Pierre Drouin, INSERM, CHRU de Nancy Brabois, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Institut Lorrain du Coeur Et Des Vaisseaux Louis Mathieu, Université de Lorraine, 4 Rue du Morvan, 54500, Vandoeuvre-Lès-Nancy, France
| | - Julie-Anne Nazare
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Hospices Civils de Lyon, F-CRIN/FORCE Network, Univ-Lyon, CarMeN Laboratory, Université Claude Bernard Lyon1, Lyon, France
| | - Laurie Van Den Berghe
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Hospices Civils de Lyon, F-CRIN/FORCE Network, Univ-Lyon, CarMeN Laboratory, Université Claude Bernard Lyon1, Lyon, France
| | - Louise Seconda
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Hospices Civils de Lyon, F-CRIN/FORCE Network, Univ-Lyon, CarMeN Laboratory, Université Claude Bernard Lyon1, Lyon, France
| | - Axelle Hoge
- Département Des Sciences de La Santé Publique, Université de Liège, Liège, Belgium
| | - Michèle Guillaume
- Département Des Sciences de La Santé Publique, Université de Liège, Liège, Belgium
| | - Martine Laville
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Hospices Civils de Lyon, F-CRIN/FORCE Network, Univ-Lyon, CarMeN Laboratory, Université Claude Bernard Lyon1, Lyon, France
| | - Patrick Rossignol
- Centre d'Investigation Clinique-Plurithématique, Inserm CIC-P 1433, Pierre Drouin, INSERM, CHRU de Nancy Brabois, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Institut Lorrain du Coeur Et Des Vaisseaux Louis Mathieu, Université de Lorraine, 4 Rue du Morvan, 54500, Vandoeuvre-Lès-Nancy, France
- Medicine and Nephrology-Dialysis Departments, Princess Grace Hospital, Monaco Private Hemodialysis Centre, Monaco, Monaco
- M-CRIN, (Monaco Clinical Research Infrastructure Network), Monaco, France
| | - Jean-Marc Boivin
- Centre d'Investigation Clinique-Plurithématique, Inserm CIC-P 1433, Pierre Drouin, INSERM, CHRU de Nancy Brabois, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Institut Lorrain du Coeur Et Des Vaisseaux Louis Mathieu, Université de Lorraine, 4 Rue du Morvan, 54500, Vandoeuvre-Lès-Nancy, France
- Department of General Medicine, University of Lorraine, Vandoeuvre-Lès-Nancy, France
| | - Sandra Wagner
- Centre d'Investigation Clinique-Plurithématique, Inserm CIC-P 1433, Pierre Drouin, INSERM, CHRU de Nancy Brabois, F-CRIN INI-CRCT (Cardiovascular and Renal Clinical Trialists), Institut Lorrain du Coeur Et Des Vaisseaux Louis Mathieu, Université de Lorraine, 4 Rue du Morvan, 54500, Vandoeuvre-Lès-Nancy, France
| |
Collapse
|
3
|
Falero-Diaz G, Barboza CDA, Vazquez-Padron RI, Velazquez OC, Lassance-Soares RM. Loss of c-Kit in Endothelial Cells Protects against Hindlimb Ischemia. Biomedicines 2024; 12:1358. [PMID: 38927565 PMCID: PMC11201387 DOI: 10.3390/biomedicines12061358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 06/04/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Critical limb ischemia (CLI) is the end stage of peripheral artery disease (PAD), and around 30% of CLI patients are ineligible for current treatments. The angiogenic benefits of c-Kit have been reported in the ischemia scenario; however, the present study demonstrates the effects of specific endothelial c-Kit signaling in arteriogenesis during hindlimb ischemia. METHODS We created conditional knockout mouse models that decrease c-Kit (c-Kit VE-Cadherin CreERT2-c-Kit) or its ligand (SCF VE-Cadherin CreERT2-SCF) specifically in endothelial cells (ECs) after tamoxifen treatment. These mice and a control group (wild-type VE-Cadherin CreERT2-WT) were subjected to hindlimb ischemia or aortic crush to evaluate perfusion/arteriogenesis and endothelial barrier permeability, respectively. RESULTS Our data confirmed the lower gene expression of c-Kit and SCF in the ECs of c-Kit and SCF mice, respectively. In addition, we confirmed the lower percentage of ECs positive for c-Kit in c-Kit mice. Further, we found that c-Kit and SCF mice had better limb perfusion and arteriogenesis compared to WT mice. We also demonstrated that c-Kit and SCF mice had a preserved endothelial barrier after aortic crush compared to WT. CONCLUSIONS Our data demonstrate the deleterious effects of endothelial SCF/c-Kit signaling on arteriogenesis and endothelial barrier integrity.
Collapse
Affiliation(s)
- Gustavo Falero-Diaz
- Department of Surgery, Miller School of Medicine, University of Miami, 1600 NW 10th Ave, RMSB, Miami, FL 33136, USA; (G.F.-D.); (R.I.V.-P.); (O.C.V.)
| | - Catarina de A. Barboza
- Department of Medicine, Miller School of Medicine, University of Miami, 1580 NW 10th Ave, Batchelor Building, Miami, FL 33136, USA;
| | - Roberto I. Vazquez-Padron
- Department of Surgery, Miller School of Medicine, University of Miami, 1600 NW 10th Ave, RMSB, Miami, FL 33136, USA; (G.F.-D.); (R.I.V.-P.); (O.C.V.)
| | - Omaida C. Velazquez
- Department of Surgery, Miller School of Medicine, University of Miami, 1600 NW 10th Ave, RMSB, Miami, FL 33136, USA; (G.F.-D.); (R.I.V.-P.); (O.C.V.)
| | - Roberta M. Lassance-Soares
- Department of Surgery, Miller School of Medicine, University of Miami, 1600 NW 10th Ave, RMSB, Miami, FL 33136, USA; (G.F.-D.); (R.I.V.-P.); (O.C.V.)
| |
Collapse
|
4
|
SDF-1α-Releasing Microspheres Effectively Extend Stem Cell Homing after Myocardial Infarction. Biomedicines 2023; 11:biomedicines11020343. [PMID: 36830880 PMCID: PMC9953248 DOI: 10.3390/biomedicines11020343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 01/20/2023] [Accepted: 01/24/2023] [Indexed: 01/27/2023] Open
Abstract
Ischemic heart disease (IHD) is one of the main focuses in today's healthcare due to its implications and complications, and it is predicted to be increasing in prevalence due to the ageing population. Although the conventional pharmacological and interventional methods for the treatment of IHD presents with success in the clinical setting, the long-term complications of cardiac insufficiency are on a continual incline as a result of post-infarction remodeling of the cardiac tissue. The migration and involvement of stem cells to the cardiac muscle, followed by differentiation into cardiac myocytes, has been proven to be the natural process, though at a slow rate. SDF-1α is a novel candidate to mobilize stem cells homing to the ischemic heart. Endogenous SDF-1α levels are elevated after myocardial infarction, but their presence gradually decreases after approximately seven days. Additional administration of SDF-1α-releasing microspheres could be a tool for the extension of the time the stem cells are in the cardiac tissue after myocardial infarction. This, in turn, could constitute a novel therapy for more efficient regeneration of the heart muscle after injury. Through this practical study, it has been shown that the controlled release of SDF-1α from biodegradable microspheres into the pericardial sac fourteen days after myocardial infarction increases the concentration of exogenous SDF-1α, which persists in the tissue much longer than the level of endogenous SDF-1α. In addition, administration of SDF-1α-releasing microspheres increased the expression of the factors potentially involved in the involvement and retention of myocardial stem cells, which constitutes vascular endothelial growth factor A (VEGFA), stem cell factor (SCF), and vascular cell adhesion molecules (VCAMs) at the site of damaged tissue. This exhibits the possibility of combating the basic limitations of cell therapy, including ineffective stem cell implantation and the ability to induce the migration of endogenous stem cells to the ischemic cardiac tissue and promote heart repair.
Collapse
|
5
|
Rossignol P, Duarte K, Bresso E, A Å, Devignes MD, Eriksson N, Girerd N, Glerup R, Jardine AG, Holdaas H, Lamiral Z, Leroy C, Massy Z, März W, Krämer B, Wu PH, Schmieder R, Soveri I, Christensen JH, Svensson M, Zannad F, Fellström B. NT-proBNP and stem cell factor plasma concentrations are independently associated with cardiovascular outcomes in end-stage renal disease hemodialysis patients. EUROPEAN HEART JOURNAL OPEN 2022; 2:oeac069. [PMID: 36600882 PMCID: PMC9797490 DOI: 10.1093/ehjopen/oeac069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 09/14/2022] [Accepted: 10/24/2022] [Indexed: 11/11/2022]
Abstract
Aims End-stage renal disease (ESRD) treated by chronic hemodialysis (HD) is associated with poor cardiovascular (CV) outcomes, with no available evidence-based therapeutics. A multiplexed proteomic approach may identify new pathophysiological pathways associated with CV outcomes, potentially actionable for precision medicine. Methods and results The AURORA trial was an international, multicentre, randomized, double-blind trial involving 2776 patients undergoing maintenance HD. Rosuvastatin vs. placebo had no significant effect on the composite primary endpoint of death from CV causes, nonfatal myocardial infarction or nonfatal stroke. We first compared CV risk-matched cases and controls (n = 410) to identify novel biomarkers using a multiplex proximity extension immunoassay (276 proteomic biomarkers assessed with OlinkTM). We replicated our findings in 200 unmatched cases and 200 controls. External validation was conducted from a multicentre real-life Danish cohort [Aarhus-Aalborg (AA), n = 331 patients] in which 92 OlinkTM biomarkers were assessed. In AURORA, only N-terminal pro-brain natriuretic peptide (NT-proBNP, positive association) and stem cell factor (SCF) (negative association) were found consistently associated with the trial's primary outcome across exploration and replication phases, independently from the baseline characteristics. Stem cell factor displayed a lower added predictive ability compared with NT-ProBNP. In the AA cohort, in multivariable analyses, BNP was found significantly associated with major CV events, while higher SCF was associated with less frequent CV deaths. Conclusions Our findings suggest that NT-proBNP and SCF may help identify ESRD patients with respectively high and low CV risk, beyond classical clinical predictors and also point at novel pathways for prevention and treatment.
Collapse
Affiliation(s)
- P Rossignol
- Corresponding author. Tel: +33383157322, Fax: +33383157324,
| | - K Duarte
- Université de Lorraine, Inserm, Centre d’Investigations Cliniques- 1433, and Inserm U1116, CHRU Nancy, F-CRIN INI-CRCT, 4, rue du Morvan, 54500 Nancy, France
| | - E Bresso
- Université de Lorraine, Inserm, Centre d’Investigations Cliniques- 1433, and Inserm U1116, CHRU Nancy, F-CRIN INI-CRCT, 4, rue du Morvan, 54500 Nancy, France,LORIA (CNRS, Inria NGE, Université de Lorraine), F-CRIN INI-CRCT, Vandœuvre-lès-Nancy, France
| | - Åsberg A
- Department of Transplantation Medicine Oslo University Hospital–Rikshospitalet, Oslo, Norway,Norway and Department of Pharmaceutical Biosciences, School of Pharmacy, University of Oslo, Oslo, Norway
| | - M D Devignes
- LORIA (CNRS, Inria NGE, Université de Lorraine), F-CRIN INI-CRCT, Vandœuvre-lès-Nancy, France
| | - N Eriksson
- UCR Uppsala Clinical Research Center, Uppsala Science Park, Uppsala, Sweden
| | - N Girerd
- Université de Lorraine, Inserm, Centre d’Investigations Cliniques- 1433, and Inserm U1116, CHRU Nancy, F-CRIN INI-CRCT, 4, rue du Morvan, 54500 Nancy, France
| | - R Glerup
- Department of Nephrology, Aalborg University Hospital, Aalborg, Denmark
| | - A G Jardine
- Renal Research Group, British Heart Foundation Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | | | - Z Lamiral
- Université de Lorraine, Inserm, Centre d’Investigations Cliniques- 1433, and Inserm U1116, CHRU Nancy, F-CRIN INI-CRCT, 4, rue du Morvan, 54500 Nancy, France
| | - C Leroy
- Université de Lorraine, Inserm, Centre d’Investigations Cliniques- 1433, and Inserm U1116, CHRU Nancy, F-CRIN INI-CRCT, 4, rue du Morvan, 54500 Nancy, France
| | - Z Massy
- CESP, Center for Research in Epidemiology and Population Health, University Paris-Saclay, University Paris-Sud, UVSQ, Villejuif, France,Division of Nephrology, Ambroise Paré University Hospital, APHP, Boulogne, Billancourt and FCRIN INI-CRCT, Paris, France
| | - W März
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria,Mannheim Institute of Public Health, Social and Preventive Medicine, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany,SYNLAB Academy, SYNLAB Holding Deutschland GmbH, Mannheim and Augsburg, Germany
| | - B Krämer
- Medical Clinic V, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - P H Wu
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden,Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - R Schmieder
- Department of Nephrology and Hypertension, University Hospital Erlangen, Erlangen, Germany
| | - I Soveri
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - J H Christensen
- Department of Nephrology, Aalborg University Hospital, Aalborg, Denmark
| | - M Svensson
- Department of Nephrology, Aarhus University Hospital, Aarhus, Denmark
| | | | | |
Collapse
|
6
|
Li H, Wu F, Huang G, Wu D, Wang T, Wang X, Wang K, Feng Y, Xu A. Cardiomyocytes induced from hiPSCs by well-defined compounds have therapeutic potential in heart failure by secreting PDGF-BB. Signal Transduct Target Ther 2022; 7:253. [PMID: 35902567 PMCID: PMC9334380 DOI: 10.1038/s41392-022-01045-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 04/27/2022] [Accepted: 05/30/2022] [Indexed: 01/14/2023] Open
Abstract
Recent studies have suggested that transplant of hiPS-CMs is a promising approach for treating heart failure. However, the optimally clinical benefits have been hampered by the immature nature of the hiPS-CMs, and the hiPS-CMs-secreted proteins contributing to the repair of cardiomyocytes remain largely unidentified. Here, we established a saponin+ compound optimally induced system to generate hiPS-CMs with stable functional attributes in vitro and transplanted in heart failure mice. Our study showed enhanced therapeutic effects of optimally induced hiPS-CMs by attenuating cardiac remodeling and dysfunction, these beneficial effects were concomitant with reduced cardiomyocytes death and increased angiogenesis. Moreover, the optimally induced hiPS-CMs could gathering to the injured heart and secret an abundant PDGF-BB. The reparative effect of the optimally induced hiPS-CMs in the hypoxia-injured HCMs was mimicked by PDGF-BB but inhibited by PDGF-BB neutralizing antibody, which was accompanied by the changed expression of p-PI3K and p-Akt proteins. It is highly possible that the PI3K/Akt pathway is regulated by the PDGF-BB secreted from the compound induced hiPS-CMs to achieve a longer lasting myocardial repair effect compared with the standard induced hiPS-CMs. Taken together, our data strongly implicate that the compound induced hiPS-CMs promote the recovery of injured hearts via paracrine action. In this process, the paracrine factor PDGF-BB derived from the compound induced hiPS-CMs reduces isoproterenol-induced adverse cardiac remodeling, which is associated with improved cardiac function, and these effects are mediated by the PI3K/Akt pathway, suggesting that the optimally induced hiPS-CMs may serve as a new promising cell therapy for clinical applications.
Collapse
Affiliation(s)
- Hongmei Li
- School of Life Science, Beijing University of Chinese Medicine, Beijing, China.,Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Fenfang Wu
- Shenzhen Hospital, Beijing University of Chinese Medicine, Shenzhen, China
| | - Guangrui Huang
- School of Life Science, Beijing University of Chinese Medicine, Beijing, China
| | - Di Wu
- College of Life Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Ting Wang
- School of Life Science, Beijing University of Chinese Medicine, Beijing, China
| | - Xiashuang Wang
- College of Life Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Kai Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yuyin Feng
- School of Life Science, Beijing University of Chinese Medicine, Beijing, China
| | - Anlong Xu
- School of Life Science, Beijing University of Chinese Medicine, Beijing, China. .,College of Life Sciences, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
7
|
Omatsu-Kanbe M, Fukunaga R, Mi X, Matsuura H. Atypically Shaped Cardiomyocytes (ACMs): The Identification, Characterization and New Insights into a Subpopulation of Cardiomyocytes. Biomolecules 2022; 12:biom12070896. [PMID: 35883452 PMCID: PMC9313223 DOI: 10.3390/biom12070896] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/17/2022] [Accepted: 06/24/2022] [Indexed: 02/01/2023] Open
Abstract
In the adult mammalian heart, no data have yet shown the existence of cardiomyocyte-differentiable stem cells that can be used to practically repair the injured myocardium. Atypically shaped cardiomyocytes (ACMs) are found in cultures of the cardiomyocyte-removed fraction obtained from cardiac ventricles from neonatal to aged mice. ACMs are thought to be a subpopulation of cardiomyocytes or immature cardiomyocytes, most closely resembling cardiomyocytes due to their spontaneous beating, well-organized sarcomere and the expression of cardiac-specific proteins, including some fetal cardiac gene proteins. In this review, we focus on the characteristics of ACMs compared with ventricular myocytes and discuss whether these cells can be substitutes for damaged cardiomyocytes. ACMs reside in the interstitial spaces among ventricular myocytes and survive under severely hypoxic conditions fatal to ventricular myocytes. ACMs have not been observed to divide or proliferate, similar to cardiomyocytes, but they maintain their ability to fuse with each other. Thus, it is worthwhile to understand the role of ACMs and especially how these cells perform cell fusion or function independently in vivo. It may aid in the development of new approaches to cell therapy to protect the injured heart or the clarification of the pathogenesis underlying arrhythmia in the injured heart.
Collapse
|
8
|
Wu PH, Glerup RI, Svensson MHS, Eriksson N, Christensen JH, de Laval P, Soveri I, Westerlund M, Linde T, Ljunggren Ö, Fellström B. Novel Biomarkers Detected by Proteomics Predict Death and Cardiovascular Events in Hemodialysis Patients. Biomedicines 2022; 10:biomedicines10040740. [PMID: 35453489 PMCID: PMC9026983 DOI: 10.3390/biomedicines10040740] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 03/06/2022] [Accepted: 03/16/2022] [Indexed: 11/16/2022] Open
Abstract
End-stage kidney disease increases mortality and the risk of cardiovascular (CV) disease. It is crucial to explore novel biomarkers to predict CV disease in the complex setting of patients receiving hemodialysis (HD). This study investigated the association between 92 targeted proteins with all-cause death, CV death, and composite vascular events (CVEs) in HD patients. From December 2010 to March 2011, 331 HD patients were included and followed prospectively for 5 years. Serum was analyzed for 92 CV-related proteins using Proseek Multiplex Cardiovascular I panel, a high-sensitivity assay based on proximity extension assay (PEA) technology. The association between biomarkers and all-cause death, CV death, and CVEs was evaluated using Cox-regression analyses. Of the PEA-based proteins, we identified 20 proteins associated with risk of all-cause death, 7 proteins associated with risk of CV death, and 17 proteins associated with risk of CVEs, independent of established risk factors. Interleukin-8 (IL-8), T-cell immunoglobulin and mucin domain 1 (TIM-1), and C-C motif chemokine 20 (CCL20) were associated with increased risk of all-cause death, CV death, and CVE in multivariable-adjusted models. Stem cell factor (SCF) and Galanin peptides (GAL) were associated with both decreased risk of all-cause death and CV death. In conclusion, IL-8, TIM-1, and CCL20 predicted death and CV outcomes in HD patients. Novel findings were that SCF and GAL were associated with a lower risk of all-cause death and CV death. The SCF warrants further study with regard to its possible biological effect in HD patients.
Collapse
Affiliation(s)
- Ping-Hsun Wu
- Department of Medical Sciences, Uppsala University, 75236 Uppsala, Sweden; (P.-H.W.); (P.d.L.); (I.S.); (M.W.); (T.L.); (Ö.L.)
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 80708, Taiwan
- Center for Big Data Research, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Rie Io Glerup
- Department of Nephrology, Aalborg University Hospital, 9000 Aalborg, Denmark; (R.I.G.); (J.H.C.)
| | - My Hanna Sofia Svensson
- Division of Medicine, Department of Nephrology, Akershus University Hospital, 1478 Oslo, Norway;
| | - Niclas Eriksson
- Uppsala Clinical Research Center, Uppsala University, 75185 Uppsala, Sweden;
| | | | - Philip de Laval
- Department of Medical Sciences, Uppsala University, 75236 Uppsala, Sweden; (P.-H.W.); (P.d.L.); (I.S.); (M.W.); (T.L.); (Ö.L.)
| | - Inga Soveri
- Department of Medical Sciences, Uppsala University, 75236 Uppsala, Sweden; (P.-H.W.); (P.d.L.); (I.S.); (M.W.); (T.L.); (Ö.L.)
| | - Magnus Westerlund
- Department of Medical Sciences, Uppsala University, 75236 Uppsala, Sweden; (P.-H.W.); (P.d.L.); (I.S.); (M.W.); (T.L.); (Ö.L.)
| | - Torbjörn Linde
- Department of Medical Sciences, Uppsala University, 75236 Uppsala, Sweden; (P.-H.W.); (P.d.L.); (I.S.); (M.W.); (T.L.); (Ö.L.)
| | - Östen Ljunggren
- Department of Medical Sciences, Uppsala University, 75236 Uppsala, Sweden; (P.-H.W.); (P.d.L.); (I.S.); (M.W.); (T.L.); (Ö.L.)
| | - Bengt Fellström
- Department of Medical Sciences, Uppsala University, 75236 Uppsala, Sweden; (P.-H.W.); (P.d.L.); (I.S.); (M.W.); (T.L.); (Ö.L.)
- Correspondence: ; Tel.: +46-18-6114348
| |
Collapse
|
9
|
Schmidt IM, Sarvode Mothi S, Wilson PC, Palsson R, Srivastava A, Onul IF, Kibbelaar ZA, Zhuo M, Amodu A, Stillman IE, Rennke HG, Humphreys BD, Waikar SS. Circulating Plasma Biomarkers in Biopsy-Confirmed Kidney Disease. Clin J Am Soc Nephrol 2022; 17:27-37. [PMID: 34759008 PMCID: PMC8763150 DOI: 10.2215/cjn.09380721] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 11/02/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND AND OBJECTIVES Biomarkers for noninvasive assessment of histopathology and prognosis are needed in patients with kidney disease. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS Using a proteomics assay, we measured a multimarker panel of 225 circulating plasma proteins in a prospective cohort study of 549 individuals with biopsy-confirmed kidney diseases and semiquantitative assessment of histopathology. We tested the associations of each biomarker with histopathologic lesions and the risks of kidney disease progression (defined as ≥40% decline in eGFR or initiation of KRT) and death. RESULTS After multivariable adjustment and correction for multiple testing, 46 different proteins were associated with histopathologic lesions. The top-performing markers positively associated with acute tubular injury and interstitial fibrosis/tubular atrophy were kidney injury molecule-1 (KIM-1) and V-set and Ig domain-containing protein 2 (VSIG2), respectively. Thirty proteins were significantly associated with kidney disease progression, and 35 were significantly associated with death. The top-performing markers for kidney disease progression were placental growth factor (hazard ratio per doubling, 5.4; 95% confidence interval, 3.4 to 8.7) and BMP and activin membrane-bound inhibitor (hazard ratio, 3.0; 95% confidence interval, 2.1 to 4.2); the top-performing markers for death were TNF-related apoptosis-inducing ligand receptor-2 (hazard ratio, 2.9; 95% confidence interval, 2.0 to 4.0) and CUB domain-containing protein-1 (hazard ratio, 2.4; 95% confidence interval, 1.8 to 3.3). CONCLUSION We identified several plasma protein biomarkers associated with kidney disease histopathology and adverse clinical outcomes in individuals with a diverse set of kidney diseases. PODCAST This article contains a podcast at https://www.asn-online.org/media/podcast/CJASN/2021_12_28_CJN09380721.mp3.
Collapse
Affiliation(s)
- Insa M. Schmidt
- Section of Nephrology, Department of Medicine, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts
- Renal Division, Brigham & Women’s Hospital, Boston, Massachusetts
| | - Suraj Sarvode Mothi
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Parker C. Wilson
- Department of Pathology and Immunology, Washington University, St. Louis, Missouri
| | - Ragnar Palsson
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Anand Srivastava
- Division of Nephrology and Hypertension, Center for Translational Metabolism and Health, Institute for Public Health and Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Ingrid F. Onul
- Section of Nephrology, Department of Medicine, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts
- Renal Division, Brigham & Women’s Hospital, Boston, Massachusetts
| | - Zoe A. Kibbelaar
- Section of Nephrology, Department of Medicine, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts
- Renal Division, Brigham & Women’s Hospital, Boston, Massachusetts
| | - Min Zhuo
- Renal Division, Brigham & Women’s Hospital, Boston, Massachusetts
- Division of Nephrology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Afolarin Amodu
- Section of Nephrology, Department of Medicine, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts
- Renal Division, Brigham & Women’s Hospital, Boston, Massachusetts
| | - Isaac E. Stillman
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Helmut G. Rennke
- Department of Pathology, Brigham & Women’s Hospital, Boston, Massachusetts
| | - Benjamin D. Humphreys
- Division of Nephrology, Department of Medicine, Washington University, St. Louis, Missouri
| | - Sushrut S. Waikar
- Section of Nephrology, Department of Medicine, Boston University School of Medicine, Boston Medical Center, Boston, Massachusetts
- Renal Division, Brigham & Women’s Hospital, Boston, Massachusetts
| |
Collapse
|
10
|
Huizer K, Sacchetti A, Swagemakers S, van der Spek PJ, Dik W, Mustafa DA, Kros JM. Circulating angiogenic cells in glioblastoma: toward defining crucial functional differences in CAC-induced neoplastic versus reactive neovascularization. Neurooncol Adv 2020; 2:vdaa040. [PMID: 32642695 PMCID: PMC7276933 DOI: 10.1093/noajnl/vdaa040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Background In order to identify suitable therapeutic targets for glioma anti-angiogenic therapy, the process of neovascularization mediated by circulating angiogenic cells (CACs) needs to be scrutinized. Methods In the present study, we compared the expression of neovascularization-related genes by 3 circulating CAC subsets (hematopoietic progenitor cells [HPCs], CD34+, and KDR+ cells; internal controls: peripheral blood mononuclear cells and circulating endothelial cells) of treatment-naïve patients with glioblastoma (GBM) to those of patients undergoing reactive neovascularization (myocardial infarction (MI). CACs from umbilical cord (representing developmental neovascularization) and healthy subjects served as controls. Fluorescent-activated cell sorting was used to isolate CACs, RT-PCR to determine the expression levels of a panel of 48 neovascularization-related genes, and Luminex assays to measure plasma levels of 21 CAC-related circulating molecules. Results We found essential differences in gene expression between GBM and MI CACs. GBM CACs had a higher expression of proangiogenic factors (especially, KITL, CXCL12, and JAG1), growth factor and chemotactic receptors (IGF1R, TGFBR2, CXCR4, and CCR2), adhesion receptor monomers (ITGA5 and ITGA6), and matricellular factor POSTN. In addition, we found major differences in the levels of neovascularization-related plasma factors. A strong positive correlation between plasma MMP9 levels and expression of CXCR4 in the CAC subset of HPCs was found in GBM patients. Conclusions Our findings indicate that CAC-mediated neovascularization in GBM is characterized by more efficient CAC homing to target tissue and a more potent proangiogenic response than in physiologic tissue repair in MI. Our findings can aid in selecting targets for therapeutic strategies acting against GBM-specific CACs.
Collapse
Affiliation(s)
- Karin Huizer
- Laboratory for Tumor Immuno-Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Andrea Sacchetti
- Laboratory for Tumor Immuno-Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Sigrid Swagemakers
- Department of Pathology and Clinical Bio-Informatics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Peter J van der Spek
- Department of Pathology and Clinical Bio-Informatics, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Wim Dik
- Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Dana A Mustafa
- Laboratory for Tumor Immuno-Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Johan M Kros
- Laboratory for Tumor Immuno-Pathology, Erasmus Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
11
|
Pawitan JA, Margiana R, Aman RA, Jusuf AA, Ibrahim N, Wibowo H. The effect of human umbilical cord-derived mesenchymal stem cell conditioned medium on the peripheral nerve regeneration of injured rats. ELECTRONIC JOURNAL OF GENERAL MEDICINE 2019. [DOI: 10.29333/ejgm/115468] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
12
|
Popielarczyk TL, Huckle WR, Barrett JG. Human Bone Marrow-Derived Mesenchymal Stem Cells Home via the PI3K-Akt, MAPK, and Jak/Stat Signaling Pathways in Response to Platelet-Derived Growth Factor. Stem Cells Dev 2019; 28:1191-1202. [PMID: 31190615 DOI: 10.1089/scd.2019.0003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) have great potential to improve clinical outcomes for many inflammatory and degenerative diseases either through intravenously delivered MSCs or through mobilization and migration of endogenous MSCs to injury sites, termed "stem cell homing." Stem cell homing involves the processes of attachment to and transmigration through endothelial cells lining the vasculature and migration through the tissue stroma to a site of injury or inflammation. Although the process of leukocyte transendothelial migration (TEM) is well understood, far less is known about stem cell homing. In this study, a transwell-based model was developed to monitor adherence and TEM of human MSCs in response to chemokine exposure. Specifically, transwell membranes lined with human synovial microvascular endothelial cells were partitioned from the tissue injury-mimetic site containing chemokine stromal cell-derived factor-1 (SDF-1). Two population subsets of MSCs were studied: migratory cells that initiated transmigration on the endothelial lining and nonmigratory cells. We hypothesized that cells would adhere to and migrate through the endothelial lining in response to SDF-1 exposure and that gene and protein expression changes would be observed between migratory and nonmigratory cells. We validated a vasculature model for MSC transmigration that showed increased expression of several genes and activation of proteins of the PI3K-Akt, MAPK, and Jak/Stat signaling pathways. These findings showed that MSC homing may be driven by activation of PDGFRA/PI3K/Akt, PDGFRA/MAPK/Grb2, and PDGFRA/Jak2/Stat signaling, as a result of SDF-1-stimulated endothelial cell production of platelet-derived growth factor. This model can be used to further investigate these key regulatory molecules toward the development of targeted therapies.
Collapse
Affiliation(s)
- Tracee L Popielarczyk
- Department of Large Animal Clinical Sciences, Marion duPont Scott Equine Medical Center, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Leesburg, Virginia
| | - William R Huckle
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia
| | - Jennifer G Barrett
- Department of Large Animal Clinical Sciences, Marion duPont Scott Equine Medical Center, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Leesburg, Virginia
| |
Collapse
|
13
|
Legere SA, Haidl ID, Légaré JF, Marshall JS. Mast Cells in Cardiac Fibrosis: New Insights Suggest Opportunities for Intervention. Front Immunol 2019; 10:580. [PMID: 31001246 PMCID: PMC6455071 DOI: 10.3389/fimmu.2019.00580] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 03/04/2019] [Indexed: 12/19/2022] Open
Abstract
Mast cells (MC) are innate immune cells present in virtually all body tissues with key roles in allergic disease and host defense. MCs recognize damage-associated molecular patterns (DAMPs) through expression of multiple receptors including Toll-like receptors and the IL-33 receptor ST2. MCs can be activated to degranulate and release pre-formed mediators, to synthesize and secrete cytokines and chemokines without degranulation, and/or to produce lipid mediators. MC numbers are generally increased at sites of fibrosis. They are potent, resident, effector cells producing mediators that regulate the fibrotic process. The nature of the secretory products produced by MCs depend on micro-environmental signals and can be both pro- and anti-fibrotic. MCs have been repeatedly implicated in the pathogenesis of cardiac fibrosis and in angiogenic responses in hypoxic tissues, but these findings are controversial. Several rodent studies have indicated a protective role for MCs. MC-deficient mice have been reported to have poorer outcomes after coronary artery ligation and increased cardiac function upon MC reconstitution. In contrast, MCs have also been implicated as key drivers of fibrosis. MC stabilization during a hypertensive rat model and an atrial fibrillation mouse model rescued associated fibrosis. Discrepancies in the literature could be related to problems with mouse models of MC deficiency. To further complicate the issue, mice generally have a much lower density of MCs in their cardiac tissue than humans, and as such comparing MC deficient and MC containing mouse models is not necessarily reflective of the role of MCs in human disease. In this review, we will evaluate the literature regarding the role of MCs in cardiac fibrosis with an emphasis on what is known about MC biology, in this context. MCs have been well-studied in allergic disease and multiple pharmacological tools are available to regulate their function. We will identify potential opportunities to manipulate human MC function and the impact of their mediators with a view to preventing or reducing harmful fibrosis. Important therapeutic opportunities could arise from increased understanding of the impact of such potent, resident immune cells, with the ability to profoundly alter long term fibrotic processes.
Collapse
Affiliation(s)
- Stephanie A. Legere
- Departments of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Ian D. Haidl
- Departments of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Jean-François Légaré
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
- Department of Surgery, Dalhousie Medicine New Brunswick, Saint John, NB, Canada
| | - Jean S. Marshall
- Departments of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
- Department of Pathology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
14
|
Choe G, Park J, Jo H, Kim YS, Ahn Y, Lee JY. Studies on the effects of microencapsulated human mesenchymal stem cells in RGD-modified alginate on cardiomyocytes under oxidative stress conditions using in vitro biomimetic co-culture system. Int J Biol Macromol 2018; 123:512-520. [PMID: 30445088 DOI: 10.1016/j.ijbiomac.2018.11.115] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 10/16/2018] [Accepted: 11/12/2018] [Indexed: 12/20/2022]
Abstract
Stem cell therapy has been recognized as a promising approach for myocardium regeneration post myocardial infarction (MI); however, it unfortunately often remains a challenge because of poor survival of transplanted cells and a lack of clear understanding of their interactions with host cells. High oxidative stress at heart tissues post MI is considered one of the important factors damaging transplanted cells and native cells/tissues. Here, we employed an in vitro co-culture system, capable of mimicking cases of stem cell transplantation into the myocardium presenting high oxidative stress, using human mesenchymal stem cells (hMSCs) encapsulated in alginate or cell interactive Arg-Gly-Asp (RGD) peptide-modified alginate micro-hydrogels. Under H2O2-induced oxidative stress conditions, viabilities of hMSCs and CMs were significantly higher in their co-culture than in their individual monolayer cultures. Expression of cardiac muscle markers remained high even with H2O2 treatment when cardiomyocytes (CMs) were co-cultured with hMSCs in RGD-alginate. Higher levels of various growth factors (associated with angiogenesis, cardiac regeneration, and contractility) were found in co-culture (noticeably with RGD-alginate) compared to monolayer cultures of CMs or hMSCs. These results can benefit the study of in vivo MI progression with transplanted stem cells and the development of effective stem cell-based therapeutic strategies for various oxidative stress-related diseases.
Collapse
Affiliation(s)
- Goeun Choe
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Junggeon Park
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Hyerim Jo
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea
| | - Yong Sook Kim
- Biomedical Research Institute, Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| | - Youngkeun Ahn
- Department of Cardiology, Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| | - Jae Young Lee
- School of Materials Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea; Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology (GIST), Gwangju 61005, Republic of Korea.
| |
Collapse
|
15
|
Wang Y, Xia Y, Kuang D, Duan Y, Wang G. PP2A regulates SCF-induced cardiac stem cell migration through interaction with p38 MAPK. Life Sci 2017; 191:59-67. [DOI: 10.1016/j.lfs.2017.10.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 09/22/2017] [Accepted: 10/02/2017] [Indexed: 12/29/2022]
|
16
|
Björkbacka H, Yao Mattisson I, Wigren M, Melander O, Fredrikson GN, Bengtsson E, Gonçalves I, Almgren P, Lagerstedt JO, Orho-Melander M, Engström G, Nilsson J. Plasma stem cell factor levels are associated with risk of cardiovascular disease and death. J Intern Med 2017; 282:508-521. [PMID: 28842933 DOI: 10.1111/joim.12675] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Stem cell factor (SCF) is a key growth factor for several types of stem and progenitor cells. There is experimental evidence that such cells are of importance for maintaining the integrity of the cardiovascular system. We investigated the association between circulating levels of SCF and risk for development of cardiovascular events and death. METHODS SCF was analysed by the proximity extension assay technique in plasma from 4742 subjects participating in the Malmö Diet and Cancer Study. Cardiovascular events and death were monitored through national registers with a mean follow-up time of 19.2 years. RESULTS Subjects with high baseline levels of SCF had lower cardiovascular (n = 340) and all-cause mortality (n = 1159) as well as a lower risk of heart failure (n = 177), stroke (n = 318) and myocardial infarction (n = 452). Smoking, diabetes and high alcohol consumption were associated with lower levels of SCF. Single nucleotide polymorphisms in the gene region encoding PDX1 C-terminal inhibiting factor 1 (PCIF1) and matrix metalloproteinase-9 were associated with plasma SCF levels. The highest SCF quartile remained independently associated with a lower risk of a lower risk of cardiovascular [hazard ratio and 95% confidence interval 0.59 (0.43-0.81)] and all-cause mortality [0.68 (0.57-0.81)], heart failure [0.50 (0.31-0.80)] and stroke [0.66 (0.47-0.92)], but not with MI [0.96 (0.72-1.27)] as compared with the lowest quartile when adjusting for traditional cardiovascular risk factors in Cox proportional hazard regression models. CONCLUSIONS This prospective population-based study demonstrates that subjects with high levels of SCF have a lower risk of cardiovascular events and death. The findings provide clinical support for a protective role of SCF in maintaining cardiovascular integrity.
Collapse
Affiliation(s)
- H Björkbacka
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - I Yao Mattisson
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - M Wigren
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - O Melander
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - G N Fredrikson
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - E Bengtsson
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - I Gonçalves
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden.,Department of Cardiology - Coronary diseases, Skåne University Hospital, Malmö, Sweden
| | - P Almgren
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - J O Lagerstedt
- Department of Experimental Medical Science, Lund University, Malmö, Sweden
| | - M Orho-Melander
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - G Engström
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| | - J Nilsson
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden
| |
Collapse
|
17
|
Omatsu-Kanbe M, Nozuchi N, Nishino Y, Mukaisho KI, Sugihara H, Matsuura H. Identification of cardiac progenitors that survive in the ischemic human heart after ventricular myocyte death. Sci Rep 2017; 7:41318. [PMID: 28120944 PMCID: PMC5264617 DOI: 10.1038/srep41318] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 12/19/2016] [Indexed: 12/21/2022] Open
Abstract
Atypically-shaped cardiomyocytes (ACMs) are beating heart cells identified in the cultures of cardiomyocyte-removed fractions obtained from adult mouse hearts. Since ACMs spontaneously develop into beating cells in the absence of hormones or chemicals, these cells are likely to be a type of cardiac progenitors rather than stem cells. “Native ACMs” are found as small interstitial cells among ventricular myocytes that co-express cellular prion protein (PrP) and cardiac troponin T (cTnT) in mouse and human heart tissues. However, the endogenous behavior of human ACMs is unclear. In the present study, we demonstrate that PrP+ cTnT+ cells are present in the human heart tissue with myocardial infarction (MI). These cells were mainly found in the border of necrotic cardiomyocytes caused by infarcts and also in the hibernating myocardium subjected to the chronic ischemia. The ratio of PrP+ cTnT+ cells to the total cells observed in the normal heart tissue section of mouse and human was estimated to range from 0.3–0.8%. Notably, living human PrP+ cTnT+ cells were identified in the cultures obtained at pathological autopsy despite exposure to lethal ischemic conditions for hours after death. These findings suggest that ACMs could survive in the ischemic human heart and develop into a sub-population of cardiac myocytes.
Collapse
Affiliation(s)
- Mariko Omatsu-Kanbe
- Department of Physiology, Shiga University of Medical Science, Otsu, Shiga, 520-2192, Japan
| | - Nozomi Nozuchi
- Department of Physiology, Shiga University of Medical Science, Otsu, Shiga, 520-2192, Japan
| | - Yuka Nishino
- Department of Physiology, Shiga University of Medical Science, Otsu, Shiga, 520-2192, Japan
| | - Ken-Ichi Mukaisho
- Department of Pathology, Shiga University of Medical Science, Otsu, Shiga, 520-2192, Japan
| | - Hiroyuki Sugihara
- Department of Pathology, Shiga University of Medical Science, Otsu, Shiga, 520-2192, Japan
| | - Hiroshi Matsuura
- Department of Physiology, Shiga University of Medical Science, Otsu, Shiga, 520-2192, Japan
| |
Collapse
|
18
|
Ruangsawasdi N, Zehnder M, Patcas R, Ghayor C, Siegenthaler B, Gjoksi B, Weber FE. Effects of Stem Cell Factor on Cell Homing During Functional Pulp Regeneration in Human Immature Teeth. Tissue Eng Part A 2016; 23:115-123. [PMID: 27762658 DOI: 10.1089/ten.tea.2016.0227] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Conventional root canal treatment in immature permanent teeth can lead to early tooth loss in children because root formation is discontinued. We investigated whether the stem cell factor (SCF) could facilitate cell homing in the pulpless immature root canal and promote regeneration of a functional pulp. In vitro, human mesenchymal stem cells (hMSCs) were exposed to SCF at various concentrations for assessing cell migration, proliferation, and differentiation toward odonto/osteoblasts by 3D-chemotaxis slides, WST-1 assay, and alkaline phosphatase activity, respectively. Fibrin gels were used to deliver 15 μg/mL SCF for in vivo experiments. The release kinetic of SCF was assessed in vitro. Two corresponding human immature premolars, with or without SCF, were placed at rat calvariae for 6 and 12 weeks. All tooth specimens were either analyzed histologically and the percentage of tissue ingrowth determined or the cells were extracted from the pulp space, and the mRNA level of DMP1, DSPP, Col1, NGF, and VEGF were assessed by quantitative polymerase chain reaction. In the presence of SCF, we saw an increase in hMSCs directional migration, proliferation, and odonto/osteogenic differentiation. SCF also increased the extent of tissue ingrowth at 6 weeks but not at 12 weeks. However, at this time point, the formed tissue appeared more mature in samples with SCF. In terms of gene transcription, DMP1, Col1, and VEGF were the significantly upregulated genes, while DSPP and NGF were not affected. Our results suggest that SCF can accelerate cell homing and the maturation of the pulp-dentin complex in human immature teeth.
Collapse
Affiliation(s)
- Nisarat Ruangsawasdi
- 1 Oral Biotechnology and Bioengineering, Center of Dental Medicine, University of Zürich , Zürich, Switzerland .,2 Zurich Center for Integrative Human Physiology (ZIHP), University of Zürich , Zürich, Switzerland .,3 Department of Pharmacology, Faculty of Dentistry, Mahidol University , Bangkok, Thailand
| | - Matthias Zehnder
- 4 Clinic for Preventive Dentistry, Periodontology, and Cariology, University of Zürich , Zürich, Switzerland
| | - Raphael Patcas
- 5 Clinic for Orthodontics and Pediatric Dentistry, Center of Dental Medicine, University of Zürich , Zürich, Switzerland
| | - Chafik Ghayor
- 1 Oral Biotechnology and Bioengineering, Center of Dental Medicine, University of Zürich , Zürich, Switzerland
| | - Barbara Siegenthaler
- 1 Oral Biotechnology and Bioengineering, Center of Dental Medicine, University of Zürich , Zürich, Switzerland .,2 Zurich Center for Integrative Human Physiology (ZIHP), University of Zürich , Zürich, Switzerland
| | - Bebeka Gjoksi
- 1 Oral Biotechnology and Bioengineering, Center of Dental Medicine, University of Zürich , Zürich, Switzerland
| | - Franz E Weber
- 1 Oral Biotechnology and Bioengineering, Center of Dental Medicine, University of Zürich , Zürich, Switzerland .,2 Zurich Center for Integrative Human Physiology (ZIHP), University of Zürich , Zürich, Switzerland
| |
Collapse
|
19
|
Di Siena S, Gimmelli R, Nori SL, Barbagallo F, Campolo F, Dolci S, Rossi P, Venneri MA, Giannetta E, Gianfrilli D, Feigenbaum L, Lenzi A, Naro F, Cianflone E, Mancuso T, Torella D, Isidori AM, Pellegrini M. Activated c-Kit receptor in the heart promotes cardiac repair and regeneration after injury. Cell Death Dis 2016; 7:e2317. [PMID: 27468693 PMCID: PMC4973348 DOI: 10.1038/cddis.2016.205] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 06/01/2016] [Accepted: 06/03/2016] [Indexed: 12/20/2022]
Abstract
The role of endogenous c-Kit receptor activation on cardiac cell homeostasis and repair remains largely unexplored. Transgenic mice carrying an activating point mutation (TgD814Y) in the kinase domain of the c-Kit gene were generated. c-KitTgD814Y receptor was expressed in the heart during embryonic development and postnatal life, in a similar timing and expression pattern to that of the endogenous gene, but not in the hematopoietic compartment allowing the study of a cardiac-specific phenotype. c-KitTgD814Y mutation produced a constitutive active c-Kit receptor in cardiac tissue and cells from transgenic mice as demonstrated by the increased phosphorylation of ERK1/2 and AKT, which are the main downstream molecular effectors of c-Kit receptor signaling. In adult transgenic hearts, cardiac morphology, size and total c-Kit+ cardiac cell number was not different compared with wt mice. However, when c-KitTgD814Y mice were subjected to transmural necrotic heart damage by cryoinjury (CI), all transgenic survived, compared with half of wt mice. In the sub-acute phase after CI, transgenic and wt mice showed similar heart damage. However, 9 days after CI, transgenic mice exhibited an increased number of c-Kit+CD31+ endothelial progenitor cells surrounding the necrotic area. At later follow-up, a consistent reduction of fibrotic area, increased capillary density and increased cardiomyocyte replenishment rate (as established by BrdU incorporation) were observed in transgenic compared with wt mice. Consistently, CD45−c-Kit+ cardiac stem cells isolated from transgenic c-KitTgD814Y mice showed an enhanced endothelial and cardiomyocyte differentiation potential compared with cells isolated from the wt. Constitutive activation of c-Kit receptor in mice is associated with an increased cardiac myogenic and vasculogenic reparative potential after injury, with a significant improvement of survival.
Collapse
Affiliation(s)
- S Di Siena
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University, Rome, Italy
| | - R Gimmelli
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - S L Nori
- Department of Medicine and Surgery, University of Salerno, Baronissi, Italy
| | - F Barbagallo
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - F Campolo
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - S Dolci
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - P Rossi
- Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - M A Venneri
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - E Giannetta
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - D Gianfrilli
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - L Feigenbaum
- Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer research, Frederick, MD, USA
| | - A Lenzi
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - F Naro
- Department of Anatomical, Histological, Forensic and Orthopedic Sciences, Sapienza University, Rome, Italy
| | - E Cianflone
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - T Mancuso
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - D Torella
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - A M Isidori
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - M Pellegrini
- Institute of Cell Biology and Neurobiology, CNR, Rome, Italy
| |
Collapse
|
20
|
Cho H, Balaji S, Hone NL, Moles CM, Sheikh AQ, Crombleholme TM, Keswani SG, Narmoneva DA. Diabetic wound healing in a MMP9-/- mouse model. Wound Repair Regen 2016; 24:829-840. [PMID: 27292154 DOI: 10.1111/wrr.12453] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 05/12/2016] [Indexed: 12/12/2022]
Abstract
Reduced mobilization of endothelial progenitor cells (EPCs) from the bone marrow (BM) and impaired EPC recruitment into the wound represent a fundamental deficiency in the chronic ulcers. However, mechanistic understanding of the role of BM-derived EPCs in cutaneous wound neovascularization and healing remains incomplete, which impedes development of EPC-based wound healing therapies. The objective of this study was to determine the role of EPCs in wound neovascularization and healing both under normal conditions and using single deficiency (EPC) or double-deficiency (EPC + diabetes) models of wound healing. MMP9 knockout (MMP9 KO) mouse model was utilized, where impaired EPC mobilization can be rescued by stem cell factor (SCF). The hypotheses were: (1) MMP9 KO mice exhibit impaired wound neovascularization and healing, which are further exacerbated with diabetes; (2) these impairments can be rescued by SCF administration. Full-thickness excisional wounds with silicone splints to minimize contraction were created on MMP9 KO mice with/without streptozotocin-induced diabetes in the presence or absence of tail-vein injected SCF. Wound morphology, vascularization, inflammation, and EPC mobilization and recruitment were quantified at day 7 postwounding. Results demonstrate no difference in wound closure and granulation tissue area between any groups. MMP9 deficiency significantly impairs wound neovascularization, increases inflammation, decreases collagen deposition, and decreases peripheral blood EPC (pb-EPC) counts when compared with wild-type (WT). Diabetes further increases inflammation, but does not cause further impairment in vascularization, as compared with MMP9 KO group. SCF improves neovascularization and increases EPCs to WT levels (both nondiabetic and diabetic MMP9 KO groups), while exacerbating inflammation in all groups. SCF rescues EPC-deficiency and impaired wound neovascularization in both diabetic and nondiabetic MMP9 KO mice. Overall, the results demonstrate that BM-derived EPCs play a significant role during wound neovascularization and that the SCF-based therapy with controlled inflammation could be a viable approach to enhance healing in chronic diabetic wounds.
Collapse
Affiliation(s)
- Hongkwan Cho
- Department of Biomedical, Chemical and Environmental Engineering, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, Ohio
| | - Swathi Balaji
- Department of Biomedical, Chemical and Environmental Engineering, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, Ohio.,Division of Pediatric Surgery, Baylor College of Medicine, Houston, Texas
| | - Natalie L Hone
- Department of Biomedical, Chemical and Environmental Engineering, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, Ohio
| | - Chad M Moles
- Division of Pediatric Surgery, Baylor College of Medicine, Houston, Texas
| | - Abdul Q Sheikh
- Department of Biomedical, Chemical and Environmental Engineering, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, Ohio
| | - Timothy M Crombleholme
- Children's Hospital Colorado and the University of Colorado School of Medicine, Aurora, Colorado
| | - Sundeep G Keswani
- Division of Pediatric Surgery, Baylor College of Medicine, Houston, Texas
| | - Daria A Narmoneva
- Department of Biomedical, Chemical and Environmental Engineering, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, Ohio.
| |
Collapse
|
21
|
In Vivo Tracking of Cell Therapies for Cardiac Diseases with Nuclear Medicine. Stem Cells Int 2016; 2016:3140120. [PMID: 26880951 PMCID: PMC4737458 DOI: 10.1155/2016/3140120] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Revised: 10/18/2015] [Accepted: 10/20/2015] [Indexed: 12/31/2022] Open
Abstract
Even though heart diseases are amongst the main causes of mortality and morbidity in the world, existing treatments are limited in restoring cardiac lesions. Cell transplantations, originally developed for the treatment of hematologic ailments, are presently being explored in preclinical and clinical trials for cardiac diseases. Nonetheless, little is known about the possible efficacy and mechanisms for these therapies and they are the center of continuous investigation. In this scenario, noninvasive imaging techniques lead to greater comprehension of cell therapies. Radiopharmaceutical cell labeling, firstly developed to track leukocytes, has been used successfully to evaluate the migration of cell therapies for myocardial diseases. A substantial rise in the amount of reports employing this methodology has taken place in the previous years. We will review the diverse radiopharmaceuticals, imaging modalities, and results of experimental and clinical studies published until now. Also, we report on current limitations and potential advances of radiopharmaceutical labeling for cell therapies in cardiac diseases.
Collapse
|
22
|
Vajravelu BN, Hong KU, Al-Maqtari T, Cao P, Keith MCL, Wysoczynski M, Zhao J, Moore IV JB, Bolli R. C-Kit Promotes Growth and Migration of Human Cardiac Progenitor Cells via the PI3K-AKT and MEK-ERK Pathways. PLoS One 2015; 10:e0140798. [PMID: 26474484 PMCID: PMC4608800 DOI: 10.1371/journal.pone.0140798] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 09/29/2015] [Indexed: 01/01/2023] Open
Abstract
A recent phase I clinical trial (SCIPIO) has shown that autologous c-kit+ cardiac progenitor cells (CPCs) improve cardiac function and quality of life when transplanted into patients with ischemic heart disease. Although c-kit is widely used as a marker of resident CPCs, its role in the regulation of the cellular characteristics of CPCs remains unknown. We hypothesized that c-kit plays a role in the survival, growth, and migration of CPCs. To test this hypothesis, human CPCs were grown under stress conditions in the presence or absence of SCF, and the effects of SCF-mediated activation of c-kit on CPC survival/growth and migration were measured. SCF treatment led to a significant increase in cell survival and a reduction in cell death under serum depletion conditions. In addition, SCF significantly promoted CPC migration in vitro. Furthermore, the pro-survival and pro-migratory effects of SCF were augmented by c-kit overexpression and abrogated by c-kit inhibition with imatinib. Mechanistically, c-kit activation in CPCs led to activation of the PI3K and the MAPK pathways. With the use of specific inhibitors, we confirmed that the SCF/c-kit-dependent survival and chemotaxis of CPCs are dependent on both pathways. Taken together, our findings suggest that c-kit promotes the survival/growth and migration of human CPCs cultured ex vivo via the activation of PI3K and MAPK pathways. These results imply that the efficiency of CPC homing to the injury site as well as their survival after transplantation may be improved by modulating the activity of c-kit.
Collapse
Affiliation(s)
- Bathri N. Vajravelu
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY 40202, United States of America
| | - Kyung U. Hong
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY 40202, United States of America
| | - Tareq Al-Maqtari
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY 40202, United States of America
| | - Pengxiao Cao
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY 40202, United States of America
| | - Matthew C. L. Keith
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY 40202, United States of America
| | - Marcin Wysoczynski
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY 40202, United States of America
| | - John Zhao
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY 40202, United States of America
| | - Joseph B. Moore IV
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY 40202, United States of America
| | - Roberto Bolli
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, Louisville, KY 40202, United States of America
- * E-mail:
| |
Collapse
|
23
|
Lee SH, Lee KB, Lee JH, Kang S, Kim HG, Asahara T, Kwon SM. Selective Interference Targeting of Lnk in Umbilical Cord-Derived Late Endothelial Progenitor Cells Improves Vascular Repair, Following Hind Limb Ischemic Injury, via Regulation of JAK2/STAT3 Signaling. Stem Cells 2015; 33:1490-500. [DOI: 10.1002/stem.1938] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 11/28/2014] [Indexed: 12/27/2022]
Affiliation(s)
- Sang Hun Lee
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital; Seoul South Korea
- Department of Biochemistry; School of Medicine, Soonchunhyang University; Cheonan South Korea
- Laboratory for Vascular Medicine and Stem Cell Biology; Department of Physiology; School of Medicine; Pusan National University; Medical Research Institute, School of Medicine, Pusan National University; Yangsan Gyeongnam South Korea
| | - Kyeung Bin Lee
- Laboratory for Vascular Medicine and Stem Cell Biology; Department of Physiology; School of Medicine; Pusan National University; Medical Research Institute, School of Medicine, Pusan National University; Yangsan Gyeongnam South Korea
| | - Jun Hee Lee
- Laboratory for Vascular Medicine and Stem Cell Biology; Department of Physiology; School of Medicine; Pusan National University; Medical Research Institute, School of Medicine, Pusan National University; Yangsan Gyeongnam South Korea
- Convergence Stem Cell Research Center, Immunoregulatory Therapeutics Group in Brain Busan 21 Project; Pusan National University, Yangsan Gyeongnam South Korea
| | - Songhwa Kang
- Laboratory for Vascular Medicine and Stem Cell Biology; Department of Physiology; School of Medicine; Pusan National University; Medical Research Institute, School of Medicine, Pusan National University; Yangsan Gyeongnam South Korea
- Convergence Stem Cell Research Center, Immunoregulatory Therapeutics Group in Brain Busan 21 Project; Pusan National University, Yangsan Gyeongnam South Korea
| | - Hwi Gon Kim
- Department of Obstetrics and Gynecology; Pusan National University, School of Medicine; Busan South Korea
| | - Takayuki Asahara
- Department of Regenerative Medicine Science; Tokai University School of Medicine; Shimokasuya Isehara Kanagawa Japan
| | - Sang Mo Kwon
- Laboratory for Vascular Medicine and Stem Cell Biology; Department of Physiology; School of Medicine; Pusan National University; Medical Research Institute, School of Medicine, Pusan National University; Yangsan Gyeongnam South Korea
- Convergence Stem Cell Research Center, Immunoregulatory Therapeutics Group in Brain Busan 21 Project; Pusan National University, Yangsan Gyeongnam South Korea
| |
Collapse
|
24
|
Ishikawa K, Fish K, Aguero J, Yaniz-Galende E, Jeong D, Kho C, Tilemann L, Fish L, Liang L, Eltoukhy AA, Anderson DG, Zsebo K, Costa KD, Hajjar RJ. Stem cell factor gene transfer improves cardiac function after myocardial infarction in swine. Circ Heart Fail 2015; 8:167-74. [PMID: 25342737 PMCID: PMC4303518 DOI: 10.1161/circheartfailure.114.001711] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 10/21/2014] [Indexed: 11/16/2022]
Abstract
BACKGROUND Stem cell factor (SCF), a ligand of the c-kit receptor, is a critical cytokine, which contributes to cell migration, proliferation, and survival. It has been shown that SCF expression increases after myocardial infarction (MI) and may be involved in cardiac repair. The aim of this study was to determine whether gene transfer of membrane-bound human SCF improves cardiac function in a large animal model of MI. METHODS AND RESULTS A transmural MI was created by implanting an embolic coil in the left anterior descending artery in Yorkshire pigs. One week after the MI, the pigs received direct intramyocardial injections of either a recombinant adenovirus encoding for SCF (Ad.SCF, n=9) or β-gal (Ad.β-gal, n=6) into the infarct border area. At 3 months post-MI, ejection fraction increased by 12% relative to baseline after Ad.SCF therapy, whereas it decreased by 4.2% (P=0.004) in pigs treated with Ad.β-gal. Preload-recruitable stroke work was significantly higher in pigs after SCF treatment (Ad.SCF, 55.5±11.6 mm Hg versus Ad.β-gal, 31.6±12.6 mm Hg, P=0.005), indicating enhanced cardiac function. Histological analyses confirmed the recruitment of c-kit(+) cells as well as a reduced degree of apoptosis 1 week after Ad.SCF injection. In addition, increased capillary density compared with pigs treated with Ad.β-gal was found at 3 months and suggests an angiogenic role of SCF. CONCLUSIONS Local overexpression of SCF post-MI induces the recruitment of c-kit(+) cells at the infarct border area acutely. In the chronic stages, SCF gene transfer was associated with improved cardiac function in a preclinical model of ischemic cardiomyopathy.
Collapse
Affiliation(s)
- Kiyotake Ishikawa
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (K.I., K.F., J.A., E.Y.-G., D.J., C.K., L.T., L.F., L.L., K.D.C., R.J.H.); Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)-Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain (J.A.); David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge (A.A.E., D.G.A.); and Celladon Corporation, San Diego, CA (K.Z.).
| | - Kenneth Fish
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (K.I., K.F., J.A., E.Y.-G., D.J., C.K., L.T., L.F., L.L., K.D.C., R.J.H.); Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)-Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain (J.A.); David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge (A.A.E., D.G.A.); and Celladon Corporation, San Diego, CA (K.Z.)
| | - Jaume Aguero
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (K.I., K.F., J.A., E.Y.-G., D.J., C.K., L.T., L.F., L.L., K.D.C., R.J.H.); Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)-Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain (J.A.); David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge (A.A.E., D.G.A.); and Celladon Corporation, San Diego, CA (K.Z.)
| | - Elisa Yaniz-Galende
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (K.I., K.F., J.A., E.Y.-G., D.J., C.K., L.T., L.F., L.L., K.D.C., R.J.H.); Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)-Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain (J.A.); David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge (A.A.E., D.G.A.); and Celladon Corporation, San Diego, CA (K.Z.)
| | - Dongtak Jeong
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (K.I., K.F., J.A., E.Y.-G., D.J., C.K., L.T., L.F., L.L., K.D.C., R.J.H.); Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)-Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain (J.A.); David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge (A.A.E., D.G.A.); and Celladon Corporation, San Diego, CA (K.Z.)
| | - Changwon Kho
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (K.I., K.F., J.A., E.Y.-G., D.J., C.K., L.T., L.F., L.L., K.D.C., R.J.H.); Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)-Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain (J.A.); David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge (A.A.E., D.G.A.); and Celladon Corporation, San Diego, CA (K.Z.)
| | - Lisa Tilemann
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (K.I., K.F., J.A., E.Y.-G., D.J., C.K., L.T., L.F., L.L., K.D.C., R.J.H.); Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)-Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain (J.A.); David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge (A.A.E., D.G.A.); and Celladon Corporation, San Diego, CA (K.Z.)
| | - Lauren Fish
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (K.I., K.F., J.A., E.Y.-G., D.J., C.K., L.T., L.F., L.L., K.D.C., R.J.H.); Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)-Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain (J.A.); David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge (A.A.E., D.G.A.); and Celladon Corporation, San Diego, CA (K.Z.)
| | - Lifan Liang
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (K.I., K.F., J.A., E.Y.-G., D.J., C.K., L.T., L.F., L.L., K.D.C., R.J.H.); Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)-Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain (J.A.); David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge (A.A.E., D.G.A.); and Celladon Corporation, San Diego, CA (K.Z.)
| | - Ahmed A Eltoukhy
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (K.I., K.F., J.A., E.Y.-G., D.J., C.K., L.T., L.F., L.L., K.D.C., R.J.H.); Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)-Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain (J.A.); David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge (A.A.E., D.G.A.); and Celladon Corporation, San Diego, CA (K.Z.)
| | - Daniel G Anderson
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (K.I., K.F., J.A., E.Y.-G., D.J., C.K., L.T., L.F., L.L., K.D.C., R.J.H.); Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)-Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain (J.A.); David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge (A.A.E., D.G.A.); and Celladon Corporation, San Diego, CA (K.Z.)
| | - Krisztina Zsebo
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (K.I., K.F., J.A., E.Y.-G., D.J., C.K., L.T., L.F., L.L., K.D.C., R.J.H.); Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)-Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain (J.A.); David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge (A.A.E., D.G.A.); and Celladon Corporation, San Diego, CA (K.Z.)
| | - Kevin D Costa
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (K.I., K.F., J.A., E.Y.-G., D.J., C.K., L.T., L.F., L.L., K.D.C., R.J.H.); Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)-Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain (J.A.); David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge (A.A.E., D.G.A.); and Celladon Corporation, San Diego, CA (K.Z.)
| | - Roger J Hajjar
- From the Cardiovascular Research Center, Icahn School of Medicine at Mount Sinai, New York, NY (K.I., K.F., J.A., E.Y.-G., D.J., C.K., L.T., L.F., L.L., K.D.C., R.J.H.); Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC)-Epidemiology, Atherothrombosis and Imaging Department, Madrid, Spain (J.A.); David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge (A.A.E., D.G.A.); and Celladon Corporation, San Diego, CA (K.Z.)
| |
Collapse
|
25
|
Guo J, Jie W, Shen Z, Li M, Lan Y, Kong Y, Guo S, Li T, Zheng S. SCF increases cardiac stem cell migration through PI3K/AKT and MMP‑2/‑9 signaling. Int J Mol Med 2014; 34:112-8. [PMID: 24804928 PMCID: PMC4072340 DOI: 10.3892/ijmm.2014.1773] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Accepted: 04/29/2014] [Indexed: 12/15/2022] Open
Abstract
The transplantation of cardiac stem cells (CSCs) is thought to be responsible for improving the performance of injured heart induced by myocardial infarction (MI). However, the mechanisms involved in the migration of activated CSCs post-MI remain to be clarified. In this study, CSCs were isolated from rat hearts and a cellular migration assay was performed using a 24-well Transwell system. Stem cell factor (SCF) induced CSC migration in a concentration-dependent manner, which could be blocked with an SCF antibody as well as a PI3K/AKT inhibitor, LY294002. Moreover, SCF induced the expression and activity of matrix metalloproteinase (MMP)-2 and MMP-9 in a concentration- and time-dependent manner, as measured by quantitative RT-PCR, western blot analysis and gelatin zymography. Results of western blot analysis revealed phosphorylated AKT was markedly increased in SCF-treated CSCs and that inhibition of SCF/c-Kit signaling or phospho-AKT activity significantly attenuated the SCF-induced expression of MMP-2 and MMP-9. Thus, our results showed that SCF partially mediated CSC migration via the activation of PI3K/AKT/MMP-2/-9 signaling.
Collapse
Affiliation(s)
- Junli Guo
- Cardiovascular Institute of Affiliated Hospital, Hainan Medical College, Haikou 571199, P.R. China
| | - Wei Jie
- Department of Pathology, School of Basic Medicine Science, Guangdong Medical College, Zhanjiang 524023, P.R. China
| | - Zhihua Shen
- Department of Pathology, School of Basic Medicine Science, Guangdong Medical College, Zhanjiang 524023, P.R. China
| | - Mengsen Li
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical College, Haikou 571199, P.R. China
| | - Youling Lan
- Cardiovascular Institute of Affiliated Hospital, Hainan Medical College, Haikou 571199, P.R. China
| | - Yueqiong Kong
- Cardiovascular Institute of Affiliated Hospital, Hainan Medical College, Haikou 571199, P.R. China
| | - Shaoli Guo
- Cardiovascular Institute of Affiliated Hospital, Hainan Medical College, Haikou 571199, P.R. China
| | - Tianfa Li
- Cardiovascular Institute of Affiliated Hospital, Hainan Medical College, Haikou 571199, P.R. China
| | - Shaojiang Zheng
- Cardiovascular Institute of Affiliated Hospital, Hainan Medical College, Haikou 571199, P.R. China
| |
Collapse
|
26
|
Ale A, Siebenhaar F, Kosanke K, Aichler M, Radrich K, Heydrich S, Schiemann M, Bielicki I, Noel PB, Braren R, Maurer M, Walch AK, Rummeny EJ, Ntziachristos V, Wildgruber M. Cardioprotective C-kit⁺ bone marrow cells attenuate apoptosis after acute myocardial infarction in mice - in-vivo assessment with fluorescence molecular imaging. Theranostics 2013; 3:903-13. [PMID: 24312159 PMCID: PMC3841340 DOI: 10.7150/thno.5938] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 04/28/2013] [Indexed: 11/05/2022] Open
Abstract
Cardiomyocyte loss via apoptosis plays a crucial role in ventricular remodeling following myocardial infarction (MI). Cell-based therapy approaches using bone marrow derived c-kit⁺ pluripotent cells may attenuate apoptosis following ischemic injury. We therefore thought to examine the early course of apoptosis following myocardial infarction - in-vivo - and non-invasively determine the effect of c-kit⁺ bone marrow cells on post-MI remodeling. We studied apoptosis in wild-type Kit(+/+) , c-kit mutant Kit(W)/Kit(W-v) and Kit(W)/Kit(W-v) mice after cell therapy with bone-marrow derived c-kit⁺ cells after ischemia-reperfusion injury. Mice were followed by hybrid Fluorescence Molecular Tomography/X-ray Computed Tomography (FMT-XCT) at 6h, 24h and 7 days after ischemia-reperfusion injury using an Annexin V-based fluorescent nanosensor targeting phosphatidylserine. Kit(W)/Kit(W-v) mice showed increased and prolonged apoptosis compared to control Kit(+/+) mice while c-kit cell therapy was able to attenuate the altered apoptosis rates. Increased apoptosis was accompanied by severe decline in heart function, determined by cardiac Magnetic Resonance Imaging, and cell therapy was able to rescue the animals from deleterious heart failure. Post-mortem cryoslicing and immunohistochemistry localized the fluorescence signal of the Annexin V sensor within the infarcted myocardium. Flow cytometry of digested infarct specimens identified apoptotic cardiomyocytes as the major source for the in-vivo Annexin V signal. In-vivo molecular imaging using hybrid FMT-XCT reveals increased cardiomyocyte apoptosis in Kit(W)/Kit(W-v) mice and shows that c-kit⁺ cardioprotective cells are able to attenuate post-MI apoptosis and rescue mice from progressive heart failure.
Collapse
|
27
|
Affiliation(s)
- Maurilio Sampaolesi
- Laboratory of Translational Cardiomyology, Department of Development and Regeneration, KU Leuven, Belgium
| | | |
Collapse
|
28
|
Omatsu-Kanbe M, Matsuura H. Ischemic survival and constitutively active autophagy in self-beating atypically-shaped cardiomyocytes (ACMs): characterization of a new subpopulation of heart cells. J Physiol Sci 2013; 63:17-29. [PMID: 23055023 PMCID: PMC10717659 DOI: 10.1007/s12576-012-0236-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Accepted: 09/18/2012] [Indexed: 11/29/2022]
Abstract
Atypically-shaped cardiomyocytes (ACMs) are a new subpopulation of spontaneously beating heart cells with a peculiar morphology identified within a culture of cardiac myocyte-depleted fraction (CMDF) cells obtained from adult mouse heart. ACMs originate from small cells in CMDF and grow in size and start beating within ~3 days culture without appreciable proliferation or express stem cell marker proteins, but stay in the heart until elderly stages. However, the characteristics of ACMs are largely unclear. The present study examined whether pre-exposure of CMDF cells to severe ischemia abolished the ability of ACMs to develop into beating cells. Of ACMs that underwent ischemia, ~50 % grew in size, changed the morphology, and started beating during the subsequent culture under normoxia. ACMs displayed constitutively active autophagy during the culture. The results suggest the possibility that the development of beating ACMs could occur in injured heart, even if the surviving cell population is small.
Collapse
Affiliation(s)
- Mariko Omatsu-Kanbe
- Department of Physiology, Shiga University of Medical Science, Otsu, Shiga, 520-2192, Japan.
| | - Hiroshi Matsuura
- Department of Physiology, Shiga University of Medical Science, Otsu, Shiga, 520-2192, Japan
| |
Collapse
|
29
|
Sun Z, Lee CJ, Mejia-Guerrero S, Zhang Y, Higuchi K, Li RK, Medin JA. Neonatal Transfer of Membrane-Bound Stem Cell Factor Improves Survival and Heart Function in Aged Mice After Myocardial Ischemia. Hum Gene Ther 2012; 23:1280-9. [DOI: 10.1089/hum.2012.063] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Zhuo Sun
- University Health Network, Toronto, ON M5G 1L7, Canada
- Division of Cardiac Surgery, Department of Surgery, University of Toronto, Toronto, ON M5G 1X8, Canada
| | | | | | - Yuemei Zhang
- University Health Network, Toronto, ON M5G 1L7, Canada
- Division of Cardiac Surgery, Department of Surgery, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Koji Higuchi
- University Health Network, Toronto, ON M5G 1L7, Canada
| | - Ren-Ke Li
- University Health Network, Toronto, ON M5G 1L7, Canada
- Division of Cardiac Surgery, Department of Surgery, University of Toronto, Toronto, ON M5G 1X8, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jeffrey A. Medin
- University Health Network, Toronto, ON M5G 1L7, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 2M9, Canada
| |
Collapse
|
30
|
Wen Z, Mai Z, Zhang H, Chen Y, Geng D, Zhou S, Wang J. Local activation of cardiac stem cells for post-myocardial infarction cardiac repair. J Cell Mol Med 2012; 16:2549-63. [PMID: 22613044 PMCID: PMC4118225 DOI: 10.1111/j.1582-4934.2012.01589.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2012] [Accepted: 05/08/2012] [Indexed: 12/23/2022] Open
Abstract
The prognosis of patients with myocardial infarction (MI) and resultant chronic heart failure remains extremely poor despite continuous advancements in optimal medical therapy and interventional procedures. Animal experiments and clinical trials using adult stem cell therapy following MI have shown a global improvement of myocardial function. The emergence of stem cell transplantation approaches has recently represented promising alternatives to stimulate myocardial regeneration. Regarding their tissue-specific properties, cardiac stem cells (CSCs) residing within the heart have advantages over other stem cell types to be the best cell source for cell transplantation. However, time-consuming and costly procedures to expanse cells prior to cell transplantation and the reliability of cell culture and expansion may both be major obstacles in the clinical application of CSC-based transplantation therapy after MI. The recognition that the adult heart possesses endogenous CSCs that can regenerate cardiomyocytes and vascular cells has raised the unique therapeutic strategy to reconstitute dead myocardium via activating these cells post-MI. Several strategies, such as growth factors, mircoRNAs and drugs, may be implemented to potentiate endogenous CSCs to repair infarcted heart without cell transplantation. Most molecular and cellular mechanism involved in the process of CSC-based endogenous regeneration after MI is far from understanding. This article reviews current knowledge opening up the possibilities of cardiac repair through CSCs activation in situ in the setting of MI.
Collapse
Affiliation(s)
- Zhuzhi Wen
- Sun Yat-sen Memorial Hospital of Sun Yat-sen UniversityGuangzhou, China
| | - Zun Mai
- Sun Yat-sen Memorial Hospital of Sun Yat-sen UniversityGuangzhou, China
| | - Haifeng Zhang
- Sun Yat-sen Memorial Hospital of Sun Yat-sen UniversityGuangzhou, China
| | - Yangxin Chen
- Sun Yat-sen Memorial Hospital of Sun Yat-sen UniversityGuangzhou, China
| | - Dengfeng Geng
- Sun Yat-sen Memorial Hospital of Sun Yat-sen UniversityGuangzhou, China
| | - Shuxian Zhou
- Sun Yat-sen Memorial Hospital of Sun Yat-sen UniversityGuangzhou, China
| | - Jingfeng Wang
- Sun Yat-sen Memorial Hospital of Sun Yat-sen UniversityGuangzhou, China
| |
Collapse
|
31
|
She T, Wang X, Gan Y, Kuang D, Yue J, Ni J, Zhao X, Wang G. Hyperglycemia suppresses cardiac stem cell homing to peri-infarcted myocardium via regulation of ERK1/2 and p38 MAPK activities. Int J Mol Med 2012; 30:1313-20. [PMID: 22965067 DOI: 10.3892/ijmm.2012.1125] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Accepted: 07/23/2012] [Indexed: 11/05/2022] Open
Abstract
Hyperglycemia in the acute phase of myocardial infarction (MI) is a marker of worse prognosis in both diabetic and non-diabetic patients; however, the role of hyperglycemia in the homing of cardiac stem cells (CSCs) to damaged myocardium post-MI and the possible mechanisms involved are not well understood. In this study, an MI model was induced in normoglycemic and hyperglycemic rats by left coronary artery ligation. Immunofluorescence was used to examine the migration of CSCs in vivo by injecting BrdU-labeled CSCs into the atrium-ventricle groove (AV-groove). Immunohistochemistry, western blot analysis and ELISA were carried out to detect the expression of stem cell factor (SCF) protein and RT-PCR was conducted for the expression of SCF mRNA. Phosphorylation of ERK1/2 and p38 MAPK was detected by western blot analysis. Afterwards, cardiac function was evaluated by hemodynamic measurement. On Day 5 post-MI, the accumulation of CSCs significantly increased in the peri-infarcted myocardium in normoglycemic rats, which led to an improvement in cardiac function 3 weeks after MI. However, the accumulation of CSCs markedly decreased in hyperglycemic rats, followed by the decline of cardiac function. SCF expression, followed with phosphorylation of ERK1/2 and p38 MAPK, were also significantly downregulated in the peri-infarcted myocardium in hyperglycemic rats compared to normoglycemic rats. Moreover, SCF expression and the migration of CSCs were blocked by either the MEK-specific inhibitor PD98059 or the p38 MAPK-selective inhibitor SB203580. The experiments in vitro confirmed that hyperglycemia decreased SCF expression via reduction in ERK1/2 and p38 MAPK activities and further inhibited the migration of CSCs. The results suggest that hyperglycemia suppresses CSC migration towards the ischemic area post-MI. This is possibly due to decreased myocardial SCF expression via reduction of ERK1/2 and p38 MAPK activities in hyperglycemic rats.
Collapse
Affiliation(s)
- Tonghui She
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Yaniz-Galende E, Chen J, Chemaly E, Liang L, Hulot JS, McCollum L, Arias T, Fuster V, Zsebo KM, Hajjar RJ. Stem cell factor gene transfer promotes cardiac repair after myocardial infarction via in situ recruitment and expansion of c-kit+ cells. Circ Res 2012; 111:1434-45. [PMID: 22931954 DOI: 10.1161/circresaha.111.263830] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
RATIONALE There is growing evidence that the myocardium responds to injury by recruiting c-kit(+) cardiac progenitor cells to the damage tissue. Even though the ability of exogenously introducing c-kit(+) cells to injured myocardium has been established, the capability of recruiting these cells through modulation of local signaling pathways by gene transfer has not been tested. OBJECTIVE To determine whether stem cell factor gene transfer mediates cardiac regeneration in a rat myocardial infarction model, through survival and recruitment of c-kit(+) progenitors and cell-cycle activation in cardiomyocytes, and explore the mechanisms involved. METHODS AND RESULTS Infarct size, cardiac function, cardiac progenitor cells recruitment, fibrosis, and cardiomyocyte cell-cycle activation were measured at different time points in controls (n=10) and upon stem cell factor gene transfer (n=13) after myocardial infarction. We found a regenerative response because of stem cell factor overexpression characterized by an enhancement in cardiac hemodynamic function: an improvement in survival; a reduction in fibrosis, infarct size and apoptosis; an increase in cardiac c-kit(+) progenitor cells recruitment to the injured area; an increase in cardiomyocyte cell-cycle activation; and Wnt/β-catenin pathway induction. CONCLUSIONS Stem cell factor gene transfer induces c-kit(+) stem/progenitor cell expansion in situ and cardiomyocyte proliferation, which may represent a new therapeutic strategy to reverse adverse remodeling after myocardial infarction.
Collapse
Affiliation(s)
- Elisa Yaniz-Galende
- Cardiovascular Research Center, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Rosenberg M, Lutz M, Kühl C, Will R, Eckstein V, Krebs J, Katus HA, Frey N. Coculture with hematopoietic stem cells protects cardiomyocytes against apoptosis via paracrine activation of AKT. J Transl Med 2012; 10:115. [PMID: 22672705 PMCID: PMC3408384 DOI: 10.1186/1479-5876-10-115] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Accepted: 06/06/2012] [Indexed: 02/08/2023] Open
Abstract
Background Previous experimental studies concluded that stem cells (SC) may exert their beneficial effects on the ischemic heart by paracrine activation of antiapoptotic pathways. In order to identify potential cardioprotective mediators, we performed a systematic analysis of the differential gene expression of hematopoietic SC after coculture with cardiomyocytes (CM). Methods After 48 h of coculture with neonatal rat ventricular CM (NRVCM), two consecutive cell sorting steps generated a highly purified population of conditioned murine hematopoietic SC (>99%). Next, a genome-wide microarray analysis of cocultured vs. monocultured hematopoietic SC derived from three independent experiments was performed. The analysis of differentially expressed genes was focused on products that are secretable and/or membrane-bound and potentially involved in antiapoptotic signalling. Results We found CCL-12, Macrophage Inhibitory Factor, Fibronectin and connexin 40 significantly upregulated in our coculture model. An ELISA of cell culture supernatants was performed to confirm secretion of candidate genes and showed that coculture supernatants revealed markedly higher CCL-12 concentrations. Moreover, we stimulated NRVCM with concentrated coculture supernatants which resulted in a significant reduction of apoptosis compared to monoculture-derived supernatant. Mechanistically, NRVCMs stimulated with coculture supernatants showed a higher level of AKT-phosphorylation, consistent with enhanced antiapoptotic signaling. Conclusion In summary, our results show that the interaction between hematopoietic SC and NRVCM led to a modified gene expression and induction of antiapoptotic pathways. These findings may thus at least in part explain the cardioprotective effects of hematopoietic SC.
Collapse
Affiliation(s)
- Mark Rosenberg
- Department of Internal Medicine III (Cardiology and Angiology), University Medical Center Schleswig-Holstein, Campus Kiel, Schittenhelmstr, 12, D-24105, Kiel, Germany
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Chaudeurge A, Wilhelm C, Chen-Tournoux A, Farahmand P, Bellamy V, Autret G, Ménager C, Hagège A, Larghéro J, Gazeau F, Clément O, Menasché P. Can Magnetic Targeting of Magnetically Labeled Circulating Cells Optimize Intramyocardial Cell Retention? Cell Transplant 2012; 21:679-91. [DOI: 10.3727/096368911x612440] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Therapeutic intracavitary stem cell infusion currently suffers from poor myocardial homing. We examined whether cardiac cell retention could be enhanced by magnetic targeting of endothelial progenitor cells (EPCs) loaded with iron oxide nanoparticles. EPCs were magnetically labeled with citrate-coated iron oxide nanoparticles. Cell proliferation, migration, and CXCR4 chemokine receptor expression were assessed in different labeling conditions and no adverse effects of the magnetic label were observed. The magnetophoretic mobility of labeled EPCs was determined in vitro, with the same magnet as that subsequently used in vivo. Coronary artery occlusion was induced for 30 min in 36 rats (31 survivors), followed by 20 min of reperfusion. The rats were randomized to receive, during brief aortic cross-clamping, direct intraventricular injection of culture medium ( n = 7) or magnetically labeled EPCs ( n = 24), with ( n = 14) or without ( n = 10) subcutaneous insertion of a magnet over the chest cavity ( n = 14). The hearts were explanted 24 h later and engrafted cells were visualized by magnetic resonance imaging (MRI) of the heart at 1.5 T. Their abundance in the myocardium was also analyzed semiquantitatively by immunofluorescence, and quantitatively by real-time polymerase chain reaction (RT-PCR). Although differences in cell retention between groups failed to be statistically significant using RT-PCR quantification, due to the variability of the animal model, immunostaining showed that the average number of engrafted EPCs was significantly ten times higher with than without magnetic targeting. There was thus a consistent trend favoring the magnet-treated hearts, thereby suggesting magnetic targeting as a potentially new mean of enhancing myocardial homing of intravascularly delivered stem cells. Magnetic targeting has the potential to enhance myocardial retention of intravascularly delivered endothelial progenitor cells.
Collapse
Affiliation(s)
- Aurélie Chaudeurge
- INSERM U633, Laboratory of Surgical Research, Paris, France
- Assistance Publique-Hôpitaux de Paris, Ecole de Chirurgie, Paris, France
| | - Claire Wilhelm
- Laboratoire Matière et Systèmes Complexes MSC, CNRS UMR 7057, Paris, France
- Université Paris-Diderot, Paris, France
| | - Annabel Chen-Tournoux
- INSERM U633, Laboratory of Surgical Research, Paris, France
- Assistance Publique-Hôpitaux de Paris, Ecole de Chirurgie, Paris, France
| | - Patrick Farahmand
- INSERM U633, Laboratory of Surgical Research, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Cardiovascular Surgery, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Valérie Bellamy
- INSERM U633, Laboratory of Surgical Research, Paris, France
- Assistance Publique-Hôpitaux de Paris, Ecole de Chirurgie, Paris, France
| | - Gwennhael Autret
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Cardiovascular Surgery, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- INSERM, U970, Paris Cardiovascular Research Center-PARCC, Paris, France
| | | | - Albert Hagège
- INSERM U633, Laboratory of Surgical Research, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Cardiovascular Surgery, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jerome Larghéro
- Univ Paris 06-CNRS-ESPCI Laboratoire PECSA UMR7195, Paris, France
- University Paris Diderot, Paris, France
| | - Florence Gazeau
- Laboratoire Matière et Systèmes Complexes MSC, CNRS UMR 7057, Paris, France
- Université Paris-Diderot, Paris, France
| | - Olivier Clément
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Cardiovascular Surgery, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
- INSERM, U970, Paris Cardiovascular Research Center-PARCC, Paris, France
| | - Philippe Menasché
- INSERM U633, Laboratory of Surgical Research, Paris, France
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Department of Cardiovascular Surgery, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
35
|
Surgical Therapy of End-Stage Heart Failure: Understanding Cell-Mediated Mechanisms Interacting with Myocardial Damage. Int J Artif Organs 2011; 34:529-45. [DOI: 10.5301/ijao.5000004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2011] [Indexed: 01/19/2023]
Abstract
Worldwide, cardiovascular disease results in an estimated 14.3 million deaths per year, giving rise to an increased demand for alternative and advanced treatment. Current approaches include medical management, cardiac transplantation, device therapy, and, most recently, stem cell therapy. Research into cell-based therapies has shown this option to be a promising alternative to the conventional methods. In contrast to early trials, modern approaches now attempt to isolate specific stem cells, as well as increase their numbers by means of amplifying in a culture environment. The method of delivery has also been improved to minimize the risk of micro-infarcts and embolization, which were often observed after the use of coronary catheterization. The latest approach entails direct, surgical, transepicardial injection of the stem cell mixture, as well as the use of tissue-engineered meshes consisting of embedded progenitor cells.
Collapse
|
36
|
Kim KL, Meng Y, Kim JY, Baek EJ, Suh W. Direct and differential effects of stem cell factor on the neovascularization activity of endothelial progenitor cells. Cardiovasc Res 2011; 92:132-40. [PMID: 21636540 DOI: 10.1093/cvr/cvr161] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
AIMS Previous studies on the role of stem cell factor (SCF) in endothelial progenitor cell (EPC)-mediated neovascularization have focused on the EPC mobilization and homing process. However, the direct effects of SCF on neovascularization activity of EPCs have not been characterized. We sought to determine whether SCF regulates the neovascularization ability of EPCs by comparing its roles in mature endothelial cells. METHODS AND RESULTS In vitro and in vivo assays revealed that SCF substantially increased the neovascularization activity of human EPCs through the c-Kit receptor. Notably, the SCF-induced increase in neovascularization activity was substantially greater in EPCs than that in human umbilical vein endothelial cells (HUVECs). SCF-induced phosphorylation of c-Kit and downstream signalling molecules was consistently found to be more potent and longer-lasting in EPCs than in HUVECs. This high responsiveness of EPCs to SCF was explained by the finding that the cell-surface expression of c-Kit is far higher in EPCs than in HUVECs. A c-Kit promoter assay revealed that the increased expression of c-Kit in EPCs could be attributed to the greater expression of stem cell leukaemia, LIM-only 2, and GATA-binding protein 2. CONCLUSION In addition to its documented role in the mobilization and recruitment of EPCs, our findings show that SCF directly enhances the neovascularization activity of EPCs. Furthermore, the present study provides further evidence that EPCs exhibit differentially greater responsiveness to hypoxia-inducible cytokines, including SCF, than mature endothelial cells, suggesting that EPCs in ischaemic tissues function differently from mature endothelial cells, although they exhibit very similar phenotypes.
Collapse
Affiliation(s)
- Koung Li Kim
- College of Pharmacy, Ajou University, San 5, Woncheon-Dong, Yeongtong-Gu, Suwon 443-749, Korea
| | | | | | | | | |
Collapse
|
37
|
Kwon JS, Kim YS, Cho AS, Cho HH, Kim JS, Hong MH, Jeong SY, Jeong MH, Cho JG, Park JC, Kang JC, Ahn Y. The novel role of mast cells in the microenvironment of acute myocardial infarction. J Mol Cell Cardiol 2011; 50:814-25. [PMID: 21295578 DOI: 10.1016/j.yjmcc.2011.01.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2010] [Revised: 01/25/2011] [Accepted: 01/27/2011] [Indexed: 10/18/2022]
Abstract
Mast cells are multifunctional cells containing various mediators, such as cytokines, tryptase, and histamine, and they have been identified in infarct myocardium. Here, we elucidated the roles of mast cells in a myocardial infarction (MI) rat model. We studied the physiological and functional roles of mast cell granules (MCGs), isolated from rat peritoneal fluid, on endothelial cells, neonatal cardiomyocytes, and infarct heart (1-hour occlusion of left coronary artery followed by reperfusion). The number of mast cells had two peak time points of appearance in the infarct region at 1day and 21days after MI induction in rats (p<0.05 in each compared with sham-operated heart). Simultaneous injection of an optimal dose of MCGs modulated the microenvironment and resulted in the increased infiltration of macrophages and decreased apoptosis of cardiomyocytes without change in the mast cell number in infarct myocardium. Moreover, MCG injection attenuated the progression of MI through angiogenesis and preserved left ventricular function after MI. MCG-treated cardiomyocytes were more resistant to hypoxic injury through phosphorylation of Akt, and MCG-treated endothelial cells showed enhanced migration and tube formation. We have shown that MCGs have novel cardioprotective roles in MI via the prolonged survival of cardiomyocytes and the induction of angiogenesis.
Collapse
Affiliation(s)
- Jin Sook Kwon
- Stem Cell Research Center of Chonnam National University Hospital, Gwangju, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Beohar N, Rapp J, Pandya S, Losordo DW. Rebuilding the damaged heart: the potential of cytokines and growth factors in the treatment of ischemic heart disease. J Am Coll Cardiol 2010; 56:1287-97. [PMID: 20888519 DOI: 10.1016/j.jacc.2010.05.039] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2008] [Revised: 04/21/2010] [Accepted: 05/10/2010] [Indexed: 12/15/2022]
Abstract
Cytokine therapy promises to provide a noninvasive treatment option for ischemic heart disease. Cytokines are thought to influence angiogenesis directly via effects on endothelial cells or indirectly through progenitor cell-based mechanisms or by activating the expression of other angiogenic agents. Several cytokines mobilize progenitor cells from the bone marrow or are involved in the homing of mobilized cells to ischemic tissue. The recruited cells contribute to myocardial regeneration both as a structural component of the regenerating tissue and by secreting angiogenic or antiapoptotic factors, including cytokines. To date, randomized, controlled clinical trials have not reproduced the efficacy observed in pre-clinical and small-scale clinical investigations. Nevertheless, the list of promising cytokines continues to grow, and combinations of cytokines, with or without concurrent progenitor cell therapy, warrant further investigation.
Collapse
|
39
|
Degeorge BR, Rosenberg M, Eckstein V, Gao E, Herzog N, Katus HA, Koch WJ, Frey N, Most P. BMP-2 and FGF-2 synergistically facilitate adoption of a cardiac phenotype in somatic bone marrow c-kit+/Sca-1+ stem cells. Clin Transl Sci 2010; 1:116-25. [PMID: 20443832 DOI: 10.1111/j.1752-8062.2008.00034.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The aim of this study was to explore the effect of bone morphogenetic protein-2 (BMP-2) and fibroblast growth factor-2 (FGF-2)- paracrine factors implicated in both cardiac embryogenesis and cardiac repair following myocardial infarction (MI)-on murine bone marrow stem cell (mBMSC) differentiation in an ex vivo cardiac microenvironment. For this purpose, green fluorescent protein (GFP) expressing hematopoietic lineage negative (lin-) c-kit ligand (c-kit) and stem cell antigen-1 (Sca-1) positive (GFP-lin-/c-kit+/sca+) mBMSC were co-cultured with neonatal rat ventricular cardiomyocytes (NVCMs). GFP+ mBMSC significantly induced the expression of BMP-2 and FGF-2 in NVCMs, and approximately 4% GFP+ mBMSCs could be recovered from the co-culture at day 10. The addition of BMP-2 in concert with FGF-2 significantly enhanced the amount of integrated GFP+ mBMSCs by 5-fold ( approximately 20%), whereas the addition of anti-BMP-2 and/or anti-FGF-2 antibodies completely abolished this effect. An analysis of calcium cycling revealed robust calcium transients in GFP+ mBMSCs treated with BMP-2/FGF-2 compared to untreated co-cultures. BMP-2 and FGF-2 addition led to a significant induction of early (NK2 transcription factor related, locus 5; Nkx2.5, GATA binding protein 4; GATA-4) and late (myosin light chain kinase [MLC-2v], connexin 43 [Cx43]) cardiac marker mRNA expression in mBMSCs following co-culture. In addition, re-cultured fluorescence-activated cell sorting (FACS)-purified BMP-2/FGF-2-treated mBMSCs revealed robust calcium transients in response to electrical field stimulation which were inhibited by the L-type calcium channel (LTCC) inhibitor, nifedipine, and displayed caffeine-sensitive intracellular calcium stores. In summary, our results show that mBMSCs can adopt a functional cardiac phenotype through treatment with factors essential to embryonic cardiogenesis that are induced after cardiac ischemia. This study provides the first evidence that mBMSCs with long-term self-renewal potential possess the capability to serve as a functional cardiomyocyte precursor through the appropriate paracrine input and cross-talk within an appropriate cardiac microenvironment.
Collapse
Affiliation(s)
- Brent R Degeorge
- Center for Translational Medicine, Department of Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Kamei N, Kwon SM, Alev C, Ishikawa M, Yokoyama A, Nakanishi K, Yamada K, Horii M, Nishimura H, Takaki S, Kawamoto A, Ii M, Akimaru H, Tanaka N, Nishikawa SI, Ochi M, Asahara T. Lnk deletion reinforces the function of bone marrow progenitors in promoting neovascularization and astrogliosis following spinal cord injury. Stem Cells 2010; 28:365-75. [PMID: 19859984 DOI: 10.1002/stem.243] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Lnk is an intracellular adaptor protein reported as a negative regulator of proliferation in c-Kit positive, Sca-1 positive, lineage marker-negative (KSL) bone marrow cells. The KSL fraction in mouse bone marrow is believed to represent a population of hematopoietic and endothelial progenitor cells (EPCs). We report here that, in vitro, Lnk(-/-) KSL cells form more EPC colonies than Lnk(+/+) KSL cells and show higher expression levels of endothelial marker genes, including CD105, CD144, Tie-1, and Tie2, than their wild-type counterparts. In vivo, the administration of Lnk(+/+) KSL cells to a mouse spinal cord injury model promoted angiogenesis, astrogliosis, axon growth, and functional recovery following injury, with Lnk(-/-) KSL being significantly more effective in inducing and promoting these regenerative events. At day 3 following injury, large vessels could be observed in spinal cords treated with KSL cells, and reactive astrocytes were found to have migrated along these large vessels. We could further show that the enhancement of astrogliosis appears to be caused in conjunction with the acceleration of angiogenesis. These findings suggest that Lnk deletion reinforces the commitment of KSL cells to EPCs, promoting subsequent repair of injured spinal cord through the acceleration of angiogenesis and astrogliosis.
Collapse
Affiliation(s)
- Naosuke Kamei
- Group of Vascular Regeneration, Institute of Biomedical Research and Innovation, 2-2 Minatojima-minamimachi, Chuo-ku, Kobe, Hyogo 650-0047, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Grajales L, García J, Banach K, Geenen DL. Delayed enrichment of mesenchymal cells promotes cardiac lineage and calcium transient development. J Mol Cell Cardiol 2010; 48:735-45. [PMID: 20060001 DOI: 10.1016/j.yjmcc.2009.12.022] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2009] [Revised: 12/09/2009] [Accepted: 12/25/2009] [Indexed: 12/11/2022]
Abstract
Bone marrow-derived mesenchymal stem cells (BM-MSCs) can be induced to differentiate into myogenic cells. Despite their potential, previous studies have not been successful in producing a high percentage of cardiac-like cells with a muscle phenotype. We hypothesized that cardiac lineage development in BM-MSC is related to cell passage, culture milieu, and enrichment for specific cell subtypes before and during differentiation. Our study demonstrated that Lin(-) BM-MSC at an intermediate passage (IP; P8-P12) expressed cardiac troponin T (cTnT) after 21 days in culture. Cardiac TnT expression was similar whether IP cells were differentiated in media containing 5-azacytidine+2% FBS (AZA; 14%) or 2% FBS alone (LS; 12%) and both were significantly higher than AZA+5% FBS. This expression was potentiated by first enriching for CD117/Sca-1 cells followed by differentiation (AZA, 39% and LS, 28%). A second sequential enrichment for the dihydropyridine receptor subunit alpha2delta1 (DHPR-alpha2) resulted in cardiac TnT expressed in 54% of cultured cells compared to 28% of cells after CD117/Sca-1(+) enrichment. Cells enriched for CD117/Sca-1 and subjected to differentiation displayed spontaneous intracellular Ca(2+) transients with an increase in transient frequency and a 60% decrease in the transient duration amplitude between days 14 and 29. In conclusion, IP CD117/Sca-1(+) murine BM-MSCs display robust cardiac muscle lineage development that can be induced independent of AZA but is diminished under higher serum concentrations. Furthermore, temporal changes in calcium kinetics commensurate with increased cTnT expression suggest progressive maturation of a cardiac muscle lineage. Enrichment with CD117/Sca-1 to establish lineage commitment followed by DHPR-alpha2 in lineage developing cells may enhance the therapeutic potential of these cells for transplantation.
Collapse
Affiliation(s)
- Liliana Grajales
- Department of Physiology and Biophysics, Section of Cardiology and the Center for Cardiovascular Research, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | | | | | |
Collapse
|
42
|
Xiang FL, Lu X, Hammoud L, Zhu P, Chidiac P, Robbins J, Feng Q. Cardiomyocyte-specific overexpression of human stem cell factor improves cardiac function and survival after myocardial infarction in mice. Circulation 2009; 120:1065-74, 9 p following 1074. [PMID: 19738140 DOI: 10.1161/circulationaha.108.839068] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND Soluble stem cell factor (SCF) has been shown to mobilize bone marrow stem cells and improve cardiac repair after myocardial infarction (MI). However, the effect of membrane-associated SCF on cardiac remodeling after MI is not known. The present study investigated the effects of cardiomyocyte-specific overexpression of the membrane-associated isoform of human SCF (hSCF) on cardiac function after MI. METHODS AND RESULTS A novel mouse model with tetracycline-inducible and cardiac-specific overexpression of membrane-associated hSCF was generated. MI was induced by left coronary artery ligation. Thirty-day mortality after MI was decreased in hSCF/tetracycline transactivator (tTA) compared with wild-type mice. In vivo cardiac function was significantly improved in hSCF/tTA mice at 5 and 30 days after MI compared with wild-type mice. Endothelial progenitor cell recruitment and capillary density were increased and myocardial apoptosis was decreased in the peri-infarct area of hSCF/tTA mice. Myocyte size was decreased in hSCF/tTA mice 30 days after MI compared with WT mice. Furthermore, hSCF overexpression promoted de novo angiogenesis as assessed by matrigel implantation into the left ventricular myocardium. CONCLUSIONS Cardiomyocyte-specific overexpression of hSCF improves myocardial function and survival after MI. These beneficial effects of hSCF may result from increases in endothelial progenitor cell recruitment and neovascularization and decreases in myocardial apoptosis and cardiac remodeling.
Collapse
Affiliation(s)
- Fu-Li Xiang
- Department of Physiology and Pharmacology, University of Western Ontario, London, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
43
|
Guo J, Jie W, Kuang D, Ni J, Chen D, Ao Q, Wang G. Ischaemia/reperfusion induced cardiac stem cell homing to the injured myocardium by stimulating stem cell factor expression via NF-kappaB pathway. Int J Exp Pathol 2009; 90:355-64. [PMID: 19563618 DOI: 10.1111/j.1365-2613.2009.00659.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Ischaemia/reperfusion (I/R) is a major cause of heart failure. Recently cardiac stem cells (CSCs) were proposed as the most appropriate cell type for heart disease therapy. However, it is still unclear whether I/R can stimulate the CSCs homing to the injured myocardium. Male Sprague-Dawley rats were subjected to a 30-min ischaemia followed by reperfusion of different intervals. RT-PCR, western blotting and immunohistochemistry were performed to detect stem cell factor (SCF) expression at mRNA and protein levels respectively. Activation of nuclear factor-kappaB (NF-kappaB) was determined by electrophoretic mobility shift assay. To assess the homing of CSCs in vivo, BrdU-labelled CSCs were injected into AV-groove before induction of ischaemia and examined by immunofluorescent staining in the injured myocardium after I/R. From day 3 to day 6 after reperfusion, the accumulation of CSCs was significantly elevated in the injured area, which was matched with the increased SCF expression during I/R. Pretreatment of rats with NF-kappaB inhibitor, N-acetyl-L-cysteine (NAC) not only suppressed NF-kappaB activation induced by I/R but also attenuated SCF expression. Further analysis revealed that I/R induced phosphorylation of IkappaBalpha after 15 min of reperfusion, and the raised phosphor-IkappaBalpha returned to the basal level at 2 h of reperfusion. In simulated I/R(SI/R) in vitro, it enhanced NF-kappaB activation and SCF expression in cultured neonatal rat cardiomyocytes, which was markedly inhibited by NF-kappaB decoy oligodeoxynucleotide or NAC. Taken together, our results demonstrated that I/R induced CSCs homing to the injured myocardium by stimulating myocardial SCF expression via activation of NF-kappaB.
Collapse
Affiliation(s)
- Junli Guo
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | | | | | | | | | | | | |
Collapse
|
44
|
Menasché P. Current status and future prospects for cell transplantation to prevent congestive heart failure. Semin Thorac Cardiovasc Surg 2008; 20:131-7. [PMID: 18707646 DOI: 10.1053/j.semtcvs.2008.03.001] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2008] [Indexed: 01/14/2023]
Abstract
Although most cardiac cell therapy trials have focused on patients with acute myocardial infarction, attempts at "regenerating" chronically failing hearts have also been performed. These studies have entailed use of skeletal myoblasts and bone marrow-derived cells. In the case of skeletal myoblasts, the randomized placebo-controlled myoblast autologous grafting in ischemic cardiomyopathy (MAGIC) trial has failed to show that myoblast injections increased ejection fraction beyond that seen in controls but the finding that the highest dose of myoblasts resulted in a significant antiremodeling effect compared with the placebo group provides an encouraging signal. In the case of bone marrow cells, surgical injections of the mononuclear fraction combined with coronary artery bypass surgery have not shown a substantial benefit but positive results have been reported with intraoperative epicardial injections of CD133(+) progenitors. There are three possible reasons for these mixed results. The first is the marked heterogeneity of cell functionality (particularly in the case of bone marrow), which would expectedly translate into variable clinical outcomes. The second reason is the low rate of sustained engraftment. The third possible explanation is a mismatch between the choice of end points and the presumed mechanism of action of the cells. The initial assumption that adult stem cells could effect myocardial tissue regeneration has led to usual focus on ejection fraction as the major surrogate endpoint. It is now increasingly recognized that adult stem cells, in contrast to their embryonic counterparts, have little if any regenerative capacity and that their presumed beneficial effects more likely involve paracrine signaling, in which case infarct size, perfusion, or left ventricular volumes might be more appropriate markers. Altogether, these observations provide a framework for future research, the results of which will then have to be integrated in the protocol design of second-generation clinical trials.
Collapse
Affiliation(s)
- Philippe Menasché
- Department of Cardiovascular Surgery, Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France.
| |
Collapse
|
45
|
Tateishi K, Takehara N, Matsubara H, Oh H. Stemming heart failure with cardiac- or reprogrammed-stem cells. J Cell Mol Med 2008; 12:2217-32. [PMID: 18754813 PMCID: PMC4514101 DOI: 10.1111/j.1582-4934.2008.00487.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Despite extensive efforts to control myocyte growth by genetic targeting of the cell cycle machinery and small molecules for cardiac repair, adult myocytes themselves appeared to divide a limited number of times in response to a variety of cardiac muscle stresses. Rare tissue-resident stem cells are thought to exist in many adult organs that are capable of self-renewal and differentiation and possess a range of actions that are potentially therapeutic. Recent studies suggest that a population of cardiac stem cells (CSCs) is maintained after cardiac development in the adult heart in mammals including human beings; however, homeostatic cardiomyocyte replacement might be stem cell-dependent, and functional myocardial regeneration after cardiac muscle damage is not yet considered as sufficient to fully maintain or reconstitute the cardiovascular system and function. Although it is clear that adult CSCs have limitations in their capabilities to proliferate extensively and differentiate in response to injury in vivo for replenishing mature car-diomyocytes and potentially function as resident stem cells. Transplantation of CSCs expanded ex vivo seems to require an integrated strategy of cell growth-enhancing factor(s) and tissue engineering technologies to support the donor cell survival and subsequent proliferation and differentiation in the host microenvironment. There has been substantial interest regarding the evidence that mammalian fibroblasts can be genetically reprogrammed to induced pluripotent stem (iPS) cells, which closely resemble embryonic stem (ES) cell properties capable of differentiating into functional cardiomyocytes, and these cells may provide an alternative cell source for generating patient-specific CSCs for therapeutic applications.
Collapse
Affiliation(s)
- Kento Tateishi
- Department of Experimental Therapeutics, Translational Research Center, Kyoto University Hospital, and Department of Cardiovascular Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | | | | | | |
Collapse
|