1
|
Organ-Specific Differentiation of Human Adipose-Derived Stem Cells in Various Organs of Xenotransplanted Rats: A Pilot Study. LIFE (BASEL, SWITZERLAND) 2022; 12:life12081116. [PMID: 35892918 PMCID: PMC9330795 DOI: 10.3390/life12081116] [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: 06/23/2022] [Revised: 07/19/2022] [Accepted: 07/22/2022] [Indexed: 11/17/2022]
Abstract
Adipose-derived stem cells (ADSCs) are potential therapeutics considering their self-renewal capacity and ability to differentiate into all somatic cell types in vitro. The ideal ADSC-based therapy is a direct injection into the relevant organs. The objective of this study was to investigate the viability and safety of intra-organ human ADSC (h-ADSC) xenotransplants in vivo. Subcutaneous adipose tissue from the abdominal area of 10 patients was sampled. h-ADSCs were isolated from adipose tissue samples and identified using immunofluorescence antibodies. Multi-differentiation potential assays for adipocytes, osteocytes, and chondrocytes were performed. Cultured h-ADSCs at passage 4 were transplanted into multiple organs of 17 rats, including the skin, subcutaneous layer, liver, kidney, pancreas, and spleen. The h-ADSC-injected organs excised after 100 days were examined, and the survival of h-ADSCs was measured by quantitative real-time polymerase chain reaction (qRT-PCR) using specific human and rat target genes. h-ADSCs confirmed by stem cell phenotyping were induced to differentiate into adipogenic, osteogenic, and chondrogenic lineages in vitro. All rats were healthy and exhibited no side effects during the study; the transplanted h-ADSCs did not cause inflammation and were indiscernible from the native organ cells. The presence of transplanted h-ADSCs was confirmed using qRT-PCR. However, the engrafted survival rates varied as follows: subcutaneous fat (70.6%), followed by the liver (52.9%), pancreas (50.0%), kidney (29.4%), skin (29.4%), and spleen (12.5%). h-ADSCs were successfully transplanted into a rat model, with different survival rates depending on the organ.
Collapse
|
2
|
Sun Y, Xiong X, Wang X. The miR-590-3p/VEGFA axis modulates secretion of VEGFA from adipose-derived stem cells, which acts as a paracrine regulator of human dermal microvascular endothelial cell angiogenesis. Hum Cell 2020; 33:479-489. [DOI: 10.1007/s13577-019-00315-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 12/13/2019] [Indexed: 12/25/2022]
|
3
|
Malik R, Darche FA, Rivinius R, Seckinger A, Krause U, Koenen M, Thomas D, Katus HA, Schweizer PA. Quantitative Efficacy and Fate of Mesenchymal Stromal Cells Targeted to Cardiac Sites by Radiofrequency Catheter Ablation. Cell Transplant 2020; 29:963689720914236. [PMID: 32207339 PMCID: PMC7444233 DOI: 10.1177/0963689720914236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Engraftment and functional integration of stem cells or stem cell-derived cells within cardiac tissue is an important prerequisite for cell replacement therapy aiming at the treatment of heart disease. Recently, a novel intravenous approach for application of mesenchymal stromal cells (MSCs) to cardiac sites has been established using radiofrequency catheter ablation (RFCA)-guided targeting, bypassing the need for open chest surgery or direct myocardial cell injection. However, little is known about the quantitative efficacy and longevity of this strategy. We performed selective power-controlled RFCA with eight ablation pulses (30 W, 60 s each) to induce heat-mediated lesions at the right atrial appendices (RAAs) of pigs. Different concentrations of human bone marrow-derived MSCs (105 to 1.6 × 106 cells/kg bodyweight) labeled with superparamagnetic iron oxide (SPIO) particles were infused intravenously in nine pigs one d after RFCA treatment and hearts were explanted 8 d later to quantify the number of engrafted cells. Prussian blue staining revealed high numbers of SPIO-labeled cells in areas surrounding the RFCA-induced lesions. Cell numbers were evaluated by quantitative real-time polymerase chain reaction using specific primers for human MSCs (hMSCs), which indicated that up to 106 hMSCs, corresponding to ∼3.9% of the systemically applied human cells, engrafted within the RAAs of RFCA-treated pigs. Of note, infused hMSCs were observed in nontargeted organs, as well, but appeared at very low concentrations. To assess long-term deposition of MSCs, RAAs of three pigs were analyzed after 6 months, which revealed few persisting hMSCs at targeted sites. RFCA-mediated targeting of MSCs provides a novel minimal invasive strategy for cardiac stem cell engraftment. Qualitative and quantitative results of our large animal experiments indicate an efficient guidance of MSCs to selected cardiac regions, although only few cells remained at targeted sites 6 mo after cell transplantation.
Collapse
Affiliation(s)
- Rizwan Malik
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany
| | - Fabrice A Darche
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Rasmus Rivinius
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Anja Seckinger
- Department of Hematology, Oncology and Rheumatology, Medical University Hospital Heidelberg, Heidelberg, Germany
| | - Ulf Krause
- Department of Hematology, Oncology and Rheumatology, Medical University Hospital Heidelberg, Heidelberg, Germany.,Institute for Transfusion Medicine and Cellular Therapy, University Hospital Muenster, Domagstrasse, Muenster, Germany
| | - Michael Koenen
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany.,Department of Molecular Neurobiology, Max-Planck-Institute for Medical Research, Jahnstrasse, Heidelberg, Germany
| | - Dierk Thomas
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Hugo A Katus
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Patrick A Schweizer
- Department of Cardiology, Medical University Hospital Heidelberg, Heidelberg, Germany.,DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
4
|
Ni H, Zhao Y, Ji Y, Shen J, Xiang M, Xie Y. Adipose-derived stem cells contribute to cardiovascular remodeling. Aging (Albany NY) 2019; 11:11756-11769. [PMID: 31800397 PMCID: PMC6932876 DOI: 10.18632/aging.102491] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 11/17/2019] [Indexed: 02/06/2023]
Abstract
Obesity is an independent risk factor for cardiovascular disease. Adipose tissue was initially thought to be involved in metabolism through paracrine. Recent researches discovered mesenchymal stem cells inside adipose tissue which could differentiate into vascular lineages in vitro and in vivo, participating vascular remodeling. However, there were few researches focusing on distinct characteristics and functions of adipose-derived stem cells (ADSCs) from different regions. This is the first comprehensive review demonstrating the variances of ADSCs from the perspective of their origins.
Collapse
Affiliation(s)
- Hui Ni
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yiming Zhao
- Department of Endocrinology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yongli Ji
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Shen
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Meixiang Xiang
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yao Xie
- Department of Cardiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
5
|
|
6
|
Kolecka MA, Arnhold S, Schmidt M, Reich C, Kramer M, Failing K, von Pückler K. Behaviour of adipose-derived canine mesenchymal stem cells after superparamagnetic iron oxide nanoparticles labelling for magnetic resonance imaging. BMC Vet Res 2017; 13:62. [PMID: 28235414 PMCID: PMC5324251 DOI: 10.1186/s12917-017-0980-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 02/17/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Therapy with mesenchymal stem cells (MSCs) has been reported to provide beneficial effects in the treatment of neurological and orthopaedic disorders in dogs. The exact mechanism of action is poorly understood. Magnetic resonance imaging (MRI) gives the opportunity to observe MSCs after clinical administration. To visualise MSCs with the help of MRI, labelling with an MRI contrast agent is necessary. However, it must be clarified whether there is any negative influence on cell function and viability after labelling prior to clinical administration. RESULTS For the purpose of the study, seven samples with canine adipose-derived stem cells were incubated with superparamagnetic iron oxide nanoparticles (SPIO: 319.2 μg/mL Fe) for 24 h. The internalisation of the iron particles occurred via endocytosis. SPIO particles were localized as free clusters in the cytoplasm or within lysosomes depending on the time of investigation. The efficiency of the labelling was investigated using Prussian blue staining and MACS assay. After 3 weeks the percentage of SPIO labelled canine stem cells decreased. Phalloidin staining showed no negative effect on the cytoskeleton. Labelled cells underwent osteogenic and adipogenic differentiation. Chondrogenic differentiation occurred to a lesser extent compared with a control sample. MTT-Test and wound healing assay showed no influence of labelling on the proliferation. The duration of SPIO labelling was assessed using a 1 Tesla clinical MRI scanner and T2 weighted turbo spin echo and T2 weighted gradient echo MRI sequences 1, 2 and 3 weeks after labelling. The hypointensity caused by SPIO lasted for 3 weeks in both sequences. CONCLUSIONS An Endorem labelling concentration of 319.2 μg/mL Fe (448 μg/mL SPIO) had no adverse effects on the viability of canine ASCs. Therefore, this contrast agent could be used as a model for iron oxide labelling agents. However, the tracking ability in vivo has to be evaluated in further studies.
Collapse
Affiliation(s)
- Malgorzata Anna Kolecka
- Department of Veterinary Clinical Sciences, Clinic for Small Animal-Surgery, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Giessen, Germany.
| | - Stefan Arnhold
- Institute of Veterinary Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Giessen, Germany
| | - Martin Schmidt
- Department of Veterinary Clinical Sciences, Clinic for Small Animal-Surgery, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Giessen, Germany
| | - Christine Reich
- Institute of Veterinary Anatomy, Histology and Embryology, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Giessen, Germany
| | - Martin Kramer
- Department of Veterinary Clinical Sciences, Clinic for Small Animal-Surgery, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Giessen, Germany
| | - Klaus Failing
- Institute of Biomathematics, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Giessen, Germany
| | - Kerstin von Pückler
- Department of Veterinary Clinical Sciences, Clinic for Small Animal-Surgery, Faculty of Veterinary Medicine, Justus-Liebig-University Giessen, Giessen, Germany
| |
Collapse
|
7
|
Hu J, Fu Z, Chen Y, Tang N, Wang L, Wang F, Sun R, Yan S. Effects of autologous adipose-derived stem cell infusion on type 2 diabetic rats. Endocr J 2015; 62:339-52. [PMID: 25739585 DOI: 10.1507/endocrj.ej14-0584] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The effects and possible mechanisms of adipose-derived stem cells (ASC) infusion on type 2 diabetic rats were investigated in this study. Twenty normal male Sprague-Dawley rats were included in normal control group, and 40 male diabetic rats were randomly divided into diabetic control group and ASC group (which received ASC infusion). After therapy, levels of fasting plasma glucose (FPG), HbA1c, serum insulin and C-peptide, recovery of islet cells, inflammatory cytokines, and insulin sensitivity were analyzed. After ASC infusion, compared with diabetic control group, hyperglycemia in ASC group was ameliorated in 2 weeks and maintained for about 6 weeks, and plasma concentrations of insulin and C-peptide were significantly improved (P<0.01). Number of islet β cells and concentration of vWF in islets in ASC group increased, while activity of caspase-3 in islets was reduced. Moreover, concentrations of TNF-α, IL-6 and IL-1β in ASC group obviously decreased (P<0.05). The expression of GLUT4, INSR, and phosphorylation of insulin signaling molecules in insulin target tissues were effectively improved. ASC infusion could aid in T2DM through recovery of islet β cells and improvement of insulin sensitivity. Autologous ASC infusion might be an effective method for T2DM.
Collapse
Affiliation(s)
- Jianxia Hu
- Stem Cell Research Center, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Albersen M, Berkers J, Dekoninck P, Deprest J, Lue TF, Hedlund P, Lin CS, Bivalacqua TJ, Van Poppel H, De Ridder D, Van der Aa F. Expression of a Distinct Set of Chemokine Receptors in Adipose Tissue-Derived Stem Cells is Responsible for In Vitro Migration Toward Chemokines Appearing in the Major Pelvic Ganglion Following Cavernous Nerve Injury. Sex Med 2014; 1:3-15. [PMID: 25356281 PMCID: PMC4184711 DOI: 10.1002/sm2.1] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Adipose tissue-derived stem cells (ADSCs) herald tremendous promise for clinical application in a wide range of injuries and diseases. Several preclinical reports demonstrate their efficacy in the treatment of cavernous nerve (CN) injury-induced erectile dysfunction in rats. It was recently illustrated that these effects were established as a result of ADSC migration to the major pelvic ganglion (MPG) where these cells induced neuroregeneration in loco. AIMS The study aims to identify chemotactic factors in the MPG following injury and to match upregulated chemokines to their respective receptors in human ADSC on the genomic, structural, and functional levels. METHODS Quantitative real-time polymerase chain reaction, fluorescence-activated cell sorting (FACS), intracellular FACS, immunofluorescence microscopy, migration assays, and calcium imaging were used in this study. MAIN OUTCOME MEASURES The main outcomes are chemokine expression in the MPG following CN injury, and the functional and structural presence of chemokine receptors in ADSC. RESULTS CCR4, CX3CR1, and XCR1 are functionally and structurally present in human ADSC, and are activated by the chemokines CCL2, CX3CL1, and XCL1 respectively, which are upregulated in the MPG following CN injury. CXCR4 and its ligand CXCL12 (SDF1) are likely no major homing factors for ADSC. Expression of chemokine receptor mRNA in ADSC did not necessarily translate into receptor presence at the cell surface and/or functional activation of these receptors. Most of the expressed chemokine receptors were detected in the intracellular compartment of these cells. CONCLUSIONS We identified the ligand/chemokine receptor pairs CCL2/CCR4, CX3CL1/CX3CR1, and XCL1/XCR1 as potentially responsible for ADSC homing toward the MPG following CN injury. The intracellular localization of various chemokine receptors likely indicates redirecting of chemokine receptors to the cell surface under specific cellular conditions. Furthermore, modification of expression of these receptors at the genomic level may potentially lead to improved migration toward injury sites and thus enhancement of treatment efficacy. Albersen M, Berkers J, Dekoninck P, Deprest J, Lue TF, Hedlund P, Lin C-S, Bivalacqua TJ, Van Poppel H, De Ridder D, and Van der Aa F. Expression of a distinct set of chemokine receptors in adipose tissue-derived stem cells is responsible for in vitro migration toward chemokines appearing in the major pelvic ganglion following cavernous nerve injury. Sex Med 2013;1:3-15.
Collapse
Affiliation(s)
- Maarten Albersen
- Laboratory of Experimental Urology, Department of Development and Regeneration, University of Leuven Leuven, Belgium ; Laboratory of Experimental Gynecology, Department of Development and Regeneration, University of Leuven Leuven, Belgium
| | - Joost Berkers
- Laboratory of Experimental Urology, Department of Development and Regeneration, University of Leuven Leuven, Belgium
| | - Philip Dekoninck
- Laboratory of Experimental Gynecology, Department of Development and Regeneration, University of Leuven Leuven, Belgium
| | - Jan Deprest
- Laboratory of Experimental Gynecology, Department of Development and Regeneration, University of Leuven Leuven, Belgium
| | - Tom F Lue
- Knuppe Molecular Urology Laboratory, Department of Urology, University of California San Francisco, CA, USA
| | - Petter Hedlund
- Department of Urology, Urological Research Institute, Vita-Salute San Raffaele University Milan, Italy
| | - Ching-Shwun Lin
- Knuppe Molecular Urology Laboratory, Department of Urology, University of California San Francisco, CA, USA
| | - Trinity J Bivalacqua
- Department of Urology, The James Buchanan Brady Urological Institute, Johns Hopkins Medical Institutions Baltimore, MD, USA
| | - Hendrik Van Poppel
- Laboratory of Experimental Urology, Department of Development and Regeneration, University of Leuven Leuven, Belgium
| | - Dirk De Ridder
- Laboratory of Experimental Urology, Department of Development and Regeneration, University of Leuven Leuven, Belgium
| | - Frank Van der Aa
- Laboratory of Experimental Urology, Department of Development and Regeneration, University of Leuven Leuven, Belgium
| |
Collapse
|
9
|
Luo L, Lin T, Zheng S, Xie Z, Chen M, Lian G, Xu C, Wang H, Xie L. Adipose-derived stem cells attenuate pulmonary arterial hypertension and ameliorate pulmonary arterial remodeling in monocrotaline-induced pulmonary hypertensive rats. Clin Exp Hypertens 2014; 37:241-8. [PMID: 25271670 DOI: 10.3109/10641963.2014.954710] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
We investigated the effect of adipose-derived stem cells (ADSCs) transplantation effects on structural remodeling and pulmonary artery pressure in monocrotaline (MCT)-induced pulmonary hypertensive rats. In the first experiment, 32 male Sprague-Dawley (SD) rats were randomly divided into four groups (n = 8/group): 3 ADSCs treated groups and normal control (Ctrl). ADSCs were administered through the left jugular vein at 10(5), 10(6) and 10(7) cells, respectively, and a cell density of 10(6)cells/ml was shown to be optimal. The GFP-tagged ADSCs were identified in the lungs and differentiated into endothelial-like cells. In the second experiment, 96 male SD rats were randomly divided into three groups (n = 32/group): Ctrl, MCT-induced pulmonary arterial hypertension (PAH), and PAH treated with ADSCs (ADSCs). Two weeks post-MCT administration, the ADSCs group received 1 × 10(6) ADSCs via the external jugular vein. Compared to PAH rats, mean pulmonary arterial pressure was decreased in rats at 1, 2, and 3 weeks after ADSCs-treatment (18.63 ± 2.15 mmHg versus 24.53 ± 2.90 mmHg; 23.07 ± 2.84 mmHg versus 33.18 ± 2.30 mmHg; 22.98 ± 2.34 mmHg versus 36.38 ± 3.28 mmHg, p < 0.05). Meanwhile, the right heart hypertrophy index (36.2 1 ± 4.27% versus 41.01 ± 1.29%; 39.47 ± 4.02% versus 48.75 ± 2 .13%; 41.02 ± 0.9% versus 50.52 ± 1.49%, p < 0.05, respectively), ratio of wall/lumen thickness, as well as the wall/lumen area were significantly reduced in PAH rats at these time points following ADSCs-treatment, as compared with untreated PAH rats. In summary, ADSCs may colonize the pulmonary arteries, attenuate pulmonary arterial hypertension and ameliorate pulmonary arterial remodeling.
Collapse
Affiliation(s)
- Li Luo
- Fujian Hypertension Research Institute, the First Affiliated Hospital of Fujian Medical University , Fuzhou , P.R. China
| | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Exendin-4 pretreated adipose derived stem cells are resistant to oxidative stress and improve cardiac performance via enhanced adhesion in the infarcted heart. PLoS One 2014; 9:e99756. [PMID: 24915574 PMCID: PMC4051823 DOI: 10.1371/journal.pone.0099756] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 05/18/2014] [Indexed: 12/12/2022] Open
Abstract
Reactive oxygen species (ROS), which were largely generated after myocardial ischemia, severely impaired the adhesion and survival of transplanted stem cells. In this study, we aimed to determine whether Exendin-4 pretreatment could improve the adhesion and therapeutic efficacy of transplanted adipose derived stem cells (ADSCs) in ischemic myocardium. In vitro, H2O2 was used to provide ROS environments, in which ADSCs pretreated with Exendin-4 were incubated. ADSCs without pretreatment were used as control. Then, cell adhesion and viability were analyzed with time. Compared with control ADSCs, Exendin-4 treatment significantly increased the adhesion of ADSCs in ROS environment, while reduced intracellular ROS and cell injury as determined by dihydroethidium (DHE) staining live/Dead staining, lactate dehydrogenase-release assay and MTT assay. Western Blotting demonstrated that ROS significantly decreased the expression of adhesion-related integrins and integrin-related focal adhesion proteins, which were significantly reversed by Exendin-4 pretreatment and followed by decreases in caspase-3, indicating that Exendin-4 may facilitate cell survival through enhanced adhesion. In vivo, myocardial infarction (MI) was induced by the left anterior descending artery ligation in SD rats. Autologous ADSCs with or without Exendin-4 pretreatment were injected into the border area of infarcted hearts, respectively. Multi-techniques were used to assess the beneficial effects after transplantation. Longitudinal bioluminescence imaging and histological staining revealed that Exendin-4 pretreatment enhanced the survival and differentiation of engrafted ADSCs in ischemic myocardium, accompanied with significant benefits in cardiac function, matrix remodeling, and angiogenesis compared with non-pretreated ADSCs 4 weeks post-transplantation. In conclusion, transplantation of Exendin-4 pretreated ADSCs significantly improved cardiac performance and can be an innovative approach in the clinical perspective.
Collapse
|
11
|
Cellular kinetics of perivascular MSC precursors. Stem Cells Int 2013; 2013:983059. [PMID: 24023546 PMCID: PMC3760099 DOI: 10.1155/2013/983059] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Accepted: 07/13/2013] [Indexed: 12/26/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) and MSC-like multipotent stem/progenitor cells have been widely investigated for regenerative medicine and deemed promising in clinical applications. In order to further improve MSC-based stem cell therapeutics, it is important to understand the cellular kinetics and functional roles of MSCs in the dynamic regenerative processes. However, due to the heterogeneous nature of typical MSC cultures, their native identity and anatomical localization in the body have remained unclear, making it difficult to decipher the existence of distinct cell subsets within the MSC entity. Recent studies have shown that several blood-vessel-derived precursor cell populations, purified by flow cytometry from multiple human organs, give rise to bona fide MSCs, suggesting that the vasculature serves as a systemic reservoir of MSC-like stem/progenitor cells. Using individually purified MSC-like precursor cell subsets, we and other researchers have been able to investigate the differential phenotypes and regenerative capacities of these contributing cellular constituents in the MSC pool. In this review, we will discuss the identification and characterization of perivascular MSC precursors, including pericytes and adventitial cells, and focus on their cellular kinetics: cell adhesion, migration, engraftment, homing, and intercellular cross-talk during tissue repair and regeneration.
Collapse
|
12
|
Li K, Qin J, Wang X, Xu Y, Shen Z, Lu X, Zhang G. Magnetic resonance imaging monitoring dual-labeled stem cells for treatment of mouse nerve injury. Cytotherapy 2013; 15:1275-85. [PMID: 23727476 DOI: 10.1016/j.jcyt.2013.03.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Revised: 02/18/2013] [Accepted: 03/16/2013] [Indexed: 01/09/2023]
Abstract
BACKGROUND AIMS Adipose-derived stem cells (ADSCs) have shown great promise in the regenerative repair of injured peripheral nerves. Magnetic resonance imaging (MRI) has provided attractive advantages in tracking superparamagnetic iron oxide nanoparticle (SPION)-labeled cells and evaluating their fate after cell transplantation. This study investigated the feasibility of the use of MRI to noninvasively track ADSCs repair of peripheral nerve injury in vivo. METHODS Green fluorescent protein (GFP)-expressing ADSCs were isolated, expanded, differentiated into an SC-like phenotype (GFP-dADSCs) at early passages and subsequently labeled with SPIONs. The morphological and functional properties of the GFP-dADSCs were assessed through the use of immunohistochemistry. The intracellular stability, proliferation and viability of the labeled cells were evaluated in vitro. Through the use of a microsurgical procedure, the labeled cells were then seeded into sciatic nerve conduits in C57/BL6 mice to repair a 1-cm sciatic nerve gap. A clinical 3-T MRI was performed to investigate the GFP-dADSCs in vitro and the transplanted GFP-dADSCs inside the sciatic nerve conduits in vivo. RESULTS The GFP-dADSCs were efficiently labeled with SPIONs, without affecting their viability and proliferation. The labeled cells implanted into the mice sciatic nerve conduit exhibited a significant increase in axonal regeneration compared with the empty conduit and could be detected by MRI. Fluorescent microscopic examination, histological analysis and immunohistochemistry confirmed the axon regeneration and MRI results. CONCLUSIONS These data will elucidate the neuroplasticity of ADSCs and provide a new protocol for in vivo tracking of stem cells that are seeded to repair injured peripheral nerves.
Collapse
Affiliation(s)
- Kangan Li
- Department of Radiology, Shanghai First People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, People's Republic of China.
| | | | | | | | | | | | | |
Collapse
|
13
|
Qin JB, Li KA, Li XX, Xie QS, Lin JY, Ye KC, Jiang ME, Zhang GX, Lu XW. Long-term MRI tracking of dual-labeled adipose-derived stem cells homing into mouse carotid artery injury. Int J Nanomedicine 2012; 7:5191-203. [PMID: 23125528 PMCID: PMC3487538 DOI: 10.2147/ijn.s35647] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background Stem cell therapy has shown great promise for regenerative repair of injured or diseased tissues. Adipose-derived stem cells (ADSCs) have become increasingly attractive candidates for cellular therapy. Magnetic resonance imaging has been proven to be effective in tracking magnetic-labeled cells and evaluating their clinical relevance after cell transplantation. This study investigated the feasibility of imaging green fluorescent protein-expressing ADSCs (GFP-ADSCs) labeled with superparamagnetic iron oxide particles, and tracked them in vivo with noninvasive magnetic resonance imaging after cell transplantation in a model of mouse carotid artery injury. Methods GFP-ADSCs were isolated from the adipose tissues of GFP mice and labeled with superparamagnetic iron oxide particles. Intracellular stability, proliferation, and viability of the labeled cells were evaluated in vitro. Next, the cells were transplanted into a mouse carotid artery injury model. Clinical 3 T magnetic resonance imaging was performed immediately before and 1, 3, 7, 14, 21, and 30 days after cell transplantation. Prussian blue staining and histological analysis were performed 7 and 30 days after transplantation. Results GFP-ADSCs were found to be efficiently labeled with superparamagnetic iron oxide particles, with no effect on viability and proliferation. Homing of the labeled cells into the injured carotid artery tissue could be monitored by magnetic resonance imaging. Conclusion Magnetically labeled ADSCs with expression of GFP can home into sites of vascular injury, and may provide new insights into understanding of cell-based therapy for cardiovascular lesions.
Collapse
Affiliation(s)
- Jin-Bao Qin
- Department of Vascular Surgery, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Richards JMJ, Shaw CA, Lang NN, Williams MC, Semple SIK, MacGillivray TJ, Gray C, Crawford JH, Alam SR, Atkinson APM, Forrest EK, Bienek C, Mills NL, Burdess A, Dhaliwal K, Simpson AJ, Wallace WA, Hill AT, Roddie PH, McKillop G, Connolly TA, Feuerstein GZ, Barclay GR, Turner ML, Newby DE. In vivo mononuclear cell tracking using superparamagnetic particles of iron oxide: feasibility and safety in humans. Circ Cardiovasc Imaging 2012; 5:509-17. [PMID: 22787016 DOI: 10.1161/circimaging.112.972596] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Cell therapy is an emerging and exciting novel treatment option for cardiovascular disease that relies on the delivery of functional cells to their target site. Monitoring and tracking cells to ensure tissue delivery and engraftment is a critical step in establishing clinical and therapeutic efficacy. The study aims were (1) to develop a Good Manufacturing Practice-compliant method of labeling competent peripheral blood mononuclear cells with superparamagnetic particles of iron oxide (SPIO), and (2) to evaluate its potential for magnetic resonance cell tracking in humans. METHODS AND RESULTS Peripheral blood mononuclear cells 1-5 × 10(9) were labeled with SPIO. SPIO-labeled cells had similar in vitro viability, migratory capacity, and pattern of cytokine release to unlabeled cells. After intramuscular administration, up to 10(8) SPIO-labeled cells were readily identifiable in vivo for at least 7 days using magnetic resonance imaging scanning. Using a phased-dosing study, we demonstrated that systemic delivery of up to 10(9) SPIO-labeled cells in humans is safe, and cells accumulating in the reticuloendothelial system were detectable on clinical magnetic resonance imaging. In a healthy volunteer model, a focus of cutaneous inflammation was induced in the thigh by intradermal injection of tuberculin. Intravenously delivered SPIO-labeled cells tracked to the inflamed skin and were detectable on magnetic resonance imaging. Prussian blue staining of skin biopsies confirmed iron-laden cells in the inflamed skin. CONCLUSIONS Human peripheral blood mononuclear cells can be labeled with SPIO without affecting their viability or function. SPIO labeling for magnetic resonance cell tracking is a safe and feasible technique that has major potential for a range of cardiovascular applications including monitoring of cell therapies and tracking of inflammatory cells. Clinical Trial Registration- URL: http://www.clinicaltrials.gov; Unique identifier: NCT00972946, NCT01169935.
Collapse
Affiliation(s)
- Jennifer M J Richards
- Centre of Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom .
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Lin CS, Lin G, Lue TF. Allogeneic and xenogeneic transplantation of adipose-derived stem cells in immunocompetent recipients without immunosuppressants. Stem Cells Dev 2012; 21:2770-8. [PMID: 22621212 DOI: 10.1089/scd.2012.0176] [Citation(s) in RCA: 159] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are well known for their immunomodulatory capabilities. In particular, their immunosuppressive property is believed to permit their allogeneic or even xenogeneic transplantation into immunocompetent recipients without the use of immunosuppressants. Adipose-derived stem cell (ADSC), owing to its ease of isolation from an abundant tissue source, is a promising MSC for the treatment of a wide range of diseases. ADSC has been shown to lack major histocompatibility complex-II expression, and its immunosuppressive effects mediated by prostaglandin E2. Both preclinical and clinical studies have shown that allogeneic transplantation of ADSCs was able to control graft-versus-host disease. In regard to xenotransplantation a total of 27 preclinical studies have been published, with 20 of them performed with the investigators' intent. All 27 studies used ADSCs isolated from humans, possibly due to the wide availability of lipoaspirates. On the other hand, the recipients were mouse in 13 studies, rat in 11, rabbit in 2, and dog in 1. The targeted diseases varied greatly but all showed significant improvements after ADSC xenotransplantation. For clinical application in human medicine, ADSC xenotransplantation offers no obvious advantage over autotransplantation. But in veterinary medicine, xenotransplantation with porcine ADSC is a practical alternative to the costly and inconvenient autotransplantation.
Collapse
Affiliation(s)
- Ching-Shwun Lin
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California 94143-0738, USA.
| | | | | |
Collapse
|
16
|
Perán M, García MA, López-Ruiz E, Bustamante M, Jiménez G, Madeddu R, Marchal JA. Functionalized nanostructures with application in regenerative medicine. Int J Mol Sci 2012; 13:3847-3886. [PMID: 22489186 PMCID: PMC3317746 DOI: 10.3390/ijms13033847] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2012] [Revised: 03/03/2012] [Accepted: 03/06/2012] [Indexed: 12/16/2022] Open
Abstract
In the last decade, both regenerative medicine and nanotechnology have been broadly developed leading important advances in biomedical research as well as in clinical practice. The manipulation on the molecular level and the use of several functionalized nanoscaled materials has application in various fields of regenerative medicine including tissue engineering, cell therapy, diagnosis and drug and gene delivery. The themes covered in this review include nanoparticle systems for tracking transplanted stem cells, self-assembling peptides, nanoparticles for gene delivery into stem cells and biomimetic scaffolds useful for 2D and 3D tissue cell cultures, transplantation and clinical application.
Collapse
Affiliation(s)
- Macarena Perán
- Department of Health Sciences, University of Jaén, Jaén E-23071, Spain; E-Mails: (M.P.); (E.L.-R.)
| | - María A. García
- Research Unit, Hospital Universitario Virgen de las Nieves, Granada E-18014, Spain; E-Mail:
| | - Elena López-Ruiz
- Department of Health Sciences, University of Jaén, Jaén E-23071, Spain; E-Mails: (M.P.); (E.L.-R.)
| | - Milán Bustamante
- Biosciences Institute, University College Cork, Cork, Ireland; E-Mail:
| | - Gema Jiménez
- Biopathology and Regenerative Medicine Institute (IBIMER), Biomedical Research Centre, University of Granada, Granada E-18100, Spain; E-Mail:
| | - Roberto Madeddu
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; E-Mail:
| | - Juan A. Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Biomedical Research Centre, University of Granada, Granada E-18100, Spain; E-Mail:
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada E-18012, Spain
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +34-958-249-321; Fax: +34-958-246-296
| |
Collapse
|
17
|
Studies in adipose-derived stromal cells: migration and participation in repair of cranial injury after systemic injection. Plast Reconstr Surg 2011; 127:1130-1140. [PMID: 21364416 DOI: 10.1097/prs.0b013e3182043712] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
BACKGROUND Adipose-derived stromal cells are a multipotent cell type with the ability to undergo osteogenic differentiation. The authors sought to examine whether systemically administered adipose-derived stromal cells would migrate to and heal surgically created defects of the mouse cranial skeleton. METHODS Mouse adipose-derived stromal cells were harvested from luciferase-positive transgenic mice; human adipose-derived stromal cells were harvested from human lipoaspirate and labeled with luciferase and green fluorescent protein. A 4-mm calvarial defect (critical sized) was made in the mouse parietal bone; skin incisions alone were used as a control (n = 5 per group). Adipose-derived stromal cells were injected intravenously (200,000 cells per animal) and compared with saline injection only. Methods of analyses included micro-computed tomographic scanning, in vivo imaging system detection of luciferase activity, and standard histology. RESULTS Migration of adipose-derived stromal cells to calvarial defect sites was confirmed by accumulation of luciferase activity and green fluorescent protein stain as early as 4 days and persisting up to 4 weeks. Little activity was observed among control groups. Intravenous administration of either mouse or human adipose-derived stromal cells resulted in histologic evidence of bone formation within the defect site, in comparison with an absence of bone among control defects. By micro-computed tomographic analysis, human but not mouse adipose-derived stromal cells stimulated significant osseous healing. CONCLUSIONS Intravenously administered adipose-derived stromal cells migrate to sites of calvarial injury. Thereafter, intravenous human adipose-derived stromal cells contribute to bony calvarial repair. Intravenous administration of adipose-derived stromal cells may be an effective delivery method for future efforts in skeletal regeneration.
Collapse
|
18
|
Lin G, Yang R, Banie L, Wang G, Ning H, Li LC, Lue TF, Lin CS. Effects of transplantation of adipose tissue-derived stem cells on prostate tumor. Prostate 2010; 70:1066-73. [PMID: 20232361 PMCID: PMC2877148 DOI: 10.1002/pros.21140] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
BACKGROUND Obesity is a risk factor for prostate cancer development, but the underlying mechanism is unknown. The present study tested the hypothesis that stromal cells of the adipose tissue might be recruited by cancer cells to help tumor growth. METHODS PC3 prostate cancer cells were transplanted into the subcutaneous space of the right flank of athymic mice. One week later, adipose tissue-derived stromal or stem cells (ADSC) or phosphate-buffered saline (PBS, as control) was transplanted similarly to the left flank. Tumor size was monitored for the next 34 days; afterwards, the mice were sacrificed and their tumors harvested for histological examination. The ability of PC3 cells to attract ADSC was tested by migration assay. The involvement of the CXCL12/CXCR4 axis was tested by migration assay in the presence of a specific inhibitor AMD3100. RESULTS Throughout the entire course, the average size of PC3 tumors in ADSC-treated mice was larger than in PBS-treated mice. ADSC were identified inside the tumors of ADSC-treated mice; CXCR4 expression was also detected. Migration assay indicated the involvement of the CXCL12/CXCR4 axis in the migration of ADSC toward PC3 cells. Capillary density was twice as high in the tumors of ADSC-treated mice than in the tumors of PBS-treated mice. VEGF expression was similar but FGF2 expression was significantly higher in tumors of ADSC-treated mice than in the tumors of PBS-tread mice. CONCLUSION Prostate cancer cells recruited ADSC by the CXCL12/CXCR4 axis. ADSC helps tumor growth by increasing tumor vascularity, and which was mediated by FGF2.
Collapse
Affiliation(s)
- Guiting Lin
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California
| | - Rong Yang
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California
| | - Lia Banie
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California
| | - Guifang Wang
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California
| | - Hongxiu Ning
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California
| | - Long-Cheng Li
- Department of Urology and Helen-Diller Comprehensive Cancer Center, University of California, San Francisco, California
| | - Tom F. Lue
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California
| | - Ching-Shwun Lin
- Knuppe Molecular Urology Laboratory, Department of Urology, School of Medicine, University of California, San Francisco, California
- Correspondence to: Ching-Shwun Lin, Department of Urology, University of California, San Francisco, CA 94143-0738.
| |
Collapse
|
19
|
Abstract
Humans and other mammals have three main adipose tissue depots: visceral white adipose tissue, subcutaneous white adipose tissue and brown adipose tissue, each of which possesses unique cell-autonomous properties. In contrast to visceral adipose tissue, which can induce detrimental metabolic effects, subcutaneous white adipose tissue and brown adipose tissue have the potential to benefit metabolism by improving glucose homeostasis and increasing energy consumption. In addition, adipose tissue contains adipose-derived stem cells, which possess the ability to differentiate into multiple lineages, a property that might be of value for the repair or replacement of various damaged cell types. Adipose tissue transplantation has primarily been used as a tool to study physiology and for human reconstructive surgery. Transplantation of adipose tissue is, however, now being explored as a possible tool to promote the beneficial metabolic effects of subcutaneous white adipose tissue and brown adipose tissue, as well as adipose-derived stem cells. Ultimately, the clinical applicability of adipose tissue transplantation for the treatment of obesity and metabolic disorders will reside in the achievable level of safety, reliability and efficacy compared with other treatments.
Collapse
Affiliation(s)
- Thien T Tran
- Joslin Diabetes Center and Harvard Medical School, Boston, MA 02215, USA
| | | |
Collapse
|