151
|
Radke D, Jia W, Sharma D, Fena K, Wang G, Goldman J, Zhao F. Tissue Engineering at the Blood-Contacting Surface: A Review of Challenges and Strategies in Vascular Graft Development. Adv Healthc Mater 2018; 7:e1701461. [PMID: 29732735 PMCID: PMC6105365 DOI: 10.1002/adhm.201701461] [Citation(s) in RCA: 149] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 02/09/2018] [Indexed: 12/14/2022]
Abstract
Tissue engineered vascular grafts (TEVGs) are beginning to achieve clinical success and hold promise as a source of grafting material when donor grafts are unsuitable or unavailable. Significant technological advances have generated small-diameter TEVGs that are mechanically stable and promote functional remodeling by regenerating host cells. However, developing a biocompatible blood-contacting surface remains a major challenge. The TEVG luminal surface must avoid negative inflammatory responses and thrombogenesis immediately upon implantation and promote endothelialization. The surface has therefore become a primary focus for research and development efforts. The current state of TEVGs is herein reviewed with an emphasis on the blood-contacting surface. General vascular physiology and developmental challenges and strategies are briefly described, followed by an overview of the materials currently employed in TEVGs. The use of biodegradable materials and stem cells requires careful control of graft composition, degradation behavior, and cell recruitment ability to ensure that a physiologically relevant vessel structure is ultimately achieved. The establishment of a stable monolayer of endothelial cells and the quiescence of smooth muscle cells are critical to the maintenance of patency. Several strategies to modify blood-contacting surfaces to resist thrombosis and control cellular recruitment are reviewed, including coatings of biomimetic peptides and heparin.
Collapse
Affiliation(s)
- Daniel Radke
- Department of Biomedical Engineering, Michigan Technological University, 1400 Townsend Drive, Houghton, MI 49931, U.S
| | - Wenkai Jia
- Department of Biomedical Engineering, Michigan Technological University, 1400 Townsend Drive, Houghton, MI 49931, U.S
| | - Dhavan Sharma
- Department of Biomedical Engineering, Michigan Technological University, 1400 Townsend Drive, Houghton, MI 49931, U.S
| | - Kemin Fena
- Department of Biomedical Engineering, Michigan Technological University, 1400 Townsend Drive, Houghton, MI 49931, U.S
| | - Guifang Wang
- Department of Biomedical Engineering, Michigan Technological University, 1400 Townsend Drive, Houghton, MI 49931, U.S
| | - Jeremy Goldman
- Department of Biomedical Engineering, Michigan Technological University, 1400 Townsend Drive, Houghton, MI 49931, U.S
| | - Feng Zhao
- Department of Biomedical Engineering, Michigan Technological University, 1400 Townsend Drive, Houghton, MI 49931, U.S
| |
Collapse
|
152
|
Chen J, Wei J, Huang Y, Ma Y, Ni J, Li M, Zhu Y, Gao X, Fan G. Danhong Injection Enhances the Therapeutic Efficacy of Mesenchymal Stem Cells in Myocardial Infarction by Promoting Angiogenesis. Front Physiol 2018; 9:991. [PMID: 30093864 PMCID: PMC6070728 DOI: 10.3389/fphys.2018.00991] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 07/06/2018] [Indexed: 01/07/2023] Open
Abstract
Stem cell-based therapies have the potential to dramatically transform the treatment and prognosis of myocardial infarction (MI), and mesenchymal stem cells (MSCs) have been suggested as a promising cell population to ameliorate the heart remodeling in post-MI. However, poor implantation and survival in ischemic myocardium restrict its efficacy and application. In this study, we sought to use the unique mode of action of Chinese medicine to improve this situation. Surrounding the myocardial infarct area, we performed a multi-point MSC transplantation and administered in conjunction with Danhong injection, which is mainly used for the treatment of MI. Our results showed that the MSC survival rate and cardiac function were improved significantly through the small animal imaging system and echocardiography, respectively. Moreover, histological analysis showed that MSC combined with DHI intervention significantly reduced myocardial infarct size in myocardial infarcted mice and significantly increased MSC resident. To investigate the mechanism of DHI promoting MSC survival and cell migration, PCR and WB experiments were performed. Our results showed that DHI could promote the expression of CXC chemokine receptor 4 in MSC and enhance the expression of stromal cell–derived factor-1 in myocardium, and this effect can be inhibited by AMD3100 (an SDF1/CXCR4 antagonist). Additionally, MSC in combination with DHI interfered with MI in mice and this signifies that when combined, the duo could the expression of vascular endothelial growth factor (VEGF) in the marginal zone of infarction compared with when either MSC or DHI are used individually. Based on these results, we conclude that DHI enhances the residence of MSCs in cardiac tissue by modulating the SDF1/CXCR4 signaling pathway. These findings have important therapeutic implications for Chinese medicine-assisted cell-based therapy strategies.
Collapse
Affiliation(s)
- Jingrui Chen
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jing Wei
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuting Huang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuling Ma
- Oxford Chinese Medicine Research Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Jingyu Ni
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Min Li
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yan Zhu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiumei Gao
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
153
|
Blocki A, Beyer S, Jung F, Raghunath M. The controversial origin of pericytes during angiogenesis - Implications for cell-based therapeutic angiogenesis and cell-based therapies. Clin Hemorheol Microcirc 2018; 69:215-232. [PMID: 29758937 DOI: 10.3233/ch-189132] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Pericytes reside within the basement membrane of small vessels and are often in direct cellular contact with endothelial cells, fulfilling important functions during blood vessel formation and homeostasis. Recently, these pericytes have been also identified as mesenchymal stem cells. Mesenchymal stem cells, and especially their specialized subpopulation of pericytes, represent promising candidates for therapeutic angiogenesis applications, and have already been widely applied in pre-clinical and clinical trials. However, cell-based therapies of ischemic diseases (especially of myocardial infarction) have not resulted in significant long-term improvement. Interestingly, pericytes from a hematopoietic origin were observed in embryonic skin and a pericyte sub-population expressing leukocyte and monocyte markers was described during adult angiogenesis in vivo. Since mesenchymal stem cells do not express hematopoietic markers, the latter cell type might represent an alternative pericyte population relevant to angiogenesis. Therefore, we sourced blood-derived angiogenic cells (BDACs) from monocytes that closely resembled hematopoietic pericytes, which had only been observed in vivo thus far. BDACs displayed many pericytic features and exhibited enhanced revascularization and functional tissue regeneration in a pre-clinical model of critical limb ischemia. Comparison between BDACs and mesenchymal pericytes indicated that BDACs (while resembling hematopoietic pericytes) enhanced early stages of angiogenesis, such as endothelial cell sprouting. In contrast, mesenchymal pericytes were responsible for blood vessel maturation and homeostasis, while reducing endothelial sprouting.Since the formation of new blood vessels is crucial during therapeutic angiogenesis or during integration of implants into the host tissue, hematopoietic pericytes (and therefore BDACs) might offer an advantageous addition or even an alternative for cell-based therapies.
Collapse
Affiliation(s)
- Anna Blocki
- Institute for Tissue Engineering and Regenerative Medicine, Chinese University of Hong Kong, Hong Kong SAR.,School of Biomedical Science, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong SAR
| | - Sebastian Beyer
- Institute for Tissue Engineering and Regenerative Medicine, Chinese University of Hong Kong, Hong Kong SAR
| | - Friedrich Jung
- Institute for Clinical Hemostasiology and Transfusion Medicine, University Saarland, Homburg/Saar, Germany
| | - Michael Raghunath
- Institute of Chemistry and Biotechnology, Zurich University of Applied Sciences, Wädenswil, Switzerland
| |
Collapse
|
154
|
van Rhijn-Brouwer FCC, Gremmels H, Fledderus JO, Verhaar MC. Mesenchymal Stromal Cell Characteristics and Regenerative Potential in Cardiovascular Disease: Implications for Cellular Therapy. Cell Transplant 2018; 27:765-785. [PMID: 29895169 PMCID: PMC6047272 DOI: 10.1177/0963689717738257] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Administration of mesenchymal stromal cells (MSCs) is a promising strategy to treat cardiovascular disease (CVD). As progenitor cells may be negatively affected by both age and comorbidity, characterization of MSC function is important to guide decisions regarding use of allogeneic or autologous cells. Definitive answers on which factors affect MSC function can also aid in selecting which MSC donors would yield the most therapeutically efficacious MSCs. Here we provide a narrative review of MSC function in CVD based on a systematic search. A total of 41 studies examining CVD-related MSC (dys)function were identified. These data show that MSC characteristics and regenerative potential are often affected by CVD. However, studies presented conflicting results, and directed assessment of MSC parameters relevant to regenerative medicine applications was lacking in many studies. The predictive ability of in vitro assays for in vivo efficacy was rarely assessed. There was no correlation between quality of study reporting and study findings. Age mismatch was also not associated with study findings or effect size. Future research should focus on assays that assess regenerative potential in MSCs and parameters that relate to clinical success.
Collapse
Affiliation(s)
- F C C van Rhijn-Brouwer
- 1 Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - H Gremmels
- 1 Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - J O Fledderus
- 1 Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - M C Verhaar
- 1 Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
155
|
Potz BA, Scrimgeour LA, Pavlov VI, Sodha NR, Abid MR, Sellke FW. Extracellular Vesicle Injection Improves Myocardial Function and Increases Angiogenesis in a Swine Model of Chronic Ischemia. J Am Heart Assoc 2018; 7:e008344. [PMID: 29895586 PMCID: PMC6220556 DOI: 10.1161/jaha.117.008344] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 05/04/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Mesenchymal stem cell-derived extracellular vesicles (EVs) are believed to be cardioprotective in myocardial infarct. The objective of this study was to examine the effects of human mesenchymal cell-derived EV injection on cardiac function, myocardial blood flow, and vessel density in the setting of chronic myocardial ischemia. METHODS AND RESULTS Twenty-three Yorkshire swine underwent placement of an ameroid constrictor on their left circumflex artery. Two weeks later, the animals were split into 2 groups: the control group (CON; n=7) and the EV myocardial injection group (MVM; n=10). The MVM group underwent myocardial injection of 50 μg of EVs in 2 mL 0.9% saline into the ischemic myocardium. Five weeks later, the pigs underwent a harvest procedure, and the left ventricular myocardium was analyzed. Absolute blood flow and the ischemic/nonischemic myocardial perfusion ratio were increased in the ischemic myocardium in the MVM group compared with the CON group. Pigs in the MVM group had increased capillary and arteriolar density in the ischemic myocardial tissue compared with CON pigs. There was an increase in expression of the phospho-mitogen-activated protein kinase/mitogen-activated protein kinase ratio, the phospho-endothelial nitric oxide synthase/endothelial nitric oxide synthase ratio, and total protein kinase B in the MVM group compared with CON. There was an increase in cardiac output and stroke volume in the MVM group compared with CON. CONCLUSIONS In the setting of chronic myocardial ischemia, myocardial injection of human mesenchymal cell-derived EVs increases blood flow to ischemic myocardial tissue by induction of capillary and arteriolar growth via activation of the protein kinase B/endothelial nitric oxide synthase and mitogen-activated protein kinase signaling pathways resulting in increased cardiac output and stroke volume.
Collapse
Affiliation(s)
- Brittany A Potz
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Laura A Scrimgeour
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Vasile I Pavlov
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Neel R Sodha
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - M Ruhul Abid
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| | - Frank W Sellke
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Alpert Medical School of Brown University, Providence, RI
| |
Collapse
|
156
|
Lee SG, Joe YA. Autophagy mediates enhancement of proangiogenic activity by hypoxia in mesenchymal stromal/stem cells. Biochem Biophys Res Commun 2018; 501:941-947. [PMID: 29772235 DOI: 10.1016/j.bbrc.2018.05.086] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 05/14/2018] [Indexed: 01/07/2023]
Abstract
Mesenchymal stromal/stem cells (MSCs) have been promising source for regenerative cell therapy in ischemic diseases. To improve efficacy of MSC therapy, various priming methods have been developed, and hypoxic priming has been reported to enhance therapeutic efficacy of MSCs by increasing secretion level of growth factors and cytokines. Recently, it has been reported that bone marrow MSCs primed with hypoxic condition show an increase of autophagy. Here, we addressed whether proangiogenic activity increased by hypoxic condition is associated with autophagy. Wharton's jelly derived MSCs primed with hypoxia showed increase of autophagy with increased hypoxia inducible factor-1α level, and conditioned medium (CM) derived from these cells showed increased levels of migration and tube formation of human umbilical vein endothelial cells (HUVECs) compared to non-primed MSCs-derived CM. Pretreatment with autophagy inhibitor 3-methyladenine or chloroquine prior to exposure of hypoxia resulted in reduction of migration and tube formation of HUVECs. CM obtained under hypoxic condition from MSCs in which autophagy activity was inhibited by ATG5 and ATG7 siRNA treatment also showed decrease of migration and tube formation of HUVECs. Accordingly, secretion levels of angiogenin and VEGF that were markedly increased upon hypoxia exposure was decreased by ATG5/7 knockdown. Therefore, it may be suggested that autophagy plays an important role in hypoxia-driven enhancement of paracrine effect of MSCs.
Collapse
Affiliation(s)
- Seul-Gi Lee
- Cancer Research Institute, Department of Medical Lifescience, and Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Young Ae Joe
- Cancer Research Institute, Department of Medical Lifescience, and Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea.
| |
Collapse
|
157
|
Li Y, Wang Z, Zhao Y, Luo Y, Xu W, Marion TN, Liu Y. Successful mesenchymal stem cell treatment of leg ulcers complicated by Behcet disease: A case report and literature review. Medicine (Baltimore) 2018; 97:e0515. [PMID: 29668637 PMCID: PMC5916712 DOI: 10.1097/md.0000000000010515] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
RATIONALE Behçet disease (BD) is a recurrent vasculitis characterized by oral and genital mucous membrane ulcers, uveitis, and skin lesions but only rarely leg ulcers. To our knowledge, no efficacious therapy has been described for BD patients with complicating, destructive leg ulcers. PATIENT CONCERNS Here, We report the case of a 55-year-old woman with generalized erythema nodosum-like, papulopustular lesions, recurrent oral and genital ulcers accompanied with recurrent leg ulcers and trouble walking. DIAGNOSES Based upon the patient's clinical feature and positive pathergy test , BD was confirmed. INTERVENTIONS Conventional immunosuppressive therapy and anti-tumor necrosis factor inhibitors, adalimumab and etanercept, had no demonstrable clinical effect. Mesenchymal stem cell (MSC) injection combined with low-dose prednisone and thalidomide, however, completely ameliorated the ulcers on one leg, significantly improved ulcers on the other leg, and returned normal function to both legs. OUTCOMES The ulcerative lesions remained in remission, and the affected leg functioned normally after 34 months' follow-up. LESSONS Our experience suggests that MSC infusion might be a potentially successful therapy for intractable drug-resistant BD patients with concomitant leg ulcer.
Collapse
Affiliation(s)
- Yanhong Li
- Department of Rheumatology, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Zhongming Wang
- Department of Rheumatology, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Yi Zhao
- Department of Rheumatology, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Yubin Luo
- Department of Rheumatology, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Wangdong Xu
- Department of Rheumatology, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Tony N. Marion
- Department of Microbiology, Immunology, and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN
| | - Yi Liu
- Department of Rheumatology, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| |
Collapse
|
158
|
Comparison of trophic factors secreted from human adipose-derived stromal vascular fraction with those from adipose-derived stromal/stem cells in the same individuals. Cytotherapy 2018; 20:589-591. [DOI: 10.1016/j.jcyt.2018.02.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 02/03/2018] [Accepted: 02/05/2018] [Indexed: 12/22/2022]
|
159
|
Gaubys A, Papeckys V, Pranskunas M. Use of Autologous Stem Cells for the Regeneration of Periodontal Defects in Animal Studies: a Systematic Review and Meta-Analysis. J Oral Maxillofac Res 2018; 9:e3. [PMID: 30116515 PMCID: PMC6090251 DOI: 10.5037/jomr.2018.9203] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 05/24/2018] [Indexed: 12/14/2022]
Abstract
OBJECTIVES To overview preclinical animal trials and quantify the effect size that stem cell therapy has on the regeneration of periodontal tissue complex. MATERIAL AND METHODS A systematic MEDLINE (PubMed) online library search was conducted for preclinical animal studies in vivo , using autologous periodontal ligament, dental pulp, cementum, alveolar periosteal, gingival margin or adipose stem cell types for periodontal tissue complex regeneration purposes. Studies had to be published between 2007.09.01 and 2017.09.01 in the English language. RESULTS Online library search yielded 2099 results. After the title, abstract and full-text screening ten studies fit inclusion criteria and were pooled into meta-analysis. Overall the stem cell regenerative therapy had a statistically significant positive influence on the periodontal tissue regeneration when compared to the control groups. The biggest influence was made to the regeneration of cementum (standardised mean difference [SMD] 2.25 [95% confidence interval (CI) = 1.31 to 3.2]) while the smallest influence was made to the alveolar bone (SMD 1.47 [95% CI = 0.7 to 2.25]) the effect size for periodontal ligament regeneration was (SMD 1.8 [95% CI = 1 to 2.59]). Subgroup analysis showed statistically significant (P < 0.05) differences between different cell types in the alveolar bone and cementum regeneration groups and in alveolar bone group in relation to scaffold materials. CONCLUSIONS Stem cell therapy has a positive impact on periodontal tissue complex regeneration. Such therapy has the biggest influence on cementum regeneration meanwhile alveolar bone regeneration is influenced by the least amount. However more and less diverse preclinical studies are needed to have a greater statistical power in future meta-analyses.
Collapse
Affiliation(s)
- Algimantas Gaubys
- Department of Maxillofacial Surgery, Lithuanian University of Health Sciences, KaunasLithuania.
| | - Valdas Papeckys
- Department of Maxillofacial Surgery, Lithuanian University of Health Sciences, KaunasLithuania.
| | - Mindaugas Pranskunas
- Department of Maxillofacial Surgery, Lithuanian University of Health Sciences, KaunasLithuania.
| |
Collapse
|
160
|
Gharaei MA, Xue Y, Mustafa K, Lie SA, Fristad I. Human dental pulp stromal cell conditioned medium alters endothelial cell behavior. Stem Cell Res Ther 2018; 9:69. [PMID: 29562913 PMCID: PMC5861606 DOI: 10.1186/s13287-018-0815-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 02/01/2018] [Accepted: 02/22/2018] [Indexed: 12/13/2022] Open
Abstract
Background Angiogenesis is of utmost importance for tissue regeneration and repair. Human dental pulp stromal cells (hDPSCs) possess angiogenic potential, as they secrete paracrine factors that may alter the host microenvironment. However, more insight into how hDPSCs guide endothelial cells (ECs) in a paracrine fashion is yet to be obtained. Therefore, the current study aimed to investigate the effect(s) of conditioned medium derived from hDPSCs (hDPSC-CM) on EC behavior in vitro. Methods hDPSCs were harvested from third molars scheduled for surgical removal under informed consent. The angiogenic profile of hDPSC-CM was identified using human angiogenesis antibody array and enzyme-linked immunosorbent assay (ELISA). Using real-time reverse transcription-polymerase chain reaction (RT-PCR) and ELISA, the mRNA and protein expression level of specific angiogenic biomarkers was determined in human umbilical vein endothelial cells (HUVECs) exposed to hDPSC-CM. The effect of hDPSC-CM on HUVEC attachment, proliferation and migration was evaluated by crystal violet staining, MTT, transwell migration along with real-time cell monitoring assays (xCELLigence; ACEA Biosciences, Inc.). A Matrigel assay was included to examine the influence of hDPSC-CM on HUVEC network formation. Endothelial growth medium (EGM-2) and EGM-2 supplemented with hDPSC-CM served as experimental groups, whereas endothelial basal medium (EBM-2) was set as negative control. Results A wide range of proangiogenic and antiangiogenic factors, including vascular endothelial growth factor, tissue inhibitor of metalloproteinase protein 1, plasminogen activator inhibitor (serpin E1), urokinase plasminogen activator and stromal cell-derived factor 1, was abundantly detected in hDPSC-CM by protein profiling array and ELISA. hDPSC-CM significantly accelerated the adhesion phases, from sedimentation to attachment and spreading, the proliferation rate and migration of HUVECs as shown in both endpoint assays and real-time cell analysis recordings. Furthermore, Matrigel assay demonstrated that hDPSC-CM stimulated tubulogenesis, affecting angiogenic parameters such as the number of nodes, meshes and total tube length. Conclusions The sustained proangiogenic and promaturation effects of hDPSC-CM shown in this in vitro study strongly suggest that the trophic factors released by hDPSCs are able to trigger pronounced angiogenic responses, even beyond EGM-2 considered as an optimal culture condition for ECs.
Collapse
Affiliation(s)
- M A Gharaei
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Årstadveien 19, N-5009, Bergen, Norway
| | - Y Xue
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Årstadveien 19, N-5009, Bergen, Norway
| | - K Mustafa
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Årstadveien 19, N-5009, Bergen, Norway
| | - S A Lie
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Årstadveien 19, N-5009, Bergen, Norway
| | - I Fristad
- Department of Clinical Dentistry, Faculty of Medicine, University of Bergen, Årstadveien 19, N-5009, Bergen, Norway.
| |
Collapse
|
161
|
Comella K, Parlo M, Daly R, Dominessy K. First-in-man intravenous implantation of stromal vascular fraction in psoriasis: a case study. Int Med Case Rep J 2018; 11:59-64. [PMID: 29606893 PMCID: PMC5868735 DOI: 10.2147/imcrj.s163612] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Stromal vascular fraction (SVF) is a mixture of adipose-derived stem cells/mesenchymal stem cells, endothelial/progenitors, pericytes, fibroblasts, and other cells obtained from fat tissue. A small sample of fat or adipose tissue can be obtained under local anesthesia using a cannula. After an enzymatic digestion and centrifugation, the adipocytes (fat cells) are removed to obtain an SVF. Here, we describe the rationale and, to our knowledge, the first clinical implementation of SVF intravenously in a patient with severe psoriasis. METHODS Adipose tissue (60 mL) was collected under local anesthesia via a mini-lipoaspirate procedure. The SVF was separated from the adipocytes via centrifugation after an enzymatic digestion. The cells were resuspended in normal saline and injected via bolus push intravenous. The subject was monitored over a period of 12 months for safety (adverse events), medication changes, and quality of life parameters. RESULTS The patient did not report any safety concerns and did not experience any severe adverse events. The patient demonstrated a significant decrease in symptoms with a noticeable difference in skin quality appearance. Psoriasis area and severity index score went from 50.4 at baseline to 0.3 at 1 month follow-up. CONCLUSION Overall, the patient reported improved quality of life and willingness to continue treatments. This successful initial case study demonstrates that this may be a feasible treatment plan for patients suffering from psoriasis.
Collapse
|
162
|
He F, Luo PF, Tang T, Zhang F, Fang H, Ji SZ, Sun Y, Wu GS, Pan BH, Huo ZB, Wang GY, Xia ZF. Targeted release of stromal cell-derived factor-1α by reactive oxygen species-sensitive nanoparticles results in bone marrow stromal cell chemotaxis and homing, and repair of vascular injury caused by electrical burns. PLoS One 2018. [PMID: 29529067 PMCID: PMC5847229 DOI: 10.1371/journal.pone.0194298] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Rapid repair of vascular injury is an important prognostic factor for electrical burns. This repair is achieved mainly via stromal cell-derived factor (SDF)-1α promoting the mobilization, chemotaxis, homing, and targeted differentiation of bone marrow mesenchymal stem cells (BMSCs) into endothelial cells. Forming a concentration gradient from the site of local damage in the circulation is essential to the role of SDF-1α. In a previous study, we developed reactive oxygen species (ROS)-sensitive PPADT nanoparticles containing SDF-1α that could degrade in response to high concentration of ROS in tissue lesions, achieving the goal of targeted SDF-1α release. In the current study, a rat vascular injury model of electrical burns was used to evaluate the effects of targeted release of SDF-1α using PPADT nanoparticles on the chemotaxis of BMSCs and the repair of vascular injury. Continuous exposure to 220 V for 6 s could damage rat vascular endothelial cells, strip off the inner layer, significantly elevate the local level of ROS, and decrease the level of SDF-1α. After injection of Cy5-labeled SDF-1α-PPADT nanoparticles, the distribution of Cy5 fluorescence suggested that SDF-1α was distributed primarily at the injury site, and the local SDF-1α levels increased significantly. Seven days after injury with nanoparticles injection, aggregation of exogenous green fluorescent protein-labeled BMSCs at the injury site was observed. Ten days after injury, the endothelial cell arrangement was better organized and continuous, with relatively intact vascular morphology and more blood vessels. These results showed that SDF-1α-PPADT nanoparticles targeted the SDF-1α release at the site of injury, directing BMSC chemotaxis and homing, thereby promoting vascular repair in response to electrical burns.
Collapse
Affiliation(s)
- Fang He
- Department of Burn Surgery, the Second Military Medical University affiliated Changhai Hospital, Shanghai, China
- Department of Burn Surgery, the Nanjing Medical University affiliated Suzhou Hospital, Jiangsu, China
| | - Peng-Fei Luo
- Department of Burn Surgery, the Second Military Medical University affiliated Changhai Hospital, Shanghai, China
| | - Tao Tang
- Department of Burn Surgery, the Second Military Medical University affiliated Changhai Hospital, Shanghai, China
- Department of Surgery, Navy Hospital of PLA, Shanghai, China
| | - Fang Zhang
- Department of Burn Surgery, the Second Military Medical University affiliated Changhai Hospital, Shanghai, China
| | - He Fang
- Department of Burn Surgery, the Second Military Medical University affiliated Changhai Hospital, Shanghai, China
| | - Shi-Zhao Ji
- Department of Burn Surgery, the Second Military Medical University affiliated Changhai Hospital, Shanghai, China
| | - Yu Sun
- Department of Burn Surgery, the Second Military Medical University affiliated Changhai Hospital, Shanghai, China
| | - Guo-Sheng Wu
- Department of Burn Surgery, the Second Military Medical University affiliated Changhai Hospital, Shanghai, China
| | - Bo-Han Pan
- Department of Burn Surgery, the Second Military Medical University affiliated Changhai Hospital, Shanghai, China
| | - Zhi-Bao Huo
- School of Environmental Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
- * E-mail: (ZBH); (GYW); (ZFX)
| | - Guang-Yi Wang
- Department of Burn Surgery, the Second Military Medical University affiliated Changhai Hospital, Shanghai, China
- * E-mail: (ZBH); (GYW); (ZFX)
| | - Zhao-Fan Xia
- Department of Burn Surgery, the Second Military Medical University affiliated Changhai Hospital, Shanghai, China
- * E-mail: (ZBH); (GYW); (ZFX)
| |
Collapse
|
163
|
Ratushnyy A, Ezdakova M, Yakubets D, Buravkova L. Angiogenic Activity of Human Adipose-Derived Mesenchymal Stem Cells Under Simulated Microgravity. Stem Cells Dev 2018; 27:831-837. [PMID: 29431030 DOI: 10.1089/scd.2017.0262] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Multipotent mesenchymal stem/stromal cells (MSCs) are strongly involved in tissue homeostasis mainly through paracrine regulation. In this study, we examined the influence of simulated microgravity on the angiogenic potential of adipose-derived MSCs (ASCs). The conditioned medium (CM) from random positioning machine (RPM)-exposed ASCs stimulated the formation of vessel network in ovo, endothelial cell (EC) capillary-like network, and nondirected EC migration in vitro. These effects were driven by alteration of both angiogenesis-related gene and protein expression. The elevation of angiogenic regulators Serpin E1, Serpin F1, IGFBP, VEGF, and IL-8 was detected in ASC-CM after 3D-clinorotation. In addition, transcription of genes encoding growth factors with proangiogenic activity were upregulated including VEGF-c and VEGF-a. These data evidenced that besides direct effect on ECs, microgravity could provoke MSC-mediating specific microenvironment for ECs supporting their functions, that is, proliferation and migration via increased production of IL-8 and VEGF as well as other paracrine factors involved in angiogenesis regulation.
Collapse
Affiliation(s)
- Andrey Ratushnyy
- Cell Physiology Laboratory, Institute of Biomedical Problems of Russian Academy of Sciences , Moscow, Russia
| | - Maria Ezdakova
- Cell Physiology Laboratory, Institute of Biomedical Problems of Russian Academy of Sciences , Moscow, Russia
| | - Danila Yakubets
- Cell Physiology Laboratory, Institute of Biomedical Problems of Russian Academy of Sciences , Moscow, Russia
| | - Ludmila Buravkova
- Cell Physiology Laboratory, Institute of Biomedical Problems of Russian Academy of Sciences , Moscow, Russia
| |
Collapse
|
164
|
Wang X, Zhang J, Cui W, Fang Y, Li L, Ji S, Mao D, Ke T, Yao X, Ding D, Feng G, Kong D. Composite Hydrogel Modified by IGF-1C Domain Improves Stem Cell Therapy for Limb Ischemia. ACS APPLIED MATERIALS & INTERFACES 2018; 10:4481-4493. [PMID: 29327586 DOI: 10.1021/acsami.7b17533] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Stem cell treatment for critical limb ischemia yields a limited therapeutic effect due to cell loss and dysfunction caused by local ischemic environment. Biomimetic scaffolds emerge as ideal cell delivery vehicles for regulating cell fate via mimicking the components of stem cell niche. Herein, we prepared a bioactive hydrogel by mixing chitosan and hyaluronic acid that is immobilized with C domain peptide of insulin-like growth factor 1 (IGF-1C) and examined whether this hydrogel could augment stem cell survival and therapeutic potential. Our results showed that IGF-1C-modified hydrogel increased in vitro viability and proangiogenic activity of adipose-derived stromal cells (ADSCs). Moreover, cotransplantation of hydrogel and ADSCs into ischemic hind limbs of mice effectively ameliorated blood perfusion and muscle regeneration, leading to superior limb salvage. These therapeutic effects can be ascribed to improved ADSC retention, angiopoientin-1 secretion, and neovascularization, as well as reduced inflammatory cell infiltration. Additionally, hydrogel enhanced antifibrotic activity of ADSCs, as evidenced by decreased collagen accumulation at late stage. Together, our findings indicate that composite hydrogel modified by IGF-1C could promote survival and proangiogenic capacity of ADSCs and thereby represents a feasible option for cell-based treatment for critical limb ischemia.
Collapse
Affiliation(s)
- Xiaomin Wang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University , Tianjin 300071, China
| | - Jimin Zhang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University , Tianjin 300071, China
| | - Weilong Cui
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University , Tianjin 300071, China
| | - Yuan Fang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University , Tianjin 300071, China
| | - Li Li
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University , Tianjin 300071, China
- Department of Endocrinology, The Second Affiliated Hospital, Kunming Medical University , Kunming 650101, Yunnan, China
| | - Shenglu Ji
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University , Tianjin 300071, China
| | - Duo Mao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University , Tianjin 300071, China
| | - Tingyu Ke
- Department of Endocrinology, The Second Affiliated Hospital, Kunming Medical University , Kunming 650101, Yunnan, China
| | - Xin Yao
- Department of Genitourinary Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer , Tianjin 300060, China
| | - Dan Ding
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University , Tianjin 300071, China
| | - Guowei Feng
- Department of Genitourinary Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer , Tianjin 300060, China
| | - Deling Kong
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University , Tianjin 300071, China
| |
Collapse
|
165
|
Krajewska-Włodarczyk M, Owczarczyk-Saczonek A, Placek W, Osowski A, Engelgardt P, Wojtkiewicz J. Role of Stem Cells in Pathophysiology and Therapy of Spondyloarthropathies-New Therapeutic Possibilities? Int J Mol Sci 2017; 19:ijms19010080. [PMID: 29283375 PMCID: PMC5796030 DOI: 10.3390/ijms19010080] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Revised: 12/23/2017] [Accepted: 12/25/2017] [Indexed: 12/14/2022] Open
Abstract
Considerable progress has been made recently in understanding the complex pathogenesis and treatment of spondyloarthropathies (SpA). Currently, along with traditional disease modifying anti-rheumatic drugs (DMARDs), TNF-α, IL-12/23 and IL-17 are available for treatment of such diseases as ankylosing spondylitis (AS) and psoriatic arthritis (PsA). Although they adequately control inflammatory symptoms, they do not affect the abnormal bone formation processes associated with SpA. However, the traditional therapeutic approach does not cover the regenerative treatment of damaged tissues. In this regards, stem cells may offer a promising, safe and effective therapeutic option. The aim of this paper is to present the role of mesenchymal stromal cells (MSC) in pathogenesis of SpA and to highlight the opportunities for using stem cells in regenerative processes and in the treatment of inflammatory changes in articular structures.
Collapse
Affiliation(s)
- Magdalena Krajewska-Włodarczyk
- Department of Rheumatology, Municipal Hospital in Olsztyn, 10-900 Olsztyn, Poland.
- Department of Pathophysiology, Faculty of Medicine, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| | - Agnieszka Owczarczyk-Saczonek
- Department of Dermatology, Sexually Transmitted Diseases and Clinical Immunology, Faculty of Medicine, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| | - Waldemar Placek
- Department of Dermatology, Sexually Transmitted Diseases and Clinical Immunology, Faculty of Medicine, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| | - Adam Osowski
- Department of Pathophysiology, Faculty of Medicine, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| | - Piotr Engelgardt
- Department of Forensic Medicine, Faculty of Medicine, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
| | - Joanna Wojtkiewicz
- Department of Pathophysiology, Faculty of Medicine, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
- Laboratory for Regenerative Medicine, Faculty of Medicine, University of Warmia and Mazury, 10-900 Olsztyn, Poland.
- Foundation for Nerve Cell Regeneration, University of Warmia and Mazury in Olsztyn, 10-900 Olsztyn, Poland.
| |
Collapse
|
166
|
Abstract
Every year 13.3 million people suffer acute kidney injury (AKI), which is associated with a high risk of death or development of long-term chronic kidney disease (CKD) in a substantial percentage of patients besides other organ dysfunctions. To date, the mortality rate per year for AKI exceeds 50 % at least in patients requiring early renal replacement therapy and is higher than the mortality for breast and prostate cancer, heart failure and diabetes combined.Until now, no effective treatments able to accelerate renal recovery and improve survival post AKI have been developed. In search of innovative and effective strategies to foster the limited regeneration capacity of the kidney, several studies have evaluated the ability of mesenchymal stem cells (MSCs) of different origin as an attractive therapeutic tool. The results obtained in several models of AKI and CKD document that MSCs have therapeutic potential in repair of renal injury, preserving renal function and structure thus prolonging animal survival through differentiation-independent pathways. In this chapter, we have summarized the mechanisms underlying the regenerative processes triggered by MSC treatment, essentially due to their paracrine activity. The capacity of MSC to migrate to the site of injury and to secrete a pool of growth factors and cytokines with anti-inflammatory, mitogenic, and immunomodulatory effects is described. New modalities of cell-to-cell communication via the release of microvesicles and exosomes by MSCs to injured renal cells will also be discussed. The translation of basic experimental data on MSC biology into effective care is still limited to preliminary phase I clinical trials and further studies are needed to definitively assess the efficacy of MSC-based therapy in humans.
Collapse
Affiliation(s)
- Marina Morigi
- IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87, 24126, Bergamo, Italy.
| | - Cinzia Rota
- IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87, 24126, Bergamo, Italy
| | - Giuseppe Remuzzi
- IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Via Stezzano 87, 24126, Bergamo, Italy
- Unit of Nephrology and Dialysis, A.O. Papa Giovanni XXIII, 24127, Bergamo, Italy
| |
Collapse
|
167
|
Mohammadi Najafabadi M, Shamsasenjan K, Akbarzadehlaleh P. The Angiogenic Chemokines Expression Profile of Myeloid Cell Lines Co-Cultured with Bone Marrow-Derived Mesenchymal Stem Cells. CELL JOURNAL 2017; 20:19-24. [PMID: 29308614 PMCID: PMC5759676 DOI: 10.22074/cellj.2018.4924] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/03/2017] [Indexed: 11/04/2022]
Abstract
OBJECTIVES Angiogenesis, the process of formation of new blood vessels, is essential for development of solid tumors. At first, it was first assumed that angiogenesis is not implicated in the development of acute myeloid leukemia (AML) as a liquid tumor. One of the most important elements in bone marrow microenvironment is mesenchymal stem cells (MSCs). These cells possess an intrinsic tropism for sites of tumor in various types of cancers and have an impact on solid tumors growth by affecting the angiogenic process. But so far, our knowledge is limited about MSCs' role in liquid tumors angiogenesis. By increasing our knowledge about the role of MSCs on angiogenesis, new therapeutic strategies can be used to improve the status of patients with leukemia. MATERIALS AND METHODS In this experimental study, HL-60, K562 and U937 cells were separately co-cultured with bone marrow derived-MSCs and after 8, 16 and 24 hours, alterations in the expression of 10 chemokine genes involved in angiogenesis, were evaluated by quantitative real time-polymerase chain reaction (qRT-PCR). Mono-cultures of leukemia cell lines were used as controls. RESULTS We observed that in HL-60 and K562 cells co-cultured with MSCs, the expression of CXCL10 and CXCL3 genes are increased, respectively as compared to the control cells. Also, in U937 cells co-cultured with MSCs, the expression of CXCL6 gene was upgraded. Moreover in U937 cells, CCL2 gene expression in the first 16 hours was lower than the control cells, while within 24 hours its expression augmented. CONCLUSIONS Our observations, for the first time, demonstrated that bone marrow (BM)-MSCs are able to alter the expression profile of chemokine genes involved in angiogenesis, in acute myeloid leukemia cell lines. MSCs cause different effects on angiogenesis in different leukemia cell lines; in some cases, MSCs promote angiogenesis, and in others, inhibit it.
Collapse
Affiliation(s)
| | - Karim Shamsasenjan
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parvin Akbarzadehlaleh
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
168
|
Javorkova E, Vackova J, Hajkova M, Hermankova B, Zajicova A, Holan V, Krulova M. The effect of clinically relevant doses of immunosuppressive drugs on human mesenchymal stem cells. Biomed Pharmacother 2017; 97:402-411. [PMID: 29091890 DOI: 10.1016/j.biopha.2017.10.114] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 10/17/2017] [Accepted: 10/21/2017] [Indexed: 12/15/2022] Open
Abstract
Immunosuppressive drugs are used to suppress graft rejection after transplantation and for the treatment of various diseases. The main limitations of their use in clinical settings are severe side effects, therefore alternative approaches are desirable. In this respect, mesenchymal stem cells (MSCs) possess a regenerative and immunomodulatory capacity that has generated considerable interest for their use in cell-based therapy. Currently, MSCs are tested in many clinical trials, including the treatment of diseases which require simultaneous immunosuppressive treatment. Since the molecular targets of immunosuppressive drugs are also present in MSCs, we investigated whether immunosuppressive drugs interact with the activity of MSCs. Human MSCs isolated from the bone marrow (BM) or adipose tissue (AT) were cultured in the presence of clinical doses of five widely used immunosuppressive drugs (cyclosporine A, mycophenolate mofetil, rapamycin, prednisone and dexamethasone), and the influence of these drugs on several factors related to the immunosuppressive properties of MSCs, including the expression of immunomodulatory enzymes, various growth factors, cytokines, chemokines, adhesion molecules and proapoptotic ligands, was assessed. Glucocorticoids, especially dexamethasone, showed the most prominent effects on both types of MSCs and suppressed the expression of the majority of the factors that were tested. A significant increase of hepatocyte growth factor production in AT-MSCs and of indoleamine 2,3-dioxygenase expression in both types of MSCs were the only exceptions. In conclusion, clinically relevant doses of inhibitors of calcineurin, mTOR and IMPDH and glucocorticoids interfere with MSC functions, but do not restrain their immunosuppressive properties. These findings should be taken into account before preparing immunosuppressive strategies combining the use of immunosuppressive drugs and MSCs.
Collapse
Affiliation(s)
- Eliska Javorkova
- Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, Prague 2, 128 44, Czech Republic; Department of Transplantation Immunology, The Czech Academy of Sciences, Institute of Experimental Medicine, Videnska 1083, Prague 4, 142 20, Czech Republic.
| | - Julie Vackova
- Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, Prague 2, 128 44, Czech Republic.
| | - Michaela Hajkova
- Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, Prague 2, 128 44, Czech Republic; Department of Transplantation Immunology, The Czech Academy of Sciences, Institute of Experimental Medicine, Videnska 1083, Prague 4, 142 20, Czech Republic.
| | - Barbora Hermankova
- Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, Prague 2, 128 44, Czech Republic; Department of Transplantation Immunology, The Czech Academy of Sciences, Institute of Experimental Medicine, Videnska 1083, Prague 4, 142 20, Czech Republic.
| | - Alena Zajicova
- Department of Transplantation Immunology, The Czech Academy of Sciences, Institute of Experimental Medicine, Videnska 1083, Prague 4, 142 20, Czech Republic.
| | - Vladimir Holan
- Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, Prague 2, 128 44, Czech Republic; Department of Transplantation Immunology, The Czech Academy of Sciences, Institute of Experimental Medicine, Videnska 1083, Prague 4, 142 20, Czech Republic.
| | - Magdalena Krulova
- Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, Prague 2, 128 44, Czech Republic; Department of Transplantation Immunology, The Czech Academy of Sciences, Institute of Experimental Medicine, Videnska 1083, Prague 4, 142 20, Czech Republic.
| |
Collapse
|
169
|
Eslani M, Putra I, Shen X, Hamouie J, Afsharkhamseh N, Besharat S, Rosenblatt MI, Dana R, Hematti P, Djalilian AR. Corneal Mesenchymal Stromal Cells Are Directly Antiangiogenic via PEDF and sFLT-1. Invest Ophthalmol Vis Sci 2017; 58:5507-5517. [PMID: 29075761 PMCID: PMC5661382 DOI: 10.1167/iovs.17-22680] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Purpose To evaluate the angiogenic properties of corneal derived mesenchymal stromal cells (Co-MSC). Methods Co-MSCs were extracted from human cadaver, and wild-type (C57BL/6J) and SERPINF1−/− mice corneas. The MSC secretome was collected in a serum-free medium. Human umbilical vein endothelial cell (HUVEC) tube formation and fibrin gel bead assay (FIBA) sprout formation were used to assess the angiogenic properties of Co-MSC secretome. Complete corneal epithelial debridement was used to induce corneal neovascularization in wild-type mice. Co-MSCs embedded in fibrin gel was applied over the debrided cornea to evaluate the angiogenic effects of Co-MSCs in vivo. Immunoprecipitation was used to remove soluble fms-like tyrosine kinase-1 (sFLT-1) and pigment epithelium-derived factor (PEDF, SERPINF1 gene) from the Co-MSC secretome. Results Co-MSC secretome significantly inhibited HUVECs tube and sprout formation. Co-MSCs from different donors consistently contained high levels of antiangiogenic factors including sFLT-1 and PEDF; and low levels of the angiogenic factor VEGF-A. In vivo, application of Co-MSCs to mouse corneas after injury prevented the development of corneal neovascularization. Removing PEDF or sFLT-1 from the secretome significantly diminished the antiangiogenic effects of Co-MSCs. Co-MSCs isolated from SERPINF1−/− mice had significantly reduced antiangiogenic effects compared to SERPINF1+/+ (wild-type) Co-MSCs. Conclusions These results illustrate the direct antiangiogenic properties of Co-MSCs, the importance of sFLT-1 and PEDF, and their potential clinical application for preventing pathologic corneal neovascularization.
Collapse
Affiliation(s)
- Medi Eslani
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Ilham Putra
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Xiang Shen
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Judy Hamouie
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Neda Afsharkhamseh
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Soroush Besharat
- Department of Medicine and University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, Wisconsin, United States
| | - Mark I Rosenblatt
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, United States
| | - Reza Dana
- Schepens Eye Research Institute, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States
| | - Peiman Hematti
- Department of Medicine and University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, Wisconsin, United States
| | - Ali R Djalilian
- Department of Ophthalmology and Visual Sciences, University of Illinois at Chicago, Chicago, Illinois, United States
| |
Collapse
|
170
|
Bakopoulou A, Apatzidou D, Aggelidou E, Gousopoulou E, Leyhausen G, Volk J, Kritis A, Koidis P, Geurtsen W. Isolation and prolonged expansion of oral mesenchymal stem cells under clinical-grade, GMP-compliant conditions differentially affects "stemness" properties. Stem Cell Res Ther 2017; 8:247. [PMID: 29096714 PMCID: PMC5667471 DOI: 10.1186/s13287-017-0705-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 10/17/2017] [Accepted: 10/19/2017] [Indexed: 02/08/2023] Open
Abstract
Background Development of clinical-grade cell preparations is central to meeting the regulatory requirements for cellular therapies under good manufacturing practice-compliant (cGMP) conditions. Since addition of animal serum in culture media may compromise safe and efficient expansion of mesenchymal stem cells (MSCs) for clinical use, this study aimed to investigate the potential of two serum/xeno-free, cGMP culture systems to maintain long-term “stemness” of oral MSCs (dental pulp stem cells (DPSCs) and alveolar bone marrow MSCs (aBMMSCs)), compared to conventional serum-based expansion. Methods DPSC and aBMMSC cultures (n = 6/cell type) were established from pulp and alveolar osseous biopsies respectively. Three culture systems were used: StemPro_MSC/SFM_XenoFree (Life Technologies); StemMacs_MSC/XF (Miltenyi Biotek); and α-MEM (Life Technologies) with 15% fetal bovine serum. Growth (population doublings (PDs)), immunophenotypic (flow cytometric analysis of MSC markers) and senescence (β-galactosidase (SA-β-gal) activity; telomere length) characteristics were determined during prolonged expansion. Gene expression patterns of osteogenic (ALP, BMP-2), adipogenic (LPL, PPAR-γ) and chondrogenic (ACAN, SOX-9) markers and maintenance of multilineage differentiation potential were determined by real-time PCR. Results Similar isolation efficiency and stable growth dynamics up to passage 10 were observed for DPSCs under all expansion conditions. aBMMSCs showed lower cumulative PDs compared to DPSCs, and when StemMacs was used substantial delays in cell proliferation were noted after passages 6–7. Serum/xeno-free expansion produced cultures with homogeneous spindle-shaped phenotypes, while serum-based expansion preserved differential heterogeneous characteristics of each MSC population. Prolonged expansion of both MSC types but in particular the serum/xeno-free-expanded aBMMSCs was associated with downregulation of CD146, CD105, Stro-1, SSEA-1 and SSEA-4, but not CD90, CD73 and CD49f, in parallel with an increase of SA-gal-positive cells, cell size and granularity and a decrease in telomere length. Expansion under both serum-free systems resulted in “osteogenic pre-disposition”, evidenced by upregulation of osteogenic markers and elimination of chondrogenic and adipogenic markers, while serum-based expansion produced only minor changes. DPSCs retained a diminishing (CCM, StemPro) or increasing (StemMacs) mineralization potential with passaging, while aBMMSCs lost this potential after passages 6–7 under all expansion conditions. Conclusions These findings indicate there is still a vacant role for development of qualified protocols for clinical-grade expansion of oral MSCs; a key milestone achievement for translation of research from the bench to clinics. Electronic supplementary material The online version of this article (doi:10.1186/s13287-017-0705-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Athina Bakopoulou
- Department of Prosthodontics, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), GR-54124, Thessaloniki, Greece. .,cGMP Regenerative Medicine Facility, Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece. .,Department of Conservative Dentistry, Periodontology and Preventive Dentistry, Hannover Medical School (MHH), Hannover, Germany.
| | - Danae Apatzidou
- Department of Preventive Dentistry, Periodontology and Implant Biology, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece
| | - Eleni Aggelidou
- Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece.,cGMP Regenerative Medicine Facility, Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece
| | - Evangelia Gousopoulou
- Department of Preventive Dentistry, Periodontology and Implant Biology, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece.,Department of Conservative Dentistry, Periodontology and Preventive Dentistry, Hannover Medical School (MHH), Hannover, Germany
| | - Gabriele Leyhausen
- Department of Conservative Dentistry, Periodontology and Preventive Dentistry, Hannover Medical School (MHH), Hannover, Germany
| | - Joachim Volk
- Department of Conservative Dentistry, Periodontology and Preventive Dentistry, Hannover Medical School (MHH), Hannover, Germany
| | - Aristeidis Kritis
- Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece.,cGMP Regenerative Medicine Facility, Department of Physiology and Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), Thessaloniki, Greece
| | - Petros Koidis
- Department of Prosthodontics, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki (A.U.Th), GR-54124, Thessaloniki, Greece
| | - Werner Geurtsen
- Department of Conservative Dentistry, Periodontology and Preventive Dentistry, Hannover Medical School (MHH), Hannover, Germany
| |
Collapse
|
171
|
Scolding NJ, Pasquini M, Reingold SC, Cohen JA. Cell-based therapeutic strategies for multiple sclerosis. Brain 2017; 140:2776-2796. [PMID: 29053779 PMCID: PMC5841198 DOI: 10.1093/brain/awx154] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 04/03/2017] [Accepted: 05/06/2017] [Indexed: 12/23/2022] Open
Abstract
The availability of multiple disease-modifying medications with regulatory approval to treat multiple sclerosis illustrates the substantial progress made in therapy of the disease. However, all are only partially effective in preventing inflammatory tissue damage in the central nervous system and none directly promotes repair. Cell-based therapies, including immunoablation followed by autologous haematopoietic stem cell transplantation, mesenchymal and related stem cell transplantation, pharmacologic manipulation of endogenous stem cells to enhance their reparative capabilities, and transplantation of oligodendrocyte progenitor cells, have generated substantial interest as novel therapeutic strategies for immune modulation, neuroprotection, or repair of the damaged central nervous system in multiple sclerosis. Each approach has potential advantages but also safety concerns and unresolved questions. Moreover, clinical trials of cell-based therapies present several unique methodological and ethical issues. We summarize here the status of cell-based therapies to treat multiple sclerosis and make consensus recommendations for future research and clinical trials.
Collapse
Affiliation(s)
- Neil J Scolding
- Department of Neurology, University of Bristol Southmead Hospital, Bristol BS10 5NB, UK
| | - Marcelo Pasquini
- Center for International Blood and Marrow Transplant Research (CIBMTR), Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Stephen C Reingold
- Scientific and Clinical Research Associates, LLC, Salisbury, CT 06068, USA
| | - Jeffrey A Cohen
- Neurological Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
172
|
Lin Y, Gil CH, Yoder MC. Differentiation, Evaluation, and Application of Human Induced Pluripotent Stem Cell-Derived Endothelial Cells. Arterioscler Thromb Vasc Biol 2017; 37:2014-2025. [PMID: 29025705 DOI: 10.1161/atvbaha.117.309962] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 09/26/2017] [Indexed: 12/13/2022]
Abstract
The emergence of induced pluripotent stem cell (iPSC) technology paves the way to generate large numbers of patient-specific endothelial cells (ECs) that can be potentially delivered for regenerative medicine in patients with cardiovascular disease. In the last decade, numerous protocols that differentiate EC from iPSC have been developed by many groups. In this review, we will discuss several common strategies that have been optimized for human iPSC-EC differentiation and subsequent studies that have evaluated the potential of human iPSC-EC as a cell therapy or as a tool in disease modeling. In addition, we will emphasize the importance of using in vivo vessel-forming ability and in vitro clonogenic colony-forming potential as a gold standard with which to evaluate the quality of human iPSC-EC derived from various protocols.
Collapse
Affiliation(s)
- Yang Lin
- From the Department of Pediatrics, Herman B. Wells Center for Pediatric Research (Y.L., C.-H.G., M.C.Y.) and Department of Biochemistry and Molecular Biology (Y.L., M.C.Y.), Indiana University School of Medicine, Indianapolis
| | - Chang-Hyun Gil
- From the Department of Pediatrics, Herman B. Wells Center for Pediatric Research (Y.L., C.-H.G., M.C.Y.) and Department of Biochemistry and Molecular Biology (Y.L., M.C.Y.), Indiana University School of Medicine, Indianapolis
| | - Mervin C Yoder
- From the Department of Pediatrics, Herman B. Wells Center for Pediatric Research (Y.L., C.-H.G., M.C.Y.) and Department of Biochemistry and Molecular Biology (Y.L., M.C.Y.), Indiana University School of Medicine, Indianapolis.
| |
Collapse
|
173
|
Yin L, Huang D, Liu X, Wang Y, Liu J, Liu F, Yu B. Omentin-1 effects on mesenchymal stem cells: proliferation, apoptosis, and angiogenesis in vitro. Stem Cell Res Ther 2017; 8:224. [PMID: 29017592 PMCID: PMC5633887 DOI: 10.1186/s13287-017-0676-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 07/01/2017] [Accepted: 09/18/2017] [Indexed: 02/07/2023] Open
Abstract
Background Mesenchymal stem cells (MSCs) are emerging as an extremely promising therapeutic agent for tissue repair. However, limitations exist such as the low numbers of MSCs obtained from donors, and the poor survival and function of donor cells. Omentin-1, a new fat depot-specific secretory adipokine, exerts proproliferation, prosurvival, and proangiogenic functions in certain cells via an Akt-dependent mechanism; however, little is known about the influence of omentin-1 on MSCs. Methods MSCs were isolated from 60–80 g donor rats. Cell proliferation was assessed with CCK-8 and EdU assay. Cell cycle, apoptosis ratio, reactive oxygen species concentration, and mitochondrial membrane potential were detected by flow cytometry. Hoechst 33342 dye was used to assess morphological changes of apoptosis. Expression levels of Akt, FoxO3a, GSK-3β, and apoptosis- and cell cycle-associated proteins were detected by Western blotting. Tube formation assay was used to test the angiogenesis role of conditioned medium from MSCs in vitro. The cytokine secretion was assessed by ELISA. Results After treatment with omentin-1 (100–800 ng/ml), MSCs displayed a higher proliferative capacity with an increasing number of cells in the S and G2 phase of the cell cycle. Moreover, omentin-1 preconditioning for 1 h could protect MSCs against H2O2-induced apoptosis in a concentration-dependent manner. Furthermore, omentin-1 pretreatment reduced the excessive reactive oxygen species. Western blots revealed that increased Bcl-2 and decreased Bax appeared in MSCs after omentin-1 incubation, which inhibited the mitochondrial apoptosis pathways with evidence showing inhibition of caspase-3 cleavage and preservation of mitochondrial membrane potential. Omentin-1 could enhance angiogenic growth factor secretion and elevate the ability of MSCs to stimulate tube formation by human umbilical vein endothelial cells (HUVECs). Furthermore, omentin-1 enhanced Akt phosphorylation; however, blockade of the PI3K/Akt pathway with an inhibitor, LY294002 (20 μM), suppressed the above beneficial effects of omentin-1. Conclusion Omentin-1 can exert beneficial effects on MSCs by promoting proliferation, inhibiting apoptosis, increasing secretion of angiogenic cytokines, and enhancing the ability for stimulating tube formation by HUVECs via the PI3K/Akt signaling pathway. Thus, omentin-1 may be considered a candidate for optimizing MSC-based cell therapy.
Collapse
Affiliation(s)
- Li Yin
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China
| | - Dan Huang
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China
| | - Xinxin Liu
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China
| | - Yongshun Wang
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China
| | - Jingjin Liu
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China
| | - Fang Liu
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China
| | - Bo Yu
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China. .,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China.
| |
Collapse
|
174
|
Clavreul A, Pourbaghi-Masouleh M, Roger E, Lautram N, Montero-Menei CN, Menei P. Human mesenchymal stromal cells as cellular drug-delivery vectors for glioblastoma therapy: a good deal? JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2017; 36:135. [PMID: 28962658 PMCID: PMC5622550 DOI: 10.1186/s13046-017-0605-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 09/21/2017] [Indexed: 02/06/2023]
Abstract
Background Glioblastoma (GB) is the most malignant brain tumor in adults. It is characterized by angiogenesis and a high proliferative and invasive capacity. Standard therapy (surgery, radiotherapy and chemotherapy with temozolomide) is of limited efficacy. Innovative anticancer drugs targeting both tumor cells and angiogenesis are urgently required, together with effective systems for their delivery to the brain. We assessed the ability of human mesenchymal stromal cells (MSCs) to uptake the multikinase inhibitor, sorafenib (SFN), and to carry this drug to a brain tumor following intranasal administration. Method MSCs were primed with SFN and drug content and release were quantified by analytical chemistry techniques. The ability of SFN-primed MSCs to inhibit the survival of the human U87MG GB cell line and endothelial cells was assessed in in vitro assays. These cells were then administered intranasally to nude mice bearing intracerebral U87MG xenografts. Their effect on tumor growth and angiogenesis was evaluated by magnetic resonance imaging and immunofluorescence analyses, and was compared with the intranasal administration of unprimed MSCs or SFN alone. Results MSCs took up about 9 pg SFN per cell, with no effect on viability, and were able to release 60% of the primed drug. The cytostatic activity of the released SFN was entirely conserved, resulting in a significant inhibition of U87MG and endothelial cell survival in vitro. Two intranasal administrations of SFN-primed MSCs in U87MG-bearing mice resulted in lower levels of tumor angiogenesis than the injection of unprimed MSCs or SFN alone, but had no effect on tumor volume. We also observed an increase in the proportion of small intratumoral vessels in animals treated with unprimed MSCs; this effect being abolished if the MSCs were primed with SFN. Conclusion We show the potential of MSCs to carry SFN to brain tumors following an intranasal administration. However, the therapeutic effect is modest probably due to the pro-tumorigenic properties of MSCs, which may limit the action of the released SFN. This calls into question the suitability of MSCs for use in GB therapy and renders it necessary to find methods guaranteeing the safety of this cellular vector after drug delivery.
Collapse
Affiliation(s)
- Anne Clavreul
- Département de Neurochirurgie, CHU, Angers, France. .,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.
| | - Milad Pourbaghi-Masouleh
- CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France.,Division of Drug Delivery and Tissue Engineering, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Emilie Roger
- MINT, UNIV Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire, Angers, France
| | - Nolwenn Lautram
- MINT, UNIV Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire, Angers, France
| | | | - Philippe Menei
- Département de Neurochirurgie, CHU, Angers, France.,CRCINA, INSERM, Université de Nantes, Université d'Angers, Angers, France
| |
Collapse
|
175
|
Seo HG, Yi Y, Oh BM, Paik NJ. Neuroprotective effect of secreted factors from human adipose stem cells in a rat stroke model. Neurol Res 2017; 39:1114-1124. [DOI: 10.1080/01616412.2017.1379293] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Han Gil Seo
- Department of Rehabilitation Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Youbin Yi
- Department of Rehabilitation Medicine, Nowon Eulji Medical Center, Eulji University School of Medicine, Seoul, Republic of Korea
| | - Byung-Mo Oh
- Department of Rehabilitation Medicine, Seoul National University College of Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | - Nam-Jong Paik
- Department of Rehabilitation Medicine, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| |
Collapse
|
176
|
Dreyer CH, Kjaergaard K, Ditzel N, Jørgensen NR, Overgaard S, Ding M. Optimizing combination of vascular endothelial growth factor and mesenchymal stem cells on ectopic bone formation in SCID mice. J Biomed Mater Res A 2017; 105:3326-3332. [PMID: 28879669 DOI: 10.1002/jbm.a.36195] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Accepted: 08/24/2017] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Insufficient blood supply may limit bone regeneration in bone defects. Vascular endothelial growth factor (VEGF) promotes angiogenesis by increasing endothelial migration. This outcome, however, could depend on time of application. Sheep mesenchymal stem cells (MSCs) in severe combined immunodeficient (SCID) mice were used in this study to evaluate optimal time points for VEGF stimulation to increase bone formation. METHODS Twenty-eight SCID (NOD.CB17-Prkdcscid /J) mice had hydroxyapatite granules seeded with 5 × 105 MSCs inserted subcutaneous. Pellets released VEGF on days 1-7, days 1-14, days 1-21, days 1-42, days 7-14, and days 21-42. After 8 weeks, the implant-bone-blocks were harvested, paraffin embedded, sectioned, and stained with both hematoxylin and eosin (HE) and immunohistochemistry for human vimentin (hVim) staining. Blood samples were collected for determination of bone-related biomarkers in serum. RESULTS The groups with 5 × 105 MSCs and VEGF stimulation on days 1-14 and days 1-21 showed more bone formation when compared to the control group of 5 × 105 MSCs alone (p < 0.01). Serum biomarkers had no significant values. The hVim staining confirmed the ovine origin of the observed ectopic bone formation. CONCLUSION Optimal bone formation of MSCs was reached when stimulating with VEGF during the first 14 or 21 days after surgery. © 2017 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 3326-3332, 2017.
Collapse
Affiliation(s)
- Chris H Dreyer
- Orthopaedic Research Laboratory, Department of Orthopaedic Surgery and Traumatology, Odense University Hospital, Department of Clinical Research, University of Southern Denmark, Sdr. Boulevard 29, Odense C, DK-5000, Denmark
| | - Kristian Kjaergaard
- Orthopaedic Research Laboratory, Department of Orthopaedic Surgery and Traumatology, Odense University Hospital, Department of Clinical Research, University of Southern Denmark, Sdr. Boulevard 29, Odense C, DK-5000, Denmark
| | - Nicholas Ditzel
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital, University of Southern Denmark, J. B. Winsløws Vej 25.1, Odense C, DK-5000, Denmark
| | - Niklas R Jørgensen
- Research Centre for Aging and Osteoporosis, Department of Clinical Biochemistry, Rigshospitalet, Ndr Ringvej 57-59, Glostrup, DK-2600, Denmark.,OPEN, Odense Patient data Explorative Network, Odense University Hospital/Institute of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Søren Overgaard
- Orthopaedic Research Laboratory, Department of Orthopaedic Surgery and Traumatology, Odense University Hospital, Department of Clinical Research, University of Southern Denmark, Sdr. Boulevard 29, Odense C, DK-5000, Denmark
| | - Ming Ding
- Orthopaedic Research Laboratory, Department of Orthopaedic Surgery and Traumatology, Odense University Hospital, Department of Clinical Research, University of Southern Denmark, Sdr. Boulevard 29, Odense C, DK-5000, Denmark
| |
Collapse
|
177
|
Abstract
INTRODUCTION In specific forms of congenital heart defects and pulmonary hypertension, the right ventricle (RV) is exposed to systemic levels of pressure overload. The RV is prone to failure in these patients because of its vulnerability to chronic pressure overload. As patients with a systemic RV reach adulthood, an emerging epidemic of RV failure has become evident. Medical therapies proven for LV failure are ineffective in treating RV failure. Areas covered: In this review, the pathophysiology of the failing RV under pressure overload is discussed, with specific emphasis on the pivotal roles of angiogenesis and oxidative stress. Studies investigating the ability of stem cell therapy to improve angiogenesis and mitigate oxidative stress in the setting of pressure overload are then reviewed. Finally, clinical trials utilizing stem cell therapy to prevent RV failure under pressure overload in congenital heart disease will be discussed. Expert commentary: Although considerable hurdles remain before their mainstream clinical implementation, stem cell therapy possesses revolutionary potential in the treatment of patients with failing systemic RVs who currently have very limited long-term treatment options. Rigorous clinical trials of stem cell therapy for RV failure that target well-defined mechanisms will ensure success adoption of this therapeutic strategy.
Collapse
Affiliation(s)
- Ming-Sing Si
- a Department of Cardiac Surgery, Section of Pediatric Cardiovascular Surgery , University of Michigan Medical School , Ann Arbor , MI , USA
| | - Richard G Ohye
- a Department of Cardiac Surgery, Section of Pediatric Cardiovascular Surgery , University of Michigan Medical School , Ann Arbor , MI , USA
| |
Collapse
|
178
|
Pence JC, Clancy KBH, Harley BAC. Proangiogenic Activity of Endometrial Epithelial and Stromal Cells in Response to Estradiol in Gelatin Hydrogels. ACTA ACUST UNITED AC 2017; 1. [PMID: 29230433 DOI: 10.1002/adbi.201700056] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Biomaterial vascularization remains a major focus in the field of tissue engineering. Biomaterial culture of endometrial cells is described as a platform to inform the design of proangiogenic biomaterials. The endometrium undergoes rapid growth and shedding of dense vascular networks during each menstrual cycle mediated via estradiol and progesterone in vivo. Cocultures of endometrial epithelial and stromal cells encapsulated within a methacrylamide-functionalized gelatin hydrogel are employed. It is reported that proangiogenic gene expression profiles and vascular endothelial growth factor production are hormone dependent in endometrial epithelial cells, but that hormone signals have no effect on human telomerase reverse transcriptase (hTERT)-immortalized endometrial stromal cells. This study subsequently examines whether the magnitude of epithelial cell response is sufficient to induce changes in human umbilical vein endothelial cell network formation. Incorporation of endometrial stromal cells improves vessel formation, but co-culture with endometrial epithelial cells leads to a decrease in vascular formation, suggesting the need for stratified cocultures of endometrial epithelial and stromal cells with endothelial cells. Given the transience of hormonal signals within 3D biomaterials, the inclusion of sex hormone binding globulin (SHBG) to alter the bioavailability of estradiol within the hydrogel is reported, demonstrating a strategy to reduce diffusive losses via SHBG-mediated estradiol sequestration.
Collapse
Affiliation(s)
- Jacquelyn C Pence
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, 110 Roger Adams Laboratory, 600 S. Mathews St, Urbana, IL 61801, USA
| | - Kathryn B H Clancy
- Department of Anthropology, University of Illinois at Urbana-Champaign, 607 S. Mathews St, Urbana IL 61801, USA
| | - Brendan A C Harley
- Department of Chemical and Biomolecular Engineering, University of Illinois at Urbana-Champaign, 110 Roger Adams Laboratory, 600 S. Mathews St, Urbana, IL 61801, USA
| |
Collapse
|
179
|
Wang L, Qing L, Liu H, Liu N, Qiao J, Cui C, He T, Zhao R, Liu F, Yan F, Wang C, Liang K, Guo X, Shen YH, Hou X, Chen L. Mesenchymal stromal cells ameliorate oxidative stress-induced islet endothelium apoptosis and functional impairment via Wnt4-β-catenin signaling. Stem Cell Res Ther 2017; 8:188. [PMID: 28807051 PMCID: PMC5557510 DOI: 10.1186/s13287-017-0640-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 07/09/2017] [Accepted: 07/24/2017] [Indexed: 12/26/2022] Open
Abstract
Background Islet dysfunction and destruction are the common cause for both type 1 and type 2 diabetes mellitus (T2DM). The islets of Langerhans are highly vascularized miniorgans, and preserving the structural integrity and full function of the microvascular endothelium is vital for protecting the islets from the infiltration of immune cells and secondary inflammatory attack. Mesenchymal stromal cell (MSC)-based therapies have been proven to promote angiogenesis of the islets; however, the underlying mechanism for the protective role of MSCs in the islet endothelium is still vague. Methods In this study, we used MS-1, a murine islet microvascular endothelium cell line, and an MSC-MS1 transwell culturing system to investigate the protective mechanism of rat bone marrow-derived MSCs under oxidative stress in vitro. Cell apoptosis was detected by TUNEL staining, annexin V/PI flow cytometry analysis, and cleaved caspase 3 western blotting analysis. Endothelial cell activation was determined by expression of intercellular cell adhesion molecule (ICAM) and vascular cell adhesion molecule (VCAM), as well as eNOS phosphorylation/activation. The changes of VCAM-1, eNOS, and the β-catenin expression were also tested in the isolated islets of T2DM rats infused with MSCs. Results We observed that treating MS-1 cells with H2O2 triggered significant apoptosis, induction of VCAM expression, and reduction of eNOS phosphorylation. Importantly, coculturing MS-1 cells with MSCs prevented oxidative stress-induced apoptosis, eNOS inhibition, and VCAM elevation in MS-1 cells. Similar changes in VCAM-1 and eNOS phosphorylation could also be observed in the islets isolated from T2DM rats infused with MSCs. Moreover, MSCs cocultured with MS-1 in vitro or their administration in vivo could both result in an increase of β-catenin, which suggested activation of the β-catenin-dependent Wnt signaling pathway. In MS-1 cells, activation of the β-catenin-dependent Wnt signaling pathway partially mediated the protective effects of MSCs against H2O2-induced apoptosis and eNOS inhibition. Furthermore, MSCs produced a significant amount of Wnt4 and Wnt5a. Although both Wnt4 and Wnt5a participated in the interaction between MSCs and MS-1 cells, Wnt4 exhibited a protective role while Wnt5a seemed to show a destructive role in MS-1 cells. Conclusions Our observations provide evidence that the orchestration of the MSC-secreted Wnts could promote the survival and improve the endothelial function of the injured islet endothelium via activating the β-catenin-dependent Wnt signaling in target endothelial cells. This finding might inspire further in-vivo studies.
Collapse
Affiliation(s)
- Lingshu Wang
- Department of Endocrinology, Institute of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Li Qing
- Department of Endocrinology, Institute of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - He Liu
- Department of Endocrinology, Institute of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Na Liu
- College of Public Health, Shandong University, Jinan, Shandong, 250012, China
| | - Jingting Qiao
- Department of Endocrinology, Institute of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Chen Cui
- Department of Endocrinology, Institute of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Tianyi He
- Department of Endocrinology, Institute of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Ruxing Zhao
- Department of Endocrinology, Institute of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Fuqiang Liu
- Department of Endocrinology, Institute of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Fei Yan
- Department of Endocrinology, Institute of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Chuan Wang
- Department of Endocrinology, Institute of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Kai Liang
- Department of Endocrinology, Institute of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Xinghong Guo
- Department of Endocrinology, Institute of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China
| | - Ying H Shen
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA.,Texas Heart Institute, Houston, TX, USA
| | - Xinguo Hou
- Department of Endocrinology, Institute of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
| | - Li Chen
- Department of Endocrinology, Institute of Endocrinology and Metabolism, Qilu Hospital of Shandong University, Jinan, 250012, Shandong, China.
| |
Collapse
|
180
|
Different Angiogenic Potentials of Mesenchymal Stem Cells Derived from Umbilical Artery, Umbilical Vein, and Wharton's Jelly. Stem Cells Int 2017; 2017:3175748. [PMID: 28874910 PMCID: PMC5569878 DOI: 10.1155/2017/3175748] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 02/14/2017] [Accepted: 02/21/2017] [Indexed: 01/02/2023] Open
Abstract
Human mesenchymal stem cells derived from the umbilical cord (UC) are a favorable source for allogeneic cell therapy. Here, we successfully isolated the stem cells derived from three different compartments of the human UC, including perivascular stem cells derived from umbilical arteries (UCA-PSCs), perivascular stem cells derived from umbilical vein (UCV-PSCs), and mesenchymal stem cells derived from Wharton's jelly (WJ-MSCs). These cells had the similar phenotype and differentiation potential toward adipocytes, osteoblasts, and neuron-like cells. However, UCA-PSCs and UCV-PSCs had more CD146+ cells than WJ-MSCs (P < 0.05). Tube formation assay in vitro showed the largest number of tube-like structures and branch points in UCA-PSCs among the three stem cells. Additionally, the total tube length in UCA-PSCs and UCV-PSCs was significantly longer than in WJ-MSCs (P < 0.01). Microarray, qRT-PCR, and Western blot analysis showed that UCA-PSCs had the highest expression of the Notch ligand Jagged1 (JAG1), which is crucial for blood vessel maturation. Knockdown of Jagged1 significantly impaired the angiogenesis in UCA-PSCs. In summary, UCA-PSCs are promising cell populations for clinical use in ischemic diseases.
Collapse
|
181
|
Liang X, Zhang L, Wang S, Han Q, Zhao RC. Exosomes secreted by mesenchymal stem cells promote endothelial cell angiogenesis by transferring miR-125a. J Cell Sci 2017; 129:2182-9. [PMID: 27252357 DOI: 10.1242/jcs.170373] [Citation(s) in RCA: 402] [Impact Index Per Article: 57.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 04/05/2016] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis plays crucial roles in various physiological processes including wound healing and tissue repair. It requires a tight interaction between endothelial cells and their surrounding environment. Mesenchymal stem cells (MSCs), one of the non-endothelial cell types present in the perivascular environment, have been shown to secret exosomes to modulate intercellular communications between MSCs and their target cells. In this study, we initially isolated exosomes secreted by human adipose-derived MSCs (adMSC-Exo) and examined their roles in angiogenesis. We found that adMSC-Exo could be taken up by endothelial cells and significantly promote angiogenesis in vitro and in vivo Further study showed that miR-125a was enriched in adMSC-Exo, and repressed the expression of the angiogenic inhibitor delta-like 4 (DLL4) by targeting its 3' untranslated region. Additionally, adMSC-Exo and its exosomal transferred miR-125a could repress DLL4 expression and modulate endothelial cell angiogenesis through promoting formation of endothelial tip cells. In conclusion, our study indicates that adMSC-Exo can transfer miR-125a to endothelial cells and promote angiogenesis by repressing DLL4. adMSC-Exo, as a pro-angiogenic factor, might be a promising candidate for therapeutical tissue repair.
Collapse
Affiliation(s)
- Xiaolei Liang
- Department of Cell Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, 5# Dongdansantiao, Beijing 100005, People's Republic of China
| | - Lina Zhang
- Department of Cell Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, 5# Dongdansantiao, Beijing 100005, People's Republic of China
| | - Shihua Wang
- Department of Cell Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, 5# Dongdansantiao, Beijing 100005, People's Republic of China
| | - Qin Han
- Department of Cell Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, 5# Dongdansantiao, Beijing 100005, People's Republic of China
| | - Robert Chunhua Zhao
- Department of Cell Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, 5# Dongdansantiao, Beijing 100005, People's Republic of China Peking Union Medical College Hospital, 1# Shuaifuyuan, Beijing 100032, People's Republic of China
| |
Collapse
|
182
|
Lambrichts I, Driesen RB, Dillen Y, Gervois P, Ratajczak J, Vangansewinkel T, Wolfs E, Bronckaers A, Hilkens P. Dental Pulp Stem Cells: Their Potential in Reinnervation and Angiogenesis by Using Scaffolds. J Endod 2017; 43:S12-S16. [PMID: 28781091 DOI: 10.1016/j.joen.2017.06.001] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Dental pulp is a highly vascularized and innervated tissue containing a heterogeneous stem cell population with multilineage differentiation potential. Current endodontic treatments focus on the preservation of the pulp tissue and the regeneration of dental pulp after pathological insults. Human dental pulp stem cells (hDPSCs) are currently investigated as stem cell-based therapy for pulp regeneration and for peripheral nerve injury in which neurons and Schwann cells display limited regenerative capacity. We have developed a neuronal differentiation protocol for hDPSCs that requires neurosphere formation before neuronal maturation. Moreover, Schwann cell differentiation of hDPSCs in our group revealed that differentiated hDPSCs have acquired the ability to myelinate and guide neurites from dorsal root ganglia. Besides their dynamic differentiation capacity, hDPSCs were shown to exert a paracrine effect on neural and endothelial cells. Analysis of hDPSC conditioned medium revealed the secretion of a broad spectrum of growth factors including brain-derived neurotrophic factor, nerve growth factor, vascular endothelial growth factor, and glial-derived neurotrophic factor. Application of the conditioned medium to endothelial cells promoted cell migration and tubulogenesis, indicating a paracrine proangiogenic effect. This hypothesis was enforced by the enhanced formation of blood vessels in the chorioallantoic membrane assay in the presence of hDPSCs. In addition, transplantation of 3-dimensional-printed hydroxyapatite scaffolds containing peptide hydrogels and hDPSCs into immunocompromised mice revealed blood vessel ingrowth, pulplike tissue formation, and osteodentin deposition suggesting osteogenic/odontogenic differentiation of hDPSCs. Future studies in our research group will focus on the pulp regeneration capacity of hDPSCs and the role of fibroblasts within the pulp extracellular matrix.
Collapse
Affiliation(s)
- Ivo Lambrichts
- Laboratory of Morphology, Biomedical Research Institute (BIOMED), Hasselt University, Diepenbeek, Belgium.
| | - Ronald B Driesen
- Laboratory of Morphology, Biomedical Research Institute (BIOMED), Hasselt University, Diepenbeek, Belgium
| | - Yörg Dillen
- Laboratory of Morphology, Biomedical Research Institute (BIOMED), Hasselt University, Diepenbeek, Belgium
| | - Pascal Gervois
- Laboratory of Morphology, Biomedical Research Institute (BIOMED), Hasselt University, Diepenbeek, Belgium
| | - Jessica Ratajczak
- Laboratory of Morphology, Biomedical Research Institute (BIOMED), Hasselt University, Diepenbeek, Belgium
| | - Tim Vangansewinkel
- Laboratory of Morphology, Biomedical Research Institute (BIOMED), Hasselt University, Diepenbeek, Belgium
| | - Esther Wolfs
- Laboratory of Morphology, Biomedical Research Institute (BIOMED), Hasselt University, Diepenbeek, Belgium
| | - Annelies Bronckaers
- Laboratory of Morphology, Biomedical Research Institute (BIOMED), Hasselt University, Diepenbeek, Belgium
| | - Petra Hilkens
- Laboratory of Morphology, Biomedical Research Institute (BIOMED), Hasselt University, Diepenbeek, Belgium
| |
Collapse
|
183
|
Gonzalez-King H, García NA, Ontoria-Oviedo I, Ciria M, Montero JA, Sepúlveda P. Hypoxia Inducible Factor-1α Potentiates Jagged 1-Mediated Angiogenesis by Mesenchymal Stem Cell-Derived Exosomes. Stem Cells 2017; 35:1747-1759. [PMID: 28376567 DOI: 10.1002/stem.2618] [Citation(s) in RCA: 272] [Impact Index Per Article: 38.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 03/05/2017] [Accepted: 03/10/2017] [Indexed: 12/13/2022]
Abstract
Insufficient vessel growth associated with ischemia remains an unresolved issue in vascular medicine. Mesenchymal stem cells (MSCs) have been shown to promote angiogenesis via a mechanism that is potentiated by hypoxia. Overexpression of hypoxia inducible factor (HIF)-1α in MSCs improves their therapeutic potential by inducing angiogenesis in transplanted tissues. Here, we studied the contribution of exosomes released by HIF-1α-overexpressing donor MSCs (HIF-MSC) to angiogenesis by endothelial cells. Exosome secretion was enhanced in HIF-MSC. Omics analysis of miRNAs and proteins incorporated into exosomes pointed to the Notch pathway as a candidate mediator of exosome communication. Interestingly, we found that Jagged1 was the sole Notch ligand packaged into MSC exosomes and was more abundant in HIF-MSC than in MSC controls. The addition of Jagged1-containing exosomes from MSC and HIF-MSC cultures to endothelial cells triggered transcriptional changes in Notch target genes and induced angiogenesis in an in vitro model of capillary-like tube formation, and both processes were stimulated by HIF-1α. Finally, subcutaneous injection of Jagged 1-containing exosomes from MSC and HIF-MSC cultures in the Matrigel plug assay induced angiogenesis in vivo, which was more robust when they were derived from HIF-MSC cultures. All Jagged1-mediated effects could be blocked by prior incubation of exosomes with an anti-Jagged 1 antibody. All together, the results indicate that exosomes derived from MSCs stably overexpressing HIF-1α have an increased angiogenic capacity in part via an increase in the packaging of Jagged1, which could have potential applications for the treatment of ischemia-related disease. Stem Cells 2017;35:1747-1759.
Collapse
Affiliation(s)
- Hernán Gonzalez-King
- Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- Joint Unit for cardiovascular Repair Instituto de Investigación Sanitaria La Fe-Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Nahuel A García
- Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- Joint Unit for cardiovascular Repair Instituto de Investigación Sanitaria La Fe-Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Imelda Ontoria-Oviedo
- Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- Joint Unit for cardiovascular Repair Instituto de Investigación Sanitaria La Fe-Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - María Ciria
- Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- Joint Unit for cardiovascular Repair Instituto de Investigación Sanitaria La Fe-Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - José Anastasio Montero
- Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- Joint Unit for cardiovascular Repair Instituto de Investigación Sanitaria La Fe-Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Pilar Sepúlveda
- Regenerative Medicine and Heart Transplantation Unit, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
- Joint Unit for cardiovascular Repair Instituto de Investigación Sanitaria La Fe-Centro de Investigación Príncipe Felipe, Valencia, Spain
| |
Collapse
|
184
|
Pignon B, Sevestre MA, Kanagaratnam L, Pernod G, Stephan D, Emmerich J, Clement C, Sarlon G, Boulon C, Tournois C, Nguyen P. Autologous Bone Marrow Mononuclear Cell Implantation and Its Impact on the Outcome of Patients With Critical Limb Ischemia - Results of a Randomized, Double-Blind, Placebo-Controlled Trial. Circ J 2017; 81:1713-1720. [PMID: 28603176 DOI: 10.1253/circj.cj-17-0045] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Cell therapy is a therapeutic option for patients presenting with nonrevascularizable critical limb ischemia (CLI). However there is a lack of firm evidence on its efficacy because of the paucity of randomized controlled trials.Methods and Results:The BALI trial was a multicenter, randomized, controlled, double-blind clinical trial that included 38 patients. For all of them, 500 mL of bone marrow were collected for preparation of a BM-MNC product that was implanted in patients assigned to active treatment. For the placebo group, a placebo cell-free product was implanted. Within 6 months after inclusion, major amputations had to be performed in 5 of the 19 placebo-treated patients and in 3 of the 17 BM-MNC-treated patients. According to a classical logistic regression analysis there was no significant difference. However, when using the jackknife analysis, 6 months after inclusion BM-MNC implantation was associated with a lower risk of major amputation (odds ratio (OR): 0.55; 95% confidence interval (CI): 0.52-0.58; P<0.0001) and of occurrence of any event (major or minor amputation, or revascularization) (OR: 0.30; 95% CI: 0.29-0.31; P<0.0001). The secondary endpoints (i.e., pain, ulcers, TcPO2, and ankle-brachial index value) were not statistically different between groups. CONCLUSIONS Our results suggested that cell therapy reduced the risk of major amputation in patients presenting with nonrevascularizable CLI.
Collapse
Affiliation(s)
| | | | | | - Gilles Pernod
- Department of Vascular Medicine, University Hospital
| | | | - Joseph Emmerich
- Department of Vascular Medicine and Cardiology, University Hospital Hotel Dieu
| | | | | | - Carine Boulon
- Department of Vascular and Internal Medicine, University Hospital
| | | | - Philippe Nguyen
- Research Unit HERVI EA, Medical School, Champagne-Ardenne University
| |
Collapse
|
185
|
Sueyama Y, Kaneko T, Ito T, Kaneko R, Okiji T. Implantation of Endothelial Cells with Mesenchymal Stem Cells Accelerates Dental Pulp Tissue Regeneration/Healing in Pulpotomized Rat Molars. J Endod 2017; 43:943-948. [DOI: 10.1016/j.joen.2017.01.035] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 01/10/2017] [Accepted: 01/25/2017] [Indexed: 12/17/2022]
|
186
|
Angiogenic Capacity of Dental Pulp Stem Cell Regulated by SDF-1 α-CXCR4 Axis. Stem Cells Int 2017; 2017:8085462. [PMID: 28588623 PMCID: PMC5447288 DOI: 10.1155/2017/8085462] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 02/26/2017] [Accepted: 03/01/2017] [Indexed: 01/07/2023] Open
Abstract
Previously, the perivascular characteristics of dental pulp stem cells (DPSCs) were reported, which suggested the potential application of DPSCs as perivascular cell source. In this study, we investigated whether DPSCs had angiogenic capacity by coinjection with human umbilical vein endothelial cells (HUVECs) in vivo; in addition, we determined the role of stromal cell-derived factor 1-α (SDF-1α) and C-X-C chemokine receptor type 4 (CXCR4) axis in the mutual interaction between DPSCs and HUVECs. Primarily isolated DPSCs showed mesenchymal stem cell- (MSC-) like characteristics. Moreover, DPSCs expressed perivascular markers such as NG2, α-smooth muscle actin (α-SMA), platelet-derived growth factor receptor β (PDGFRβ), and CD146. In vivo angiogenic capacity of DPSCs was demonstrated by in vivo Matrigel plug assay. We could observe microvessel-like structures in the coinjection of DPSCs and HUVECs at 7 days postinjection. To block SDF-1α and CXCR4 axis between DPSCs and HUVECs, AMD3100, a CXCR4 antagonist, was added into Matrigel plug. No significant microvessel-like structures were observed at 7 days postinjection. In conclusion, DPSCs have perivascular characteristics that contribute to in vivo angiogenesis. The findings of this study have potential applications in neovascularization of engineered tissues and vascular diseases.
Collapse
|
187
|
Zhang Y, Xia X, Yan J, Yan L, Lu C, Zhu X, Wang T, Yin T, Li R, Chang HM, Qiao J. Mesenchymal stem cell-derived angiogenin promotes primodial follicle survival and angiogenesis in transplanted human ovarian tissue. Reprod Biol Endocrinol 2017; 15:18. [PMID: 28274269 PMCID: PMC5343383 DOI: 10.1186/s12958-017-0235-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 02/23/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND We have recently reported that human bone marrow-derived mesenchymal stem cells (MSCs) facilitate angiogenesis and prevent follicle loss in xenografted human ovarian tissues. However, the mechanism underlying this effect remains to be elucidated. Thus, determining the paracrine profiles and identifying the key secreted factors in MSCs co-transplanted with ovarian grafts are essential for the future application of MSCs. METHODS In this study, we used cytokine microarrays to identify differentially expressed proteins associated with angiogenesis in frozen-thawed ovarian tissues co-transplanted with MSCs. The function of specific secreted factors in MSCs co-transplanted with human ovarian tissues was studied via targeted blockade with short-hairpin RNAi and the use of monoclonal neutralizing antibodies. RESULTS Our results showed that angiogenin (ANG) was one of the most robustly up-regulated proteins (among 42 protein we screened, 37 proteins were up-regulated). Notably, the targeted depletion of ANG with short-hairpin RNAi (shANG) or the addition of anti-ANG monoclonal neutralizing antibodies (ANG Ab) significantly reversed the MSC-stimulated angiogenesis, increased follicle numbers and protective effect on follicle apoptosis. CONCLUSION Our results indicate that ANG plays a critical role in regulating angiogenesis and follicle survival in xenografted human ovarian tissues. Our findings provide important insights into the molecular mechanism by which MSCs promote angiogenesis and follicle survival in transplanted ovarian tissues, thus providing a theoretical basis for their further application.
Collapse
MESH Headings
- Adult
- Animals
- Antibodies, Neutralizing/pharmacology
- Apoptosis/drug effects
- Cells, Cultured
- Female
- Graft Survival/drug effects
- Humans
- Mesenchymal Stem Cell Transplantation/methods
- Mesenchymal Stem Cells/metabolism
- Mice, SCID
- Neovascularization, Physiologic/drug effects
- Neovascularization, Physiologic/genetics
- Neovascularization, Physiologic/physiology
- Ovarian Follicle/blood supply
- Ovarian Follicle/growth & development
- Ovarian Follicle/transplantation
- Ovariectomy
- RNA Interference
- Ribonuclease, Pancreatic/genetics
- Ribonuclease, Pancreatic/immunology
- Ribonuclease, Pancreatic/metabolism
- Transplantation, Heterologous
Collapse
Affiliation(s)
- Yaoyao Zhang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, No.49 North HuaYuan Road, HaiDian District, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China
| | - Xi Xia
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, No.49 North HuaYuan Road, HaiDian District, Beijing, 100191, China
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Shenzhen Hospital, No.1120 Lotus Road, FuTian District, Shenzhen, Guangdong, 518000, China
| | - Jie Yan
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, No.49 North HuaYuan Road, HaiDian District, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China
| | - Liying Yan
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, No.49 North HuaYuan Road, HaiDian District, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China
| | - Cuilin Lu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, No.49 North HuaYuan Road, HaiDian District, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China
| | - Xiaohui Zhu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, No.49 North HuaYuan Road, HaiDian District, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China
| | - Tianren Wang
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, No.49 North HuaYuan Road, HaiDian District, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, 100191, China
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Shengjing Hospital of China Medical University, Shenyang, 100004, China
| | - Tailang Yin
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, No.49 North HuaYuan Road, HaiDian District, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China
| | - Rong Li
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, No.49 North HuaYuan Road, HaiDian District, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, 100191, China
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China
| | - Hsun-Ming Chang
- Department of Obstetrics and Gynaecology, Child and Family Research Institute, University of British Columbia, Vancouver, V5Z4H4, Canada
| | - Jie Qiao
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, Peking University Third Hospital, No.49 North HuaYuan Road, HaiDian District, Beijing, 100191, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproduction, Beijing, 100191, China.
- Key Laboratory of Assisted Reproduction, Ministry of Education, Beijing, 100191, China.
| |
Collapse
|
188
|
Wang X, Li G, Guo J, Yang L, Liu Y, Sun Q, Li R, Yu W. Hybrid composites of mesenchymal stem cell sheets, hydroxyapatite, and platelet-rich fibrin granules for bone regeneration in a rabbit calvarial critical-size defect model. Exp Ther Med 2017; 13:1891-1899. [PMID: 28565782 PMCID: PMC5443196 DOI: 10.3892/etm.2017.4199] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 01/13/2017] [Indexed: 12/15/2022] Open
Abstract
The reconstruction of large bone defects remains a major clinical challenge, and tissue engineering is a promising technique for resolving this problem. Many attempts have been made to optimize bone tissue engineering protocols. The aim of the present study was to develop a process incorporating mesenchymal stem cell (MSC) sheets with nanoscale hydroxyapatite (nano-HA) and autologous platelet-rich fibrin (PRF) granules for enhanced bone formation within a critical-sized rabbit cranial defect. MSC sheets and PRF were prepared prior to in vivo experiments. The osteogenic differentiation ability of MSCs and the ultrastructure of PRF were also studied. A total of 15 New Zealand white rabbits were used in the current study and critical-size defects (CSDs) were surgically introduced in the cranium (diameter, 15 mm). The surgical defects were treated with MSC/PRF composites, MSC composites or left empty. Animals were euthanized at week 8 post-surgery. Iconography, histological and histomorphometric analysis were performed to assess de novo bone formation. The percentage of new bone in the MSC/PRF group (35.7±5.1%) was significantly higher than that in the MSC (18.3±3.2%; P<0.05) and empty defect groups (4.7±1.5%; P<0.05). The results of the present study suggest that combined application of an MSC sheet with nano-HA and granular PRF enhances bone regeneration in a rabbit calvarial CSD model, and provides a novel insight into bone tissue regeneration for large bone defects.
Collapse
Affiliation(s)
- Xi Wang
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Guanghui Li
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Jia Guo
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Lei Yang
- Department of Orthodontics, Beijing Ruitai Dental Hospital, Beijing 100107, P.R. China
| | - Yiming Liu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Qiang Sun
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Rui Li
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Weiwei Yu
- Department of Stomatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
189
|
Wang Z, Wang L, Su X, Pu J, Jiang M, He B. Rational transplant timing and dose of mesenchymal stromal cells in patients with acute myocardial infarction: a meta-analysis of randomized controlled trials. Stem Cell Res Ther 2017; 8:21. [PMID: 28129790 PMCID: PMC5273801 DOI: 10.1186/s13287-016-0450-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 11/23/2016] [Accepted: 12/03/2016] [Indexed: 02/08/2023] Open
Abstract
Background Mesenchymal stromal cells (MSCs) are considered to have a modest benefit on left ventricular ejection fraction (LVEF) in patients with acute myocardial infarction (AMI). However, the optimal injection timing and dose needed to induce beneficial cardiac effects are unknown. The purpose of this meta-analysis was to identify an optimal MSC transplantation time and cell dose in the setting of AMI to achieve better clinical endpoints. Methods The authors conducted a systematic review of studies published up to June 2016 by searching PubMed, EMBASE, MEDLINE, and the Cochrane Library for relevant randomized controlled trials (RCTs). Results Eight prospective RCTs with 449 participants were included. The pooled results revealed that patients in the MSC group had no significant increase in LVEF from baseline compared with that in the control group (1.47% increase, 95% confidence interval (CI) −4.5 to 7.45; I2 = 97%; P > 0.05). A subgroup analysis was conducted to explore the results according to differences in transplantation time and dose of MSCs injected. For transplantation timing, the LVEF of patients accepting a MSC infusion within 1 week was significantly increased by 3.22% (95% CI 1.31 to 5.14; I2 = 0; P < 0.05), but this increase was insignificant in the group that accepted an MSC infusion after 1 week (−0.35% in LVEF, 95% CI −10.22 to 9.52; I2 = 99%; P > 0.05). Furthermore, patients accepting a MSC dose of less than 107 cells exhibited an LVEF improvement of 2.25% compared with the control (95% CI 0.56 to 3.93; I2 = 9%; P < 0.05). Combining transplantation time and cell dose indicates that a significant improvement of LVEF of 3.32% was achieved in the group of patients injected with <107 MSCs within 1 week (95% CI 1.14 to 5.50; I2 = 0; P = 0.003). Conclusions Transplantation time and injected cell dose are key factors that determine the therapeutic effect of stem cell therapy. The injection of no more than 107 MSCs within 1 week for AMI after percutaneous coronary intervention might improve left ventricular systolic function. Further studies on the mechanism and the effectiveness of MSCs for long-term therapy are warranted.
Collapse
Affiliation(s)
- Zi Wang
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 160 Pujian Road, Shanghai, 200127, China
| | - Lingling Wang
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 160 Pujian Road, Shanghai, 200127, China
| | - Xuan Su
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 160 Pujian Road, Shanghai, 200127, China
| | - Jun Pu
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 160 Pujian Road, Shanghai, 200127, China
| | - Meng Jiang
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 160 Pujian Road, Shanghai, 200127, China.
| | - Ben He
- Department of Cardiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 160 Pujian Road, Shanghai, 200127, China.
| |
Collapse
|
190
|
Hayrapetyan A, Surjandi S, Lemsom EEPJ, Wolters MMMW, Jansen JA, van den Beucken JJJP. Coculture effects on the osteogenic differentiation of human mesenchymal stromal cells. Tissue Eng Regen Med 2016; 13:713-723. [PMID: 30603452 DOI: 10.1007/s13770-016-0008-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 03/04/2016] [Accepted: 03/10/2016] [Indexed: 12/16/2022] Open
Abstract
Cell-based bone regeneration is generally pursued based on single cell type approaches, for which human adipose tissue-derived mesenchymal stromal cells (AT-MSCs) are frequently used, owing to their easy accessibility and relatively large yield. In view of multiple cell types involved in physiological bone regeneration, this study aimed to evaluate the osteogenic differentiation of AT-MSCs upon co-culture with endothelial cells or macrophages in a direct or indirect in vitro co-culture set-up. Our hypotheses were that 1) endothelial cells and macrophages stimulate AT-MSCs proliferation and osteogenic differentiation and that 2) these two cell types will more profoundly affect osteogenic differentiation of AT-MSCs in a direct compared to an indirect co-culture set-up, because of the possibility for both cell-cell interactions and effects of secreted soluble factors in the former. Osteogenic differentiation of AT-MSCs was stimulated by endothelial cells, particularly in direct co-cultures. Although initial numbers of AT-MSCs in co-culture with endothelial cells were 50% compared to monoculture controls, equal levels of mineralization were achieved. Macrophages showed a variable effect on AT-MSCs behavior for indirect co-cultures and a negative effect on osteogenic differentiation of AT-MSCs in direct co-cultures, the latter likely due to species differences of the cell types used. The results of this study demonstrate potential for cell combination strategies in bone regenerative therapies.
Collapse
Affiliation(s)
| | - Soraya Surjandi
- 1Department of Biomaterials, Radboudumc, Nijmegen, the Netherlands
| | | | | | - John A Jansen
- 1Department of Biomaterials, Radboudumc, Nijmegen, the Netherlands
| | - Jeroen J J P van den Beucken
- 1Department of Biomaterials, Radboudumc, Nijmegen, the Netherlands.,2Department of Biomaterials (309), Radboudumc, Ph van Leijdenlaan 25, Nijmegen, 6525 the Netherlands
| |
Collapse
|
191
|
Supportive angiogenic and osteogenic differentiation of mesenchymal stromal cells and endothelial cells in monolayer and co-cultures. Int J Oral Sci 2016; 8:223-230. [PMID: 27910940 PMCID: PMC5168417 DOI: 10.1038/ijos.2016.39] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2016] [Indexed: 12/14/2022] Open
Abstract
Sites of implantation with compromised biology may be unable to achieve the required level of angiogenic and osteogenic regeneration. The specific function and contribution of different cell types to the formation of prevascularized, osteogenic networks in co-culture remains unclear. To determine how bone marrow-derived mesenchymal stromal cells (BMSCs) and endothelial cells (ECs) contribute to cellular proangiogenic differentiation, we analysed the differentiation of BMSCs and ECs in standardized monolayer, Transwell and co-cultures. BMSCs were derived from the iliac bone marrow of five patients, characterized and differentiated in standardized monolayers, permeable Transwells and co-cultures with human umbilical vein ECs (HUVECs). The expression levels of CD31, von Willebrand factor, osteonectin (ON) and Runx2 were assessed by quantitative reverse transcriptase polymerase chain reaction. The protein expression of alkaline phosphatase, ON and CD31 was demonstrated via histochemical and immunofluorescence analysis. The results showed that BMSCs and HUVECs were able to retain their lineage-specific osteogenic and angiogenic differentiation in direct and indirect co-cultures. In addition, BMSCs demonstrated a supportive expression of angiogenic function in co-culture, while HUVEC was able to improve the expression of osteogenic marker molecules in BMSCs.
Collapse
|
192
|
Du WJ, Chi Y, Yang ZX, Li ZJ, Cui JJ, Song BQ, Li X, Yang SG, Han ZB, Han ZC. Heterogeneity of proangiogenic features in mesenchymal stem cells derived from bone marrow, adipose tissue, umbilical cord, and placenta. Stem Cell Res Ther 2016; 7:163. [PMID: 27832825 PMCID: PMC5103372 DOI: 10.1186/s13287-016-0418-9] [Citation(s) in RCA: 133] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 09/06/2016] [Accepted: 10/04/2016] [Indexed: 01/08/2023] Open
Abstract
Background Mesenchymal stem cells (MSCs) have been widely proven effective for therapeutic angiogenesis in ischemia animal models as well as clinical vascular diseases. Because of the invasive method, limited resources, and aging problems of adult tissue-derived MSCs, more perinatal tissue-derived MSCs have been isolated and studied as promising substitutable MSCs for cell transplantation. However, fewer studies have comparatively studied the angiogenic efficacy of MSCs derived from different tissues sources. Here, we evaluated whether the in-situ environment would affect the angiogenic potential of MSCs. Methods We harvested MSCs from adult bone marrow (BMSCs), adipose tissue (AMSCs), perinatal umbilical cord (UMSCs), and placental chorionic villi (PMSCs), and studied their “MSC identity” by flow cytometry and in-vitro trilineage differentiation assay. Then we comparatively studied their endothelial differentiation capabilities and paracrine actions side by side in vitro. Results Our data showed that UMSCs and PMSCs fitted well with the minimum standard of MSCs as well as BMSCs and AMSCs. Interestingly, we found that MSCs regardless of their tissue origins could develop similar endothelial-relevant functions in vitro, including producing eNOS and uptaking ac-LDL during endothelial differentiation in spite of their feeble expression of endothelial-related genes and proteins. Additionally, we surprisingly found that BMSCs and PMSCs could directly form tubular structures in vitro on Matrigel and their conditioned medium showed significant proangiogenic bioactivities on endothelial cells in vitro compared with those of AMSCs and UMSCs. Besides, several angiogenic genes were upregulated in BMSCs and PMSCs in comparison with AMSCs and UMSCs. Moreover, enzyme-linked immunosorbent assay further confirmed that BMSCs secreted much more VEGF, and PMSCs secreted much more HGF and PGE2. Conclusions Our study demonstrated the heterogeneous proangiogenic properties of MSCs derived from different tissue origins, and the in vivo isolated environment might contribute to these differences. Our study suggested that MSCs derived from bone marrow and placental chorionic villi might be preferred in clinical application for therapeutic angiogenesis. Electronic supplementary material The online version of this article (doi:10.1186/s13287-016-0418-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wen Jing Du
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Disease, Chinese Academy of Medical Science & Peking Union Medical College, No. 288, Nanjing Road, Heping District, Tianjin, 300020, China
| | - Ying Chi
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Disease, Chinese Academy of Medical Science & Peking Union Medical College, No. 288, Nanjing Road, Heping District, Tianjin, 300020, China
| | - Zhou Xin Yang
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Disease, Chinese Academy of Medical Science & Peking Union Medical College, No. 288, Nanjing Road, Heping District, Tianjin, 300020, China
| | - Zong Jin Li
- Beijing Institute of Health and Stem Cells, No. 1, Kangding Road, BDA, Beijing, 100176, China
| | - Jun Jie Cui
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Disease, Chinese Academy of Medical Science & Peking Union Medical College, No. 288, Nanjing Road, Heping District, Tianjin, 300020, China
| | - Bao Quan Song
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Disease, Chinese Academy of Medical Science & Peking Union Medical College, No. 288, Nanjing Road, Heping District, Tianjin, 300020, China
| | - Xue Li
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Disease, Chinese Academy of Medical Science & Peking Union Medical College, No. 288, Nanjing Road, Heping District, Tianjin, 300020, China
| | - Shao Guang Yang
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Disease, Chinese Academy of Medical Science & Peking Union Medical College, No. 288, Nanjing Road, Heping District, Tianjin, 300020, China
| | - Zhi Bo Han
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Disease, Chinese Academy of Medical Science & Peking Union Medical College, No. 288, Nanjing Road, Heping District, Tianjin, 300020, China.
| | - Zhong Chao Han
- The State Key Laboratory of Experimental Hematology, Institute of Hematology and Hospital of Blood Disease, Chinese Academy of Medical Science & Peking Union Medical College, No. 288, Nanjing Road, Heping District, Tianjin, 300020, China. .,Beijing Institute of Health and Stem Cells, No. 1, Kangding Road, BDA, Beijing, 100176, China.
| |
Collapse
|
193
|
Anderson JD, Pham MT, Contreras Z, Hoon M, Fink KD, Johansson HJ, Rossignol J, Dunbar GL, Showalter M, Fiehn O, Bramlett CS, Bardini RL, Bauer G, Fury B, Hendrix KJ, Chedin F, EL-Andaloussi S, Hwang B, Mulligan MS, Lehtiö J, Nolta JA. Mesenchymal stem cell-based therapy for ischemic stroke. Chin Neurosurg J 2016. [DOI: 10.1186/s41016-016-0053-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
194
|
Vigani B, Mastracci L, Grillo F, Perteghella S, Preda S, Crivelli B, Antonioli B, Galuzzi M, Tosca MC, Marazzi M, Torre ML, Chlapanidas T. Local biological effects of adipose stromal vascular fraction delivery systems after subcutaneous implantation in a murine model. J BIOACT COMPAT POL 2016; 31:600-612. [DOI: 10.1177/0883911516635841] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2023]
Abstract
The aim of this study was to test alginate beads and silk fibroin non-woven mats as stromal vascular fraction delivery systems to support cell implantation for tissue repair and regeneration, through trophic and immunomodulant paracrine signaling. Furthermore, in vivo scaffold biocompatibility was histologically analyzed in a murine model at different time endpoints, with particular focus on construct-induced vascularization and neoangiogenesis. The fibroin mat induced a typical foreign body reaction, recruiting macrophages and giant cells and concurrently promoted neovascularization of the implanted construct. Conversely, alginate beads triggered a more circumscribed, chronic inflammatory reaction, which decreased over time. The combined in vivo implantation of alginate beads and fibroin mat with stromal vascular fraction promoted vascularization and integration of scaffolds into the surrounding subcutaneous environment. The new blood vessel ingrowth should, hopefully, support engineered cell viability and functionality, as well as the transport of soluble bioactive molecules. Due to their neovascularization properties, stromal vascular fraction administration, using alginate or fibroin scaffolds, is a new, promising, cost-effective tissue engineering approach.
Collapse
Affiliation(s)
- Barbara Vigani
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Luca Mastracci
- Pathology Section, Department of Surgical and Integrated Diagnostic Sciences (DISC), University of Genoa, IRCCS AOU San Martino—IST, Genoa, Italy
| | - Federica Grillo
- Pathology Section, Department of Surgical and Integrated Diagnostic Sciences (DISC), University of Genoa, IRCCS AOU San Martino—IST, Genoa, Italy
| | | | - Stefania Preda
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | | | - Barbara Antonioli
- Struttura Semplice Tissue Therapy, Niguarda Ca’ Granda Hospital, Milan, Italy
| | - Marta Galuzzi
- Department of Drug Sciences, University of Pavia, Pavia, Italy
- Struttura Semplice Tissue Therapy, Niguarda Ca’ Granda Hospital, Milan, Italy
| | - Marta C Tosca
- Struttura Semplice Tissue Therapy, Niguarda Ca’ Granda Hospital, Milan, Italy
| | - Mario Marazzi
- Struttura Semplice Tissue Therapy, Niguarda Ca’ Granda Hospital, Milan, Italy
| | - Maria L Torre
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | | |
Collapse
|
195
|
Nakamura M, Samii A, Lang JM, Götz F, Samii M, Krauss JK. De Novo Arteriovenous Malformation Growth Secondary to Implantation of Genetically Modified Allogeneic Mesenchymal Stem Cells in the Brain. Neurosurgery 2016; 78:E596-600. [PMID: 26382859 DOI: 10.1227/neu.0000000000001025] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND AND IMPORTANCE Local biological drug delivery in the brain is an innovative field of medicine that developed rapidly in recent years. Our report illustrates a unique case of de novo development of a cerebral arteriovenous malformation (AVM) after implantation of genetically modified allogeneic mesenchymal stem cells in the brain. CLINICAL PRESENTATION A 50-year-old man was included in a prospective clinical study (study ID number CM GLP-1/01, 2007-004516-31) investigating a novel neuroprotective approach in stroke patients to prevent perihematomal neuronal damage. In this study, alginate microcapsules containing genetically modified allogeneic mesenchymal stem cells producing the neuroprotective glucagon-like peptide-1 (GLP-1) were implanted. Three years later, the patient presented with aphasia and a focal seizure due to a new left frontal intracerebral hemorrhage. Angiography revealed a de novo left frontal AVM. CONCLUSION The development of an AVM within a period of 3 years after implantation of the glucagon-like peptide-1-secreting mesenchymal stem cells suggests a possible relationship. This case exemplifies that further investigations are necessary to assess the safety of genetically modified cell lines for local biological drug delivery in the brain.
Collapse
Affiliation(s)
- Makoto Nakamura
- *Department of Neurosurgery, Hannover Medical University, Hannover, Germany;‡Department of Neurosurgery, International Neuroscience Institute, Hannover, Germany;§Institute of Diagnostic and Interventional Neuroradiology, Hannover Medical University, Hannover, Germany
| | | | | | | | | | | |
Collapse
|
196
|
Stem Cells of Dental Origin: Current Research Trends and Key Milestones towards Clinical Application. Stem Cells Int 2016; 2016:4209891. [PMID: 27818690 PMCID: PMC5081960 DOI: 10.1155/2016/4209891] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 09/14/2016] [Indexed: 12/17/2022] Open
Abstract
Dental Mesenchymal Stem Cells (MSCs), including Dental Pulp Stem Cells (DPSCs), Stem Cells from Human Exfoliated Deciduous teeth (SHED), and Stem Cells From Apical Papilla (SCAP), have been extensively studied using highly sophisticated in vitro and in vivo systems, yielding substantially improved understanding of their intriguing biological properties. Their capacity to reconstitute various dental and nondental tissues and the inherent angiogenic, neurogenic, and immunomodulatory properties of their secretome have been a subject of meticulous and costly research by various groups over the past decade. Key milestone achievements have exemplified their clinical utility in Regenerative Dentistry, as surrogate therapeutic modules for conventional biomaterial-based approaches, offering regeneration of damaged oral tissues instead of simply “filling the gaps.” Thus, the essential next step to validate these immense advances is the implementation of well-designed clinical trials paving the way for exploiting these fascinating research achievements for patient well-being: the ultimate aim of this ground breaking technology. This review paper presents a concise overview of the major biological properties of the human dental MSCs, critical for the translational pathway “from bench to clinic.”
Collapse
|
197
|
Rombouts C, Giraud T, Jeanneau C, About I. Pulp Vascularization during Tooth Development, Regeneration, and Therapy. J Dent Res 2016; 96:137-144. [PMID: 28106505 DOI: 10.1177/0022034516671688] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The pulp is a highly vascularized tissue situated in an inextensible environment surrounded by rigid dentin walls, with the apical foramina being the only access. The pulp vascular system is not only responsible for nutrient supply and waste removal but also contributes actively to the pulp inflammatory response and subsequent regeneration. This review discusses the underlying mechanisms of pulp vascularization during tooth development, regeneration, and therapeutic procedures, such as tissue engineering and tooth transplantation. Whereas the pulp vascular system is established by vasculogenesis during embryonic development, sprouting angiogenesis is the predominant process during regeneration and therapeutic processes. Hypoxia can be considered a common driving force. Dental pulp cells under hypoxic stress release proangiogenic factors, with vascular endothelial growth factor being one of the most potent. The benefit of exogenous vascular endothelial growth factor application in tissue engineering has been well demonstrated. Interestingly, dental pulp stem cells have an important role in pulp revascularization. Indeed, recent studies show that dental pulp stem cell secretome possesses angiogenic potential that actively contributes to the angiogenic process by guiding endothelial cells and even by differentiating themselves into the endothelial lineage. Although considerable insight has been obtained in the processes underlying pulp vascularization, many questions remain relating to the signaling pathways, timing, and influence of various stress conditions.
Collapse
Affiliation(s)
- C Rombouts
- 1 Aix Marseille Univ, CNRS, ISM, Inst Movement Sci, Marseille, France
| | - T Giraud
- 1 Aix Marseille Univ, CNRS, ISM, Inst Movement Sci, Marseille, France.,2 Service d'Odontologie, Hôpital Timone, APHM, Marseille, France
| | - C Jeanneau
- 1 Aix Marseille Univ, CNRS, ISM, Inst Movement Sci, Marseille, France
| | - I About
- 1 Aix Marseille Univ, CNRS, ISM, Inst Movement Sci, Marseille, France
| |
Collapse
|
198
|
van Rhijn-Brouwer FCC, Gremmels H, Fledderus JO, Radstake TRD, Verhaar MC, van Laar JM. Cellular Therapies in Systemic Sclerosis: Recent Progress. Curr Rheumatol Rep 2016; 18:12. [PMID: 26943351 PMCID: PMC4779139 DOI: 10.1007/s11926-015-0555-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Systemic sclerosis (SSc) is a rare autoimmune connective tissue disease with a high mortality and morbidity. While progress has been made in terms of identifying high-risk patients and implementing new treatment strategies, therapeutic options remain limited. In the past few decades, various cellular therapies have emerged, which have been studied in SSc and other conditions. Here, we provide a comprehensive review of currently available cellular therapies and critically assess their merit as disease-modifying treatment for SSc. Currently, hematopoietic stem cell transplantation is the only cellular therapy that has demonstrated clinical effects on the immune system, neoangiogenesis, and fibrosis. Robust mechanistic studies as well as clinical trials are essential to move the field forward.
Collapse
Affiliation(s)
- Femke C C van Rhijn-Brouwer
- Department of Nephrology and Hypertension, Division of Internal Medicine and Dermatology, University Medical Center Utrecht, P.O. Box 85500, 3508 GA, Utrecht, The Netherlands
| | - Hendrik Gremmels
- Department of Nephrology and Hypertension, Division of Internal Medicine and Dermatology, University Medical Center Utrecht, P.O. Box 85500, 3508 GA, Utrecht, The Netherlands
| | - Joost O Fledderus
- Department of Nephrology and Hypertension, Division of Internal Medicine and Dermatology, University Medical Center Utrecht, P.O. Box 85500, 3508 GA, Utrecht, The Netherlands
| | - Timothy R D Radstake
- Department of Rheumatology & Clinical Immunology, Division of Internal Medicine and Dermatology, University Medical Center Utrecht, P.O. Box 85500, 3508 GA, Utrecht, The Netherlands
| | - Marianne C Verhaar
- Department of Nephrology and Hypertension, Division of Internal Medicine and Dermatology, University Medical Center Utrecht, P.O. Box 85500, 3508 GA, Utrecht, The Netherlands
| | - Jacob M van Laar
- Department of Rheumatology & Clinical Immunology, Division of Internal Medicine and Dermatology, University Medical Center Utrecht, P.O. Box 85500, 3508 GA, Utrecht, The Netherlands.
| |
Collapse
|
199
|
Han YS, Lee JH, Yoon YM, Yun CW, Noh H, Lee SH. Hypoxia-induced expression of cellular prion protein improves the therapeutic potential of mesenchymal stem cells. Cell Death Dis 2016; 7:e2395. [PMID: 27711081 PMCID: PMC5133977 DOI: 10.1038/cddis.2016.310] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 08/30/2016] [Accepted: 09/05/2016] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) are ‘adult' multipotent cells that promote regeneration of injured tissues in vivo. However, differences in oxygenation levels between normoxic culture conditions (21% oxygen) and both the MSC niche (2–8% oxygen) and ischemic injury-induced oxidative stress conditions in vivo have resulted in low efficacy of MSC therapies in both pre-clinical and clinical studies. To address this issue, we examined the effectiveness of hypoxia preconditioning (2% oxygen) for enhancing the bioactivity and tissue-regenerative potential of adipose-derived MSCs. Hypoxia preconditioning enhanced the proliferative potential of MSCs by promoting the expression of normal cellular prion protein (PrPC). In particular, hypoxia preconditioning-mediated MSC proliferation was regulated by PrPC-dependent JAK2 and STAT3 activation. In addition, hypoxia preconditioning-induced PrPC regulated superoxide dismutase and catalase activity, and inhibited oxidative stress-induced apoptosis via inactivation of cleaved caspase-3. In a murine hindlimb ischemia model, hypoxia preconditioning enhanced the survival and proliferation of transplanted MSCs, ultimately resulting in improved functional recovery of the ischemic tissue, including the ratio of blood flow perfusion, limb salvage, and neovascularization. These results suggest that Hypo-MSC offer a therapeutic strategy for accelerated neovasculogenesis in ischemic diseases, and that PrPC comprises a potential target for MSC-based therapies.
Collapse
Affiliation(s)
- Yong-Seok Han
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Jun Hee Lee
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham School of Medicine, Baltimore, AL 35294, USA
| | - Yeo Min Yoon
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Chul Won Yun
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea
| | - Hyunjin Noh
- Department of Internal Medicine, Soonchunhyang University, Seoul, Republic of Korea.,Hyonam Kidney Laboratory, Soonchunhyang University, Seoul, Republic of Korea
| | - Sang Hun Lee
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Republic of Korea.,Departments of Biochemistry, Soonchunhyang University College of Medicine, Cheonan 330-930, Republic of Korea
| |
Collapse
|
200
|
Dongsheng H, Zhuo Z, Jiamin L, Hailan M, Lijuan H, Fan C, Dan Y, He Z, Yun X. Proteomic Analysis of the Peri-Infarct Area after Human Umbilical Cord Mesenchymal Stem Cell Transplantation in Experimental Stroke. Aging Dis 2016; 7:623-634. [PMID: 27699085 PMCID: PMC5036957 DOI: 10.14336/ad.2016.0121] [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: 11/15/2015] [Accepted: 01/21/2016] [Indexed: 12/27/2022] Open
Abstract
Among various therapeutic approaches for stroke, treatment with human umbilical cord mesenchymal stem cells (hUC-MSCs) has acquired some promising results. However, the underlying mechanisms remain unclear. We analyzed the protein expression spectrum of the cortical peri-infarction region after ischemic stroke followed by treatment with hUC-MSCs, and found 16 proteins expressed differentially between groups treated with or without hUC-MSCs. These proteins were further determined by Gene Ontology term analysis and network with CD200-CD200R1, CCL21-CXCR3 and transcription factors. Three of them: Abca13, Grb2 and Ptgds were verified by qPCR and ELISA. We found the protein level of Abca13 and the mRNA level of Grb2 consistent with results from the proteomic analysis. Finally, the function of these proteins was described and the potential proteins that deserve to be further studied was also highlighted. Our data may provide possible underlying mechanisms for the treatment of stroke using hUC-MSCs.
Collapse
Affiliation(s)
- He Dongsheng
- 1Department of Neurology, Affiliated Drum Tower Hospital, and; 2Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing 210008, China.; 3The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210008, China; 5Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing 210008, China; 6Nanjing Neuropsychiatry Clinic Medical Center, Nanjing 210008, China
| | - Zhang Zhuo
- 4Department of Gastroenterology, Children's Hospital of Nanjing, Nanjing Medical University, Nanjing 210008, China
| | - Lao Jiamin
- 1Department of Neurology, Affiliated Drum Tower Hospital, and; 2Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing 210008, China.; 3The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210008, China; 5Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing 210008, China; 6Nanjing Neuropsychiatry Clinic Medical Center, Nanjing 210008, China
| | - Meng Hailan
- 1Department of Neurology, Affiliated Drum Tower Hospital, and; 2Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing 210008, China.; 3The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210008, China; 5Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing 210008, China; 6Nanjing Neuropsychiatry Clinic Medical Center, Nanjing 210008, China
| | - Han Lijuan
- 1Department of Neurology, Affiliated Drum Tower Hospital, and; 2Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing 210008, China.; 3The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210008, China; 5Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing 210008, China; 6Nanjing Neuropsychiatry Clinic Medical Center, Nanjing 210008, China
| | - Chen Fan
- 1Department of Neurology, Affiliated Drum Tower Hospital, and; 2Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing 210008, China.; 3The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210008, China; 5Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing 210008, China; 6Nanjing Neuropsychiatry Clinic Medical Center, Nanjing 210008, China
| | - Ye Dan
- 1Department of Neurology, Affiliated Drum Tower Hospital, and; 2Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing 210008, China.; 3The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210008, China; 5Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing 210008, China; 6Nanjing Neuropsychiatry Clinic Medical Center, Nanjing 210008, China
| | - Zhang He
- 1Department of Neurology, Affiliated Drum Tower Hospital, and; 2Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing 210008, China.; 3The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210008, China; 5Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing 210008, China; 6Nanjing Neuropsychiatry Clinic Medical Center, Nanjing 210008, China
| | - Xu Yun
- 1Department of Neurology, Affiliated Drum Tower Hospital, and; 2Jiangsu Key Laboratory for Molecular Medicine, Nanjing University Medical School, Nanjing 210008, China.; 3The State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing 210008, China; 5Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing 210008, China; 6Nanjing Neuropsychiatry Clinic Medical Center, Nanjing 210008, China
| |
Collapse
|