51
|
Mesenchymal stem cells: Immunomodulatory capability and clinical potential in immune diseases. ACTA ACUST UNITED AC 2016. [DOI: 10.1016/j.jocit.2014.12.001] [Citation(s) in RCA: 211] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
52
|
Lung Regeneration: Endogenous and Exogenous Stem Cell Mediated Therapeutic Approaches. Int J Mol Sci 2016; 17:ijms17010128. [PMID: 26797607 PMCID: PMC4730369 DOI: 10.3390/ijms17010128] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 01/07/2016] [Accepted: 01/11/2016] [Indexed: 12/25/2022] Open
Abstract
The tissue turnover of unperturbed adult lung is remarkably slow. However, after injury or insult, a specialised group of facultative lung progenitors become activated to replenish damaged tissue through a reparative process called regeneration. Disruption in this process results in healing by fibrosis causing aberrant lung remodelling and organ dysfunction. Post-insult failure of regeneration leads to various incurable lung diseases including chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis. Therefore, identification of true endogenous lung progenitors/stem cells, and their regenerative pathway are crucial for next-generation therapeutic development. Recent studies provide exciting and novel insights into postnatal lung development and post-injury lung regeneration by native lung progenitors. Furthermore, exogenous application of bone marrow stem cells, embryonic stem cells and inducible pluripotent stem cells (iPSC) show evidences of their regenerative capacity in the repair of injured and diseased lungs. With the advent of modern tissue engineering techniques, whole lung regeneration in the lab using de-cellularised tissue scaffold and stem cells is now becoming reality. In this review, we will highlight the advancement of our understanding in lung regeneration and development of stem cell mediated therapeutic strategies in combating incurable lung diseases.
Collapse
|
53
|
Xia C, Chang P, Zhang Y, Shi W, Liu B, Ding L, Liu M, Gao L, Dong L. Therapeutic effects of bone marrow-derived mesenchymal stem cells on radiation-induced lung injury. Oncol Rep 2015; 35:731-8. [PMID: 26717975 DOI: 10.3892/or.2015.4433] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 09/17/2015] [Indexed: 11/06/2022] Open
Abstract
Radiation-induced lung injury (RILI) is a fatal condition featured by interstitial pneumonitis and fibrosis. Mesenchymal stem cells (MSCs) have been widely used for treating RILI in rodent models. In the present study, we aimed to investigate whether the therapeutic effects of human bone marrow-derived mesenchymal stem cells (hBM-MSCs) on RILI were in a dose-dependent manner. A total of 100 mice were randomly divided into: a control group (n=25), subject to lung irradiation and injection of phosphate-buffered solution (PBS) via the tail vein; and the hBM-MSC group, subject to lung irradiation followed by injection of a low dose (1x103 hBM-MSCs/g), medium dose (5x103 hBM-MSCs/g) and high dose (1x104 hBM-MSCs/g) of hBM-MSCs in PBS through the tail vein, respectively. After sacrifice, the pulmonary tissues were subject to hematoxylin and eosin (H&E) staining, Masson's trichrome staining and immunohistochemical staining to investigate the pathological changes. Immunofluorescent staining was performed to evaluate the differentiation capacity of hBM-MSCs in vivo by analyzing the expression of SPC and PECAM. hBM-MSCs improved the survival rate and histopathological features in the irradiated mice, especially in the low-dose group. Marked decrease in collagen deposition was noted in the irradiated mice treated using a low dose of hBM-MSCs. In addition, hBM-MSCs attenuated secretion and expression of IL-10 and increased the expression of TNF-α. Furthermore, hBM-MSCs had the potential to differentiate into functional cells upon lung injury. Low-dose hBM-MSCs contributed to functional recovery in mice with RILI.
Collapse
Affiliation(s)
- Chengcheng Xia
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun, Jilin 130000; Key Laboratory of Radiobiology (Chinese Ministry of Health), School of Public Health, Jilin University, Changchun, Jilin 130000, P.R. China
| | - Pengyu Chang
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun, Jilin 130000; Key Laboratory of Radiobiology (Chinese Ministry of Health), School of Public Health, Jilin University, Changchun, Jilin 130000, P.R. China
| | - Yuyu Zhang
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun, Jilin 130000; Key Laboratory of Radiobiology (Chinese Ministry of Health), School of Public Health, Jilin University, Changchun, Jilin 130000, P.R. China
| | - Weiyan Shi
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun, Jilin 130000; Key Laboratory of Radiobiology (Chinese Ministry of Health), School of Public Health, Jilin University, Changchun, Jilin 130000, P.R. China
| | - Bin Liu
- Department of Orthopaedics, The First Bethune Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Lijuan Ding
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun, Jilin 130000; Key Laboratory of Radiobiology (Chinese Ministry of Health), School of Public Health, Jilin University, Changchun, Jilin 130000, P.R. China
| | - Min Liu
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun, Jilin 130000; Key Laboratory of Radiobiology (Chinese Ministry of Health), School of Public Health, Jilin University, Changchun, Jilin 130000, P.R. China
| | - Ling Gao
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun, Jilin 130000; Key Laboratory of Radiobiology (Chinese Ministry of Health), School of Public Health, Jilin University, Changchun, Jilin 130000, P.R. China
| | - Lihua Dong
- Department of Radiation Oncology, The First Bethune Hospital of Jilin University, Changchun, Jilin 130000; Key Laboratory of Radiobiology (Chinese Ministry of Health), School of Public Health, Jilin University, Changchun, Jilin 130000, P.R. China
| |
Collapse
|
54
|
Cai SX, Liu AR, Chen S, He HL, Chen QH, Xu JY, Pan C, Yang Y, Guo FM, Huang YZ, Liu L, Qiu HB. The Orphan Receptor Tyrosine Kinase ROR2 Facilitates MSCs to Repair Lung Injury in ARDS Animal Model. Cell Transplant 2015; 25:1561-74. [PMID: 26531175 DOI: 10.3727/096368915x689776] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
There are some limitations to the therapeutic effects of mesenchymal stem cells (MSCs) on acute respiratory distress syndrome (ARDS) due to their low engraftment and differentiation rates in lungs. We found previously that noncanonical Wnt5a signaling promoted the differentiation of mouse MSCs (mMSCs) into type II alveolar epithelial cells (AT II cells), conferred resistance to oxidative stress, and promoted migration of MSCs in vitro. As receptor tyrosine kinase-like orphan receptor 2 (ROR2) is an essential receptor for Wnt5a, it was reasonable to deduce that ROR2 might be one of the key molecules for the therapeutic effect of MSCs in ARDS. The mMSCs that stably overexpressed ROR2 or the green fluorescent protein (GFP) control were transplanted intratracheally into the ARDS mice [induced by intratracheal injection of lipopolysaccharide (LPS)]. The results showed that ROR2-overexpressing mMSCs led to more significant effects than the GFP controls, including the retention of the mMSCs in the lung, differentiation into AT II cells, improvement of alveolar epithelial permeability, improvement of acute LPS-induced pulmonary inflammation, and, finally, reduction of the pathological impairment of the lung tissue. In conclusion, MSCs that overexpress ROR2 could further improve MSC-mediated protection against epithelial impairment in ARDS.
Collapse
Affiliation(s)
- Shi-Xia Cai
- Department of Critical Care Medicine, Nanjing Zhong-da Hospital, School of Medicine, Southeast University, Nanjing, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
55
|
Xu X, Li L, Wang C, Liu Y, Chen C, Yan J, Ding H, Tang SY. The expansion of autologous adipose-derived stem cells in vitro for the functional reconstruction of nasal mucosal tissue. Cell Biosci 2015; 5:54. [PMID: 26388989 PMCID: PMC4574024 DOI: 10.1186/s13578-015-0045-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 09/04/2015] [Indexed: 02/07/2023] Open
Abstract
Background I
t is established that adipose-derived stem cells (ADSCs) produce and secrete cytokines/growth factors that antagonize mucosal injury. However, the exact molecular basis underlying the treatment effects exerted by ADSCs is ill understood, and whether ADSCs cooperate with adipose tissue particles to improve mucosal function in patients with empty nose syndrome (ENS) has not been explored. We investigated the impact of ADSCs on nasal mucosa, the associated mechanisms, and their use in the treatment of patients with ENS. Results
The nasal endoscope and mucociliary clearance assessments were significantly improved (P < 0.05) in patients with (n = 28) and without (n = 2) a rudimentary turbinate that received ADSCs combined with fat granules transplantation. Patients experienced a significant improvement in nasal obstruction and nasal mucociliary clearance after nasal turbinate angioplasty (P < 0.05). H&E staining, Masson’s staining, and AB-PAS staining confirmed that inflammation was significantly reduced, collagenous fibers became aligned, fewer deposits were observed, and the mucosal proteins generated from caliciform cells increased following treatment. After a 14-day incubation period, ADSCs developed a polygonal cobblestone shape characteristic of human epithelial cells. Furthermore, immunohistochemical analysis revealed the presence of epithelial markers such as cytokeratin-7, and cytokeratin-19. Western blot analysis showed the presence of specific epithelial cell markers including cytokeratin-7, cytokeratin-14 and cytokeratin-19 in these epithelial like cells (ELC); these markers had low expression levels of ADSCs. Conclusions The reconstruction of mucosal function by nasal turbinate angioplasty combined with ADSCs and autologous adipose tissue particle transplantation significantly improved the symptoms of patients with ENS. This is a new procedure that will improve mucosal restoration treatment options in patients with ENS. Furthermore, we undertook preliminary explorations of the underlying mechanisms involved, and found that transplantation of ADSCs could induce epithelial cells to improve mucosa function in patients with ENS in the micro-environment of injection areas.
Collapse
Affiliation(s)
- Xiao Xu
- Skin and Reconstructive Medicine Department, The General Hospital of Chinese People's Armed Police Forces, Beijing, People's Republic of China
| | - Liang Li
- Skin and Reconstructive Medicine Department, The General Hospital of Chinese People's Armed Police Forces, Beijing, People's Republic of China
| | - Cheng Wang
- Skin and Reconstructive Medicine Department, The General Hospital of Chinese People's Armed Police Forces, Beijing, People's Republic of China
| | - Yang Liu
- Skin and Reconstructive Medicine Department, The General Hospital of Chinese People's Armed Police Forces, Beijing, People's Republic of China
| | - Chong Chen
- Skin and Reconstructive Medicine Department, The General Hospital of Chinese People's Armed Police Forces, Beijing, People's Republic of China
| | - Junling Yan
- Skin and Reconstructive Medicine Department, The General Hospital of Chinese People's Armed Police Forces, Beijing, People's Republic of China
| | - Hong Ding
- Skin and Reconstructive Medicine Department, The General Hospital of Chinese People's Armed Police Forces, Beijing, People's Republic of China
| | - Su-Yang Tang
- Skin and Reconstructive Medicine Department, The General Hospital of Chinese People's Armed Police Forces, Beijing, People's Republic of China
| |
Collapse
|
56
|
Zhou MI, Chen DL, Jiang T, Feng YM, Han XL. Effects of bone marrow-derived mesenchymal stem cells transfected with survivin on pulmonary fibrosis in mice. Exp Ther Med 2015; 10:1857-1864. [PMID: 26640562 DOI: 10.3892/etm.2015.2715] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 07/17/2015] [Indexed: 12/15/2022] Open
Abstract
The aim of the present study was to investigate the effects of bone marrow-derived mesenchymal stem cells (BMSCs) transfected with survivin on lung fibrosis in mice. Mice with bleomycin-induced pulmonary fibrosis were allocated at random to group A, B or C, and injected with 1×106 survivin gene-expressing BMSCs, 1×106 BMSCs or normal saline, respectively. A total of 6 mice were sacrificed from each group on days 7, 14 and 28 after treatment. The extent of alveolitis and pulmonary fibrosis was assessed and the apoptotic rates of the BMSCs and survivin-expressing BMSCs were detected. The content of surfactant protein A (SP-A) in the lung and hydroxyproline (Hyp) in the serum was measured. The mRNA expression levels of transforming growth factor (TGF)-β1 and matrix metalloproteinase (MMP)-9 in the lung tissue of the mice was detected. Furthermore, the protein expression levels of caspase-3 and -9 were detected. The apoptotic rates of the BMSCs (group B) and survivin-expressing BMSCs (group A) were 14.466±1.953 and 7.718±0.493%, respectively. The degree of lung fibrosis in groups A and B was reduced compared with that in group C. The hydroxyproline content in groups A and B was reduced compared with that in group C, and the SP-A content in groups A and B was increased compared with that in group C. The mRNA expression levels of TGF-β1 in group A were reduced compared with those in group B, and the levels in group B were reduced compared with those in group C. By contrast, the mRNA expression levels of MMP-9 in group A were increased compared with those in groups B and C, and the levels in group B were increased compared with those in group A. The expression levels of caspase-3 and -9 in group A were elevated compared with those in groups B and C. In conclusion, BMSCs are effective in preventing bleomycin-induced lung fibrosis, and survivin may enhance the protective effects of BMSCs.
Collapse
Affiliation(s)
- M I Zhou
- Department of Respiratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400042, P.R. China
| | - Dong-Ling Chen
- Department of Respiratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400042, P.R. China
| | - Tao Jiang
- Department of Respiratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400042, P.R. China
| | - Yan-Mei Feng
- Department of Respiratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400042, P.R. China
| | - Xiao-Li Han
- Department of Respiratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400042, P.R. China
| |
Collapse
|
57
|
Abd El Salam NF, Hafez MS, Omar SM, el Sayed HF. The role of bone marrow-derived mesenchymal stem cells in a rat model of paraquat-induced lung fibrosis. THE EGYPTIAN JOURNAL OF HISTOLOGY 2015; 38:389-401. [DOI: 10.1097/01.ehx.0000464786.52906.32] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
58
|
Devaney J, Horie S, Masterson C, Elliman S, Barry F, O'Brien T, Curley GF, O'Toole D, Laffey JG. Human mesenchymal stromal cells decrease the severity of acute lung injury induced by E. coli in the rat. Thorax 2015; 70:625-35. [PMID: 25986435 DOI: 10.1136/thoraxjnl-2015-206813] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 04/06/2015] [Indexed: 01/08/2023]
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs) demonstrate considerable promise in preclinical acute respiratory distress syndrome models. We wished to determine the efficacy and mechanisms of action of human MSCs (hMSCs) in the setting of acute lung injury induced by prolonged Escherichia coli pneumonia in the rat. METHODS Adult male Sprague Dawley rats underwent intratracheal instillation of E. coli bacteria in all experiments. In Series 1, animals were randomised to intravenous administration of: (1) vehicle (phosphate buffered saline (PBS), 300 μL); (2) 1×10(7) fibroblasts/kg; (3) 1×10(7) hMSCs/kg or (4) 2×10(7) hMSCs/kg. Series 2 determined the lowest effective hMSC dose. Series 3 compared the efficacy of intratracheal versus intravenous hMSC administration, while Series 4 examined the efficacy of cryopreserved hMSC. Series 5 examined the efficacy of the hMSC secretome. Parallel in vitro experiments further assessed the potential for hMSCs to secrete LL-37 and modulate macrophage phagocytosis. RESULTS hMSC therapy reduced the severity of rodent E. coli pneumonia, improving survival, decreasing lung injury, reducing lung bacterial load and suppressing inflammation. Doses as low as 5×10(6) hMSCs/kg were effective. Intratracheal hMSC therapy was as effective as intravenous hMSC. Cryopreserved hMSCs were also effective, while the hMSC secretome was less effective in this model. hMSC therapy enhanced macrophage phagocytic capacity and increased lung and systemic concentrations of the antimicrobial peptide LL37. CONCLUSIONS hMSC therapy decreased E. coli induced pneumonia injury and reduced lung bacterial burden, potentially via enhanced macrophage phagocytosis and increased alveolar LL-37 concentrations.
Collapse
Affiliation(s)
- James Devaney
- Department of Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, Galway, Ireland Regenerative Medicine Institute, National University of Ireland, Galway, Ireland
| | - Shahd Horie
- Department of Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, Galway, Ireland Regenerative Medicine Institute, National University of Ireland, Galway, Ireland
| | - Claire Masterson
- Department of Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, Galway, Ireland Regenerative Medicine Institute, National University of Ireland, Galway, Ireland
| | - Steve Elliman
- Orbsen Therapeutics Ltd, National University of Ireland, Galway, Ireland
| | - Frank Barry
- Regenerative Medicine Institute, National University of Ireland, Galway, Ireland
| | - Timothy O'Brien
- Regenerative Medicine Institute, National University of Ireland, Galway, Ireland
| | - Gerard F Curley
- Department of Anesthesia, Critical Illness and Injury Research Centre, Keenan Research Centre for Biomedical Science, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Daniel O'Toole
- Department of Anaesthesia, School of Medicine, Clinical Sciences Institute, National University of Ireland, Galway, Ireland Regenerative Medicine Institute, National University of Ireland, Galway, Ireland
| | - John G Laffey
- Department of Anesthesia, Critical Illness and Injury Research Centre, Keenan Research Centre for Biomedical Science, St Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
59
|
The role of vascular endothelial growth factor receptor-1 signaling in compensatory contralateral lung growth following unilateral pneumonectomy. J Transl Med 2015; 95:456-68. [PMID: 25642830 DOI: 10.1038/labinvest.2014.159] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 10/31/2014] [Accepted: 12/02/2014] [Indexed: 01/09/2023] Open
Abstract
Compensatory lung growth models have been widely used to investigate alveolization because the remaining lung can be kept intact and volume loss can be controlled. Vascular endothelial growth factor (VEGF) plays an important role in blood formation during lung growth and repair, but the precise mechanisms involved are poorly understood; therefore, the aim of this study was to investigate the role of VEGF signaling in compensatory lung growth. After left pneumonectomy, the right lung weight was higher in VEGF transgenic mice than wild-type (WT) mice. Compensatory lung growth was suppressed significantly in mice injected with a VEGF neutralizing antibody and in VEGF receptor-1 tyrosine kinase-deficient mice (TK(-/-) mice). The mobilization of progenitor cells expressing VEGFR1(+) cells from bone marrow and the recruitment of these cells to lung tissue were also suppressed in the TK(-/-) mice. WT mice transplanted with bone marrow from TK(-/-)transgenic GFP(+) mice had significantly lower numbers of GFP(+)/aquaporin 5(+), GFP(+)/surfactant protein A(+), and GFP(+)/VEGFR1(+) cells than WT mice transplanted with bone marrow from WTGFP(+) mice. The GFP(+)/VEGFR1(+) cells also co-stained for aquaporin 5 and surfactant protein A. Overall, these results suggest that VEGF signaling contributes to compensatory lung growth by mobilizing VEGFR1(+) cells.
Collapse
|
60
|
Gao P, Yang J, Gao X, Xu D, Niu D, Li J, Wen Q. Salvianolic acid B improves bone marrow-derived mesenchymal stem cell differentiation into alveolar epithelial cells type I via Wnt signaling. Mol Med Rep 2015; 12:1971-6. [PMID: 25892295 DOI: 10.3892/mmr.2015.3632] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Accepted: 03/04/2015] [Indexed: 11/06/2022] Open
Abstract
Acute lung injury (ALI) is among the most common causes of mortality in intensive care units. Previous studies have suggested that bone marrow-derived mesenchymal stem cells (BMSCs) may attenuate pulmonary edema. In addition, alveolar epithelial cells type I (ATI) are involved in reducing the alveolar edema in response to ALI. However, the mechanism involved in improving the efficiency of differentiation of MSCs into ATI remains to be elucidated. In the present study, the effect of salvianolic acid B (Sal B) on the differentiation of BMSCs into ATI and the activities of the Wnt signaling pathways were investigated. The BMSCs were supplemented with conditioned medium (CM). The groups were as follows: i) CM group: BMSCs were supplemented with CM; ii) lithium chloride (LiCl) group: BMSCs were supplemented with CM and 5 mM LiCl; iii) Sal B group: BMSCs were supplemented with CM and 10 mM Sal B. The samples were collected and assessed on days 7 and 14. It was revealed that aquaporin (AQP)-5 and T1α were expressed in BMSCs, and induction with LiCl or Sal B increased the expression of AQP-5 and T1α. Furthermore, the Wnt-1 and Wnt-3a signaling pathways were activated during the differentiation of BMSCs into ATI. In conclusion, it was suggested that the promotive effects of Sal B on the differentiation of BMSCs into ATI occurred through the activation of Wnt signaling pathways.
Collapse
Affiliation(s)
- Peng Gao
- Department of Anesthesiology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Jingxian Yang
- Department of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, Liaoning 116600, P.R. China
| | - Xi Gao
- Department of Anesthesiology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Dan Xu
- Department of Anesthesiology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Dongge Niu
- Department of Anesthesiology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Jinglin Li
- Department of Anesthesiology, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116011, P.R. China
| | - Qingping Wen
- Department of Anesthesiology, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| |
Collapse
|
61
|
Cai SX, Liu AR, Chen S, He HL, Chen QH, Xu JY, Pan C, Yang Y, Guo FM, Huang YZ, Liu L, Qiu HB. Activation of Wnt/β-catenin signalling promotes mesenchymal stem cells to repair injured alveolar epithelium induced by lipopolysaccharide in mice. Stem Cell Res Ther 2015; 6:65. [PMID: 25889393 PMCID: PMC4414385 DOI: 10.1186/s13287-015-0060-y] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2014] [Revised: 02/03/2015] [Accepted: 03/20/2015] [Indexed: 01/11/2023] Open
Abstract
Introduction Mesenchymal stem cells (MSCs) have potential for re-epithelization and recovery in acute respiratory distress syndrome (ARDS). In a previous in vitro study, the results showed that the canonical Wnt/β-catenin pathway promoted the differentiation of MSCs into type II alveolar epithelial cells, conferred resistance to oxidative stress, and promoted their migration, suggesting that the Wnt/β-catenin pathway might be one of the key mechanisms underling the therapeutic effect of mouse MSCs in ARDS. Methods Mouse MSCs stable transfected with β-catenin or green fluorescent protein control were transplanted intratracheally into the ARDS mice induced by lipopolysaccharide. Lung tissue injury and repair assessment were examined using haematoxylin and eosin staining, lung injury scoring, Masson’s trichrome staining and fibrosis scoring. Homing and differentiation of mouse MSCs were assayed by labelling and tracing MSCs using NIR815 dye, immunofluorescent staining, and Western immunoblot analysis. The inflammation and permeability were evaluated by detecting the cytokine and protein measurements in bronchoalveolar lavage fluid using enzyme-linked immunosorbent assay. Results In this study, β-catenin-overexpressing MSC engraftment led to more significant effects than the GFP controls, including the retention of the MSCs in the lung, differentiation into type II alveolar epithelial cells, improvement in alveolar epithelial permeability, and the pathologic impairment of the lung tissue. Conclusion These results suggest that the activation of canonical Wnt/β-catenin pathway by mouse MSCs by overexpressing β-catenin could further improve the protection of mouse MSCs against epithelial impair and the therapeutic effects of mouse MSCs in ARDS mice.
Collapse
Affiliation(s)
- Shi-xia Cai
- Department of Critical Care Medicine, Zhong-da Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, People's Republic of China. .,Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266003, People's Republic of China.
| | - Ai-ran Liu
- Department of Critical Care Medicine, Zhong-da Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, People's Republic of China.
| | - Song Chen
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China.
| | - Hong-li He
- Department of Critical Care Medicine, Zhong-da Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, People's Republic of China.
| | - Qi-hong Chen
- Department of Critical Care Medicine, Zhong-da Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, People's Republic of China.
| | - Jing-yuan Xu
- Department of Critical Care Medicine, Zhong-da Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, People's Republic of China.
| | - Chun Pan
- Department of Critical Care Medicine, Zhong-da Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, People's Republic of China.
| | - Yi Yang
- Department of Critical Care Medicine, Zhong-da Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, People's Republic of China.
| | - Feng-mei Guo
- Department of Critical Care Medicine, Zhong-da Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, People's Republic of China.
| | - Ying-zi Huang
- Department of Critical Care Medicine, Zhong-da Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, People's Republic of China.
| | - Ling Liu
- Department of Critical Care Medicine, Zhong-da Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, People's Republic of China.
| | - Hai-bo Qiu
- Department of Critical Care Medicine, Zhong-da Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
62
|
Jin Z, Pan X, Zhou K, Bi H, Wang L, Yu L, Wang Q. Biological effects and mechanisms of action of mesenchymal stem cell therapy in chronic obstructive pulmonary disease. J Int Med Res 2015; 43:303-10. [PMID: 25834280 DOI: 10.1177/0300060514568733] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 12/22/2014] [Indexed: 02/06/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is the most frequent chronic respiratory disease and a leading cause of morbidity and mortality, worldwide. Given that the foremost risk factor leading to the development of COPD is cigarette smoke, the initial treatment for COPD is smoking cessation. Even after smoking cessation, inflammation, apoptosis and oxidative stress can persist and continue to contribute to COPD. Although current therapies for COPD (which are primarily based on anti-inflammatory drugs such as corticosteroids, theophylline and bronchodilators) reduce airway obstruction, limit COPD exacerbation and improve the patient's health-related quality-of-life, none can prevent disease progression or reduce mortality. Recent advances in stem cell research have provided novel insight into the potential of bone marrow mesenchymal stem cells (MSCs) in the treatment of several pulmonary diseases. This review article discusses the biological effects and mechanisms of action of MSC transplantation in COPD, and highlights the foundation that MSCs provide for novel therapeutic approaches in COPD.
Collapse
Affiliation(s)
- Zhixian Jin
- Second Department of Respiratory Medicine, The First People's Hospital of Kunming, Kunming, Yunnan Province, China
| | - Xinghua Pan
- Stem Cell Engineering Laboratory of Yunnan Province, Department of Clinical Research, Kunming General Hospital of Chengdu Military Command, Kunming, Yunnan Province, China
| | - Kaihua Zhou
- Second Department of Respiratory Medicine, The First People's Hospital of Kunming, Kunming, Yunnan Province, China
| | - Hong Bi
- Second Department of Respiratory Medicine, The First People's Hospital of Kunming, Kunming, Yunnan Province, China
| | - Liyan Wang
- Second Department of Respiratory Medicine, The First People's Hospital of Kunming, Kunming, Yunnan Province, China
| | - Lu Yu
- Department of Pathology, The First People's Hospital of Kunming, Kunming, Yunnan Province, China
| | - Qing Wang
- Second Department of Respiratory Medicine, The First People's Hospital of Kunming, Kunming, Yunnan Province, China
| |
Collapse
|
63
|
Yang JX, Zhang N, Wang HW, Gao P, Yang QP, Wen QP. CXCR4 receptor overexpression in mesenchymal stem cells facilitates treatment of acute lung injury in rats. J Biol Chem 2014; 290:1994-2006. [PMID: 25492872 DOI: 10.1074/jbc.m114.605063] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Novel therapeutic regimens for tissue renewal incorporate mesenchymal stem cells (MSCs) as they differentiate into a variety of cell types and are a stem cell type that is easy to harvest and to expand in vitro. However, surface chemokine receptors, such as CXCR4, which are involved in the mobilization of MSCs, are expressed only on the surface of a small proportion of MSCs, and the lack of CXCR4 expression may underlie the low efficiency of homing of MSCs toward tissue damage, which results in a poor curative effect. Here, a rat CXCR4 expressing lentiviral vector was constructed and introduced into MSCs freshly prepared from rat bone marrow. The influence of CXCR4 expression on migration, proliferation, differentiation, and paracrine effects of MSCs was examined in vitro. The in vivo properties of CXCR4-MSCs were also investigated in a model of acute lung injury in rats induced by lipopolysaccharide. Expression of CXCR4 in MSCs significantly enhanced the chemotactic and paracrine characteristics of the cells in vitro but did not affect self-renewal or differentiation into alveolar and vascular endothelial cells. In vivo, CXCR4 improved MSC homing and colonization of damaged lung tissue, and furthermore, the transplanted CXCR4-MSCs suppressed the development of acute lung injury in part by modulating levels of inflammatory molecules and the neutrophil count. These results indicated that efficient mobilization of MSCs to sites of tissue injury may be due to CXCR4, and therefore, increased expression of CXCR4 may improve their therapeutic potential in the treatment of diseases where tissue damage develops.
Collapse
Affiliation(s)
- Jing-Xian Yang
- From the School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China
| | - Nan Zhang
- From the School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China, School of Pharmacy, China Medical University, Shenyang 110013, China
| | - Han-Wei Wang
- First Affiliated Hospital, Dalian Medical University, Dalian 116011, China, and
| | - Peng Gao
- Department of Anesthesiology, Dalian Medical University, Dalian 116044, China
| | - Qing-Ping Yang
- From the School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian 116600, China
| | - Qing-Ping Wen
- First Affiliated Hospital, Dalian Medical University, Dalian 116011, China, and
| |
Collapse
|
64
|
Huang K, Kang X, Wang X, Wu S, Xiao J, Li Z, Wu X, Zhang W. Conversion of bone marrow mesenchymal stem cells into type II alveolar epithelial cells reduces pulmonary fibrosis by decreasing oxidative stress in rats. Mol Med Rep 2014; 11:1685-92. [PMID: 25411925 PMCID: PMC4270324 DOI: 10.3892/mmr.2014.2981] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Accepted: 11/03/2014] [Indexed: 11/29/2022] Open
Abstract
Pulmonary fibrosis is an irreversible chronic progressive fibroproliferative lung disease, which usually has a poor prognosis. Previous studies have confirmed that the transplantation of bone marrow mesenchymal stem cells (MSCs) significantly reduces lung damage in a number of animal models. However, the underlying mechanism involved in this process remains to be elucidated. In the present study, a bleomycin (BLM)-induced female Wister rat model of fibrosis was established. At 0 or 7 days following BLM administration, rats were injected into the tail vein with 5-bromo-2-deoxyuridine-labeled MSCs extracted from male Wistar rats. The lung tissue of the rats injected with MSCs expressed the sex-determining region Y gene. The level surfactant protein C (SP-C), a marker for type II alveolar epithelial cells (AEC II), was higher in the group injected with MSCs at day 0 than that in the group injected at day 7. Furthermore, SP-C mRNA, but not aquaporin 5 mRNA, a marker for type I alveolar epithelial cells, was expressed in fresh bone marrow aspirates and the fifth generation of cultured MSCs. In addition, superoxide dismutase activity and total antioxidative capability, specific indicators of oxidative stress, were significantly increased in the lung tissue of the MSC-transplanted rats (P<0.05). In conclusion, to alleviate pulmonary fibrosis, exogenous MSCs may be transplanted into damaged lung tissue where they differentiate into AEC II and exert their effect, at least in part, through blocking oxidative stress.
Collapse
Affiliation(s)
- Kun Huang
- Department of Respiratory Medicine, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Xiaowen Kang
- Department of Respiratory Medicine, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Xinyan Wang
- Department of Respiratory Medicine, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Shijie Wu
- Department of Respiratory Medicine, Daqing Oilfield General Hospital, Daqing, Heilongjiang 163316, P.R. China
| | - Jinling Xiao
- Department of Respiratory Medicine, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Zhaoguo Li
- Department of Respiratory Medicine, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Xiaomei Wu
- Department of Respiratory Medicine, The Second Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Wei Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
65
|
Sdrimas K, Kourembanas S. MSC microvesicles for the treatment of lung disease: a new paradigm for cell-free therapy. Antioxid Redox Signal 2014; 21:1905-15. [PMID: 24382303 PMCID: PMC4202925 DOI: 10.1089/ars.2013.5784] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
SIGNIFICANCE Bronchopulmonary dysplasia (BPD), also known as chronic lung disease of infancy, is a major complication of preterm birth that, despite improvements in neonatal respiratory support and perinatal care, remains an important cause of morbidity and mortality, often with severe adverse neurodevelopmental sequelae. Even with major advances in our understanding of the pathogenesis of this disease, BPD remains essentially without adequate treatment. RECENT ADVANCES Cell-based therapies arose as a promising treatment for acute and chronic lung injury in many experimental models of disease. Currently, more than 3000 human clinical trials employing cell therapy for the treatment of diverse diseases, including cardiac, neurologic, immune, and respiratory conditions, are ongoing or completed. Among the treatments, mesenchymal stem cells (MSCs) are the most studied and have been extensively tested in experimental models of BPD, pulmonary hypertension, pulmonary fibrosis, and acute lung injury. CRITICAL ISSUES Despite the promising potential, MSC therapy for human lung disease still remains at an experimental stage and optimal transplantation conditions need to be determined. Although the mechanism of MSC action can be manifold, accumulating evidence suggests a predominant paracrine, immunomodulatory, and cytoprotective effect. FUTURE DIRECTIONS The current review summarizes the effect of MSC treatment in models of lung injury, including BPD, and focuses on the MSC secretome and, specifically, MSC-derived microvesicles as potential key mediators of therapeutic action that can be the focus of future therapies.
Collapse
Affiliation(s)
- Konstantinos Sdrimas
- 1 Division of Newborn Medicine, Boston Children's Hospital , Boston, Massachusetts
| | | |
Collapse
|
66
|
Quan H, Kim SK, Heo SJ, Koak JY, Lee JH. Optimization of growth inducing factors for colony forming and attachment of bone marrow-derived mesenchymal stem cells regarding bioengineering application. J Adv Prosthodont 2014; 6:379-86. [PMID: 25352960 PMCID: PMC4211054 DOI: 10.4047/jap.2014.6.5.379] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 06/16/2014] [Accepted: 07/11/2014] [Indexed: 12/14/2022] Open
Abstract
PURPOSE These days, mesenchymal stem cells (MSCs) have received worldwide attention because of their potentiality in tissue engineering for implant dentistry. The purpose of this study was to evaluate various growth inducing factors in media for improvement of acquisition of bone marrow mesenchymal stem cells (BMMSCs) and colony forming unit-fibroblast (CFU-F). MATERIALS AND METHODS The mouse BMMSCs were freshly obtained from female C3H mouse femur and tibia. The cells seeded at the density of 106/dish in media supplemented with different density of fetal bovine serum (FBS), 1α, 25-dihydroxyvitamin (VD3) and recombinant human epidermal growth factor (rhEGF). After 14 days, CFU-F assay was conducted to analyze the cell attachment and proliferation, and moreover for VD3, the 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS) assay was additionally conducted. RESULTS The cell proliferation was increased with the increase of FBS concentration (P<.05). The cell proliferation was highest at the density of 20 ng/mL rhEGF compared with 0 ng/mL and 200 ng/mL rhEGF (P<.05). For VD3, although the colony number was increased with the increase of its concentration, the difference was not statistically significant (P>.05). CONCLUSION FBS played the main role in cell attachment and growth, and the growth factor like rhEGF played the additional effect. However, VD3 did not have much efficacy compare with the other two factors. Improvement of the conditions could be adopted to acquire more functional MSCs to apply into bony defect around implants easily.
Collapse
Affiliation(s)
- Hongxuan Quan
- Department of Prosthodontics & Dental Research Institute, Seoul National University Dental Hospital, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Seong-Kyun Kim
- Department of Prosthodontics & Dental Research Institute, Seoul National University Dental Hospital, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Seong-Joo Heo
- Department of Prosthodontics & Dental Research Institute, Seoul National University Dental Hospital, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Jai-Young Koak
- Department of Prosthodontics & Dental Research Institute, Seoul National University Dental Hospital, School of Dentistry, Seoul National University, Seoul, Republic of Korea
| | - Joo-Hee Lee
- Department of Prosthodontics, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, Republic of Korea
| |
Collapse
|
67
|
Gao F, Li Q, Hou L, Li Z, Min F, Liu Z. Mesenchymal stem cell-based angiotensin-converting enzyme 2 in treatment of acute lung injury rat induced by bleomycin. Exp Lung Res 2014; 40:392-403. [DOI: 10.3109/01902148.2014.938200] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
68
|
|
69
|
Wang C, Zhu H, Sun Z, Xiang Z, Ge Y, Ni C, Luo Z, Qian W, Han X. Inhibition of Wnt/β-catenin signaling promotes epithelial differentiation of mesenchymal stem cells and repairs bleomycin-induced lung injury. Am J Physiol Cell Physiol 2014; 307:C234-44. [PMID: 24898581 DOI: 10.1152/ajpcell.00366.2013] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Idiopathic pulmonary fibrosis is a progressive lung disorder of unknown etiology. Previous studies have shown that aberrant activation of the Wnt/β-catenin signaling cascade occurs in lungs of patients with idiopathic pulmonary fibrosis. Given the important roles of the Wnt/β-catenin signaling pathway in the development of pulmonary fibrosis, we targeted this pathway for the intervention of pulmonary fibrosis with XAV939, a small molecule that specifically inhibits Tankyrase 1/2, eventually leading to the degradation of β-catenin and suppression of the Wnt/β-catenin signaling pathway. Our results demonstrated that XAV939 significantly inhibited the activation of Wnt/β-catenin signaling and attenuated bleomycin-induced lung fibrosis in mice, and thus improved the survival of mice with lung injury. Interestingly, previous investigations have confirmed that endogenous and exogenous mesenchymal stem cells could be recruited to the injured lung, although the exact effects of these cells are debatable. To determine the effect of Wnt/β-catenin signaling in the epithelial differentiation of bone marrow-derived mesenchymal stem cells (BM-MSCs), we established a coculture system that contains BM-MSCs and alveolar type II epithelial cells. The in vitro experiments demonstrated that XAV939 could promote the differentiation of BM-MSCs into an epithelium-like phenotype in the coculture system. We also found that XAV939 could inhibit the proliferation and myofibroblast differentiation of NIH/3T3 fibroblasts. This work supports that inhibition of the Wnt/β-catenin signaling pathway may be exploited for the treatment of idiopathic pulmonary fibrosis for which effective treatment strategies are still lacking.
Collapse
Affiliation(s)
- Cong Wang
- Immunology and Reproductive Biology Laboratory, Medical School, Nanjing University, Nanjing, Jiangsu, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, China; State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu, China
| | - Huiming Zhu
- Immunology and Reproductive Biology Laboratory, Medical School, Nanjing University, Nanjing, Jiangsu, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, China; State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu, China
| | - Zhaorui Sun
- Department of Emergency, Jinling Hospital, Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Zou Xiang
- Department of Microbiology and Immunology, Mucosal Immunobiology and Vaccine Research Center, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden; and
| | - Yuanyuan Ge
- Immunology and Reproductive Biology Laboratory, Medical School, Nanjing University, Nanjing, Jiangsu, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, China; State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu, China
| | - Can Ni
- Immunology and Reproductive Biology Laboratory, Medical School, Nanjing University, Nanjing, Jiangsu, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, China; State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu, China
| | - Zhaowen Luo
- Immunology and Reproductive Biology Laboratory, Medical School, Nanjing University, Nanjing, Jiangsu, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, China; State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu, China
| | - Weiping Qian
- State Key Laboratory of Bioelectronics, Southeast University, Nanjing, China
| | - Xiaodong Han
- Immunology and Reproductive Biology Laboratory, Medical School, Nanjing University, Nanjing, Jiangsu, China; Jiangsu Key Laboratory of Molecular Medicine, Nanjing University, Nanjing, Jiangsu, China; State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing, Jiangsu, China;
| |
Collapse
|
70
|
Abstract
Ocular surface defects represent one of the most common causes of impaired vision or even blindness. For treatment, keratoplasty represents the first choice. However, if corneal defects are more extensive and associated with a limbal stem cell (LSC) deficiency, corneal transplantation is not a sufficient therapeutic procedure and only viable approach to treatment is the transplantation of LSCs. When the LSC deficiency is a bilateral disorder, autologous LSCs are not available. The use of allogeneic LSCs requires strong immunosuppression, which leads to side-effects, and the treatment is not always effective. The alternative and perspective approach to the treatment of severe ocular surface injuries and LSC deficiency is offered by the transplantation of autologous mesenchymal stem cells (MSCs). These cells can be obtained from the bone marrow or adipose tissue of the particular patient, grow well in vitro and can be transferred, using an appropriate scaffold, onto the damaged ocular surface. Here they exert beneficial effects by possible direct differentiation into corneal epithelial cells, by immunomodulatory effects and by the production of numerous trophic and growth factors. Recent experiments utilizing the therapeutic properties of MSCs in animal models with a mechanically or chemically injured ocular surface have yielded promising results and demonstrated significant corneal regeneration, improved corneal transparency and a rapid healing process associated with the restoration of vision. The use of autologous MSCs thus represents a promising therapeutic approach and offers hope for patients with severe ocular surface injuries and LSC deficiency.
Collapse
|
71
|
Mendez JJ, Ghaedi M, Steinbacher D, Niklason LE. Epithelial cell differentiation of human mesenchymal stromal cells in decellularized lung scaffolds. Tissue Eng Part A 2014; 20:1735-46. [PMID: 24393055 DOI: 10.1089/ten.tea.2013.0647] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Identification of appropriate donor cell types is important for lung cell therapy and for lung regeneration. Previous studies have indicated that mesenchymal stromal cells derived from human bone marrow (hBM-MSCs) and from human adipose tissue (hAT-MSCs) may have the ability to trans-differentiate into lung epithelial cells. However, these data remain controversial. Herein, the ability of hBM-MSCs and hAT-MSCs to repopulate acellular rodent lung tissue was evaluated. hBM-MSCs and hAT-MSCs were isolated from bone marrow aspirate and lipoaspirate, respectively. Rat lungs were decellularized with CHAPS detergent, followed by seeding the matrix with hBM-MSCs and hAT-MSCs. Under appropriate culture conditions, both human MSC populations attached to and proliferated within the lung tissue scaffold. In addition, cells were capable of type 2 pneumocyte differentiation, as assessed by marker expression of surfactant protein C (pro-SPC) at the protein and the RNA level, and by the presence of lamellar bodies by transmission electron microscopy. Additionally, hAT-MSCs contributed to Clara-like cells that lined the airways in the lung scaffolds, whereas the hBM-MSCs did not. We also tested the differentiation potential of MSCs on different extracellular matrix components in vitro, and found that protein substrate influences MSC epithelial differentiation. Together our data show the capacity for human MSCs to differentiate toward lung epithelial phenotypes, and the possibility of using these cells for lung cell therapies and tissue engineering.
Collapse
Affiliation(s)
- Julio J Mendez
- 1 Department of Anesthesiology and Biomedical Engineering, Yale University , New Haven, Connecticut
| | | | | | | |
Collapse
|
72
|
Gao P, Zhou Y, Xian L, Li C, Xu T, Plunkett B, Huang SK, Wan M, Cao X. Functional effects of TGF-β1 on mesenchymal stem cell mobilization in cockroach allergen-induced asthma. THE JOURNAL OF IMMUNOLOGY 2014; 192:4560-4570. [PMID: 24711618 DOI: 10.4049/jimmunol.1303461] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Mesenchymal stem cells (MSCs) have been suggested to participate in immune regulation and airway repair/remodeling. TGF-β1 is critical in the recruitment of stem/progenitor cells for tissue repair, remodeling, and cell differentiation. In this study, we sought to investigate the role of TGF-β1 in MSC migration in allergic asthma. We examined nestin expression (a marker for MSCs) and TGF-β1 signaling activation in airways in cockroach allergen extract (CRE)-induced mouse models. Compared with control mice, there were increased nestin(+) cells in airways and higher levels of active TGF-β1 in serum and p-Smad2/3 expression in lungs of CRE-treated mice. Increased activation of TGF-β1 signaling was also found in CRE-treated MSCs. We then assessed MSC migration induced by conditioned medium from CRE-challenged human epithelium in air/liquid interface culture in Transwell assays. MSC migration was stimulated by epithelial-conditioned medium, but was significantly inhibited by either TGF-β1-neutralizing Ab or TβR1 inhibitor. Intriguingly, increased migration of MSCs from blood and bone marrow to the airway was also observed after systemic injection of GFP(+) MSCs and from bone marrow of Nes-GFP mice following CRE challenge. Furthermore, TGF-β1-neutralizing Ab inhibited the CRE-induced MSC recruitment, but promoted airway inflammation. Finally, we investigated the role of MSCs in modulating CRE-induced T cell response and found that MSCs significantly inhibited CRE-induced inflammatory cytokine secretion (IL-4, IL-13, IL-17, and IFN-γ) by CD4(+) T cells. These results suggest that TGF-β1 may be a key promigratory factor in recruiting MSCs to the airways in mouse models of asthma.
Collapse
Affiliation(s)
- Peisong Gao
- Johns Hopkins Asthma and Allergy Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yufeng Zhou
- Johns Hopkins Asthma and Allergy Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lingling Xian
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Changjun Li
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ting Xu
- Johns Hopkins Asthma and Allergy Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Beverly Plunkett
- Johns Hopkins Asthma and Allergy Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shau-Ku Huang
- Johns Hopkins Asthma and Allergy Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,National Health Research Institutes, Taiwan
| | - Mei Wan
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Xu Cao
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
73
|
Sabin K, Kikyo N. Microvesicles as mediators of tissue regeneration. Transl Res 2014; 163:286-95. [PMID: 24231336 PMCID: PMC3976717 DOI: 10.1016/j.trsl.2013.10.005] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Revised: 10/19/2013] [Accepted: 10/21/2013] [Indexed: 12/20/2022]
Abstract
The use of stem cells in the treatment of various diseases and injuries has received increasing interest during the past decade. Injected stem cells, such as mesenchymal stem cells, stimulate tissue repair largely through the secretion of soluble factors that regulate various processes of tissue regeneration, including inflammatory responses, apoptosis, host cell proliferation, and angiogenesis. Recently, it has become apparent that stem cells also use membranous small vesicles, collectively called microvesicles, to repair damaged tissues. Microvesicles are released by many types of cells and exist in almost all types of body fluids. They serve as a vehicle to transfer protein, messenger RNA, and micro RNA to distant cells, altering the gene expression, proliferation, and differentiation of the recipient cells. Although animal models and in vitro studies have suggested promising applications for microvesicles-based regeneration therapy, its effectiveness and feasibility in clinical medicine remain to be established. Further studies of the basic mechanisms responsible for microvesicle-mediated tissue regeneration could lead to novel approaches in regenerative medicine.
Collapse
Affiliation(s)
- Keith Sabin
- Stem Cell Institute, Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minn
| | - Nobuaki Kikyo
- Stem Cell Institute, Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minn.
| |
Collapse
|
74
|
Goolaerts A, Pellan-Randrianarison N, Larghero J, Vanneaux V, Uzunhan Y, Gille T, Dard N, Planès C, Matthay MA, Clerici C. Conditioned media from mesenchymal stromal cells restore sodium transport and preserve epithelial permeability in an in vitro model of acute alveolar injury. Am J Physiol Lung Cell Mol Physiol 2014; 306:L975-85. [PMID: 24682451 DOI: 10.1152/ajplung.00242.2013] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stromal cells (MSCs) or their media (MSC-M) were reported to reverse acute lung injury (ALI)-induced decrease of alveolar fluid clearance. To determine the mechanisms by which MSC-M exert their beneficial effects, an in vitro model of alveolar epithelial injury was created by exposing primary rat alveolar epithelial cells (AECs) to hypoxia (3% O2) plus cytomix, a combination of IL-1β, TNF-α, and IFN-γ. MSC-M were collected from human MSCs exposed for 12 h to either normoxia (MSC-M) or to hypoxia plus cytomix (HCYT-MSC-M). This latter condition was used to model the effect of alveolar inflammation and hypoxia on paracrine secretion of MSCs in the injured lung. Comparison of paracrine soluble factors in MSC media showed that the IL-1 receptor antagonist and prostaglandin E2 were markedly increased while keratinocyte growth factor (KGF) was twofold lower in HCYT-MSC-M compared with MSC-M. In AECs, hypoxia plus cytomix increased protein permeability, reduced amiloride-sensitive short-circuit current (AS-Isc), and also decreased the number of α-epithelial sodium channel (α-ENaC) subunits in the apical membrane. To test the effects of MSC media, MSC-M and HCYT-MSC-M were added for an additional 12 h to AECs exposed to hypoxia plus cytomix. MSC-M and HCYT-MSC-M completely restored epithelial permeability to normal. MSC-M, but not HCYT-MSC-M, significantly prevented the hypoxia plus cytomix-induced decrease of ENaC activity and restored apical α-ENaC channels. Interestingly, KGF-deprived MSC-M were unable to restore amiloride-sensitive sodium transport, indicating a possible role for KGF in the beneficial effect of MSC-M. These results indicate that MSC-M may be a preferable therapeutic option for ALI.
Collapse
Affiliation(s)
- Arnaud Goolaerts
- Institut National de la Santé et de la Recherche Médicale, U773, Paris, France
| | - Nadia Pellan-Randrianarison
- Institut National de la Santé et de la Recherche Médicale, U773, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Unité Mixte de Recherche 773 and Unité Mixte de Recherche 940, Paris, France
| | - Jérôme Larghero
- Université Paris Diderot, Sorbonne Paris Cité, Unité Mixte de Recherche 773 and Unité Mixte de Recherche 940, Paris, France; AP-HP, Hôpital Saint Louis, Unité de Thérapie Cellulaire et CIC de Biothérapies, Paris, France; University Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Valérie Vanneaux
- Université Paris Diderot, Sorbonne Paris Cité, Unité Mixte de Recherche 773 and Unité Mixte de Recherche 940, Paris, France; AP-HP, Hôpital Saint Louis, Unité de Thérapie Cellulaire et CIC de Biothérapies, Paris, France; University Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Yurdagül Uzunhan
- Université Paris 13, Sorbonne Paris Cité, EA2363, Bobigny, France; AP-HP, Hôpital Avicenne, Bobigny, France; and
| | - Thomas Gille
- Université Paris 13, Sorbonne Paris Cité, EA2363, Bobigny, France; AP-HP, Hôpital Avicenne, Bobigny, France; and
| | - Nicolas Dard
- Université Paris 13, Sorbonne Paris Cité, EA2363, Bobigny, France
| | - Carole Planès
- Université Paris 13, Sorbonne Paris Cité, EA2363, Bobigny, France; AP-HP, Hôpital Avicenne, Bobigny, France; and
| | - Michael A Matthay
- Institut National de la Santé et de la Recherche Médicale, U773, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Unité Mixte de Recherche 773 and Unité Mixte de Recherche 940, Paris, France
| | - Christine Clerici
- Institut National de la Santé et de la Recherche Médicale, U773, Paris, France; Université Paris Diderot, Sorbonne Paris Cité, Unité Mixte de Recherche 773 and Unité Mixte de Recherche 940, Paris, France; AP-HP, Hôpital Bichat, Paris, France
| |
Collapse
|
75
|
Yang WJ, Li SH, Weisel RD, Liu SM, Li RK. Cell fusion contributes to the rescue of apoptotic cardiomyocytes by bone marrow cells. J Cell Mol Med 2014; 16:3085-95. [PMID: 22805279 PMCID: PMC4393736 DOI: 10.1111/j.1582-4934.2012.01600.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 07/09/2012] [Indexed: 12/14/2022] Open
Abstract
Cardiomyocyte apoptosis is an important contributor to the progressive cardiac dysfunction that culminates in congestive heart failure. Bone marrow cells (BMCs) restore cardiac function following ischaemia, and transplanted BMCs have been reported to fuse with cells of diverse tissues. We previously demonstrated that the myogenic conversion of bone marrow stromal cells increased nearly twofold when the cells were co-cultured with apoptotic (TNF-α treated) cardiomyocytes. We therefore hypothesized that cell fusion may be a major mechanism by which BMCs rescue cardiomyocytes from apoptosis. We induced cellular apoptosis in neonatal rat cardiomyocytes by treatment with hydrogen peroxide (H2O2). The TUNEL assay demonstrated an increase in apoptosis from 4.5 ± 1.3% in non-treated cells to 19.0 ± 4.4% (P < 0.05) in treated cells. We subsequently co-cultured the apoptotic cardiomyocytes with BMCs and assessed cell fusion using flow cytometry. Fusion was rare in the non-treated control cardiomyocytes (0.3%), whereas H2O2 treatment led to significantly higher fusion rates than the control group (P < 0.05), with the highest rate of 7.9 ± 0.3% occurring at 25 μM H2O2. We found an inverse correlation between cell fusion and completion of cardiomyocyte apoptosis (R2 = 0.9863). An in vivo mouse model provided evidence of cell fusion in the infarcted myocardium following the injection of BMCs. The percentage of cells undergoing fusion was significantly higher in mice injected with BMCs following infarction (8.8 ± 1.3%) compared to mice that did not undergo infarction (4.6 ± 0.6%, P < 0.05). Enhancing cell fusion may be one method to preserve cardiomyocytes following myocardial infarction, and this new approach may provide a novel target for cardiac regenerative therapies.
Collapse
Affiliation(s)
- Wei-Jian Yang
- Department of Cardiology, Second Affiliated Hospital of Guangzhou Medical College, Guangzhou, China
| | | | | | | | | |
Collapse
|
76
|
Gong X, Sun Z, Cui D, Xu X, Zhu H, Wang L, Qian W, Han X. Isolation and characterization of lung resident mesenchymal stem cells capable of differentiating into alveolar epithelial type II cells. Cell Biol Int 2014; 38:405-11. [DOI: 10.1002/cbin.10240] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Xuemin Gong
- Immunology and Reproductive Biology Laboratory; Medical College of Nanjing University; Nanjing 210093 China
- Jiangsu Key Laboratory of Molecular Medicine; Nanjing 210093 China
- State Key Laboratory of Analytical Chemistry for Life Science; Nanjing University; Nanjing 210093 China
| | - Zhaorui Sun
- Immunology and Reproductive Biology Laboratory; Medical College of Nanjing University; Nanjing 210093 China
- Jiangsu Key Laboratory of Molecular Medicine; Nanjing 210093 China
- State Key Laboratory of Analytical Chemistry for Life Science; Nanjing University; Nanjing 210093 China
| | - Di Cui
- Immunology and Reproductive Biology Laboratory; Medical College of Nanjing University; Nanjing 210093 China
- Jiangsu Key Laboratory of Molecular Medicine; Nanjing 210093 China
- State Key Laboratory of Analytical Chemistry for Life Science; Nanjing University; Nanjing 210093 China
| | - Xiaomeng Xu
- Immunology and Reproductive Biology Laboratory; Medical College of Nanjing University; Nanjing 210093 China
- Jiangsu Key Laboratory of Molecular Medicine; Nanjing 210093 China
- State Key Laboratory of Analytical Chemistry for Life Science; Nanjing University; Nanjing 210093 China
| | - Huiming Zhu
- Immunology and Reproductive Biology Laboratory; Medical College of Nanjing University; Nanjing 210093 China
- Jiangsu Key Laboratory of Molecular Medicine; Nanjing 210093 China
- State Key Laboratory of Analytical Chemistry for Life Science; Nanjing University; Nanjing 210093 China
| | - Lihui Wang
- Immunology and Reproductive Biology Laboratory; Medical College of Nanjing University; Nanjing 210093 China
- Jiangsu Key Laboratory of Molecular Medicine; Nanjing 210093 China
- State Key Laboratory of Analytical Chemistry for Life Science; Nanjing University; Nanjing 210093 China
| | - Weiping Qian
- State Key Laboratory of Bioelectronics; Southeast University; Nanjing 210093 China
| | - Xiaodong Han
- Immunology and Reproductive Biology Laboratory; Medical College of Nanjing University; Nanjing 210093 China
- Jiangsu Key Laboratory of Molecular Medicine; Nanjing 210093 China
- State Key Laboratory of Analytical Chemistry for Life Science; Nanjing University; Nanjing 210093 China
| |
Collapse
|
77
|
Effects of bone marrow or mesenchymal stem cell transplantation on oral mucositis (mouse) induced by fractionated irradiation. Strahlenther Onkol 2014; 190:399-404. [DOI: 10.1007/s00066-013-0510-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Accepted: 11/08/2013] [Indexed: 10/25/2022]
|
78
|
Hematopoietic and mesenchymal stem cells for the treatment of chronic respiratory diseases: role of plasticity and heterogeneity. ScientificWorldJournal 2014; 2014:859817. [PMID: 24563632 PMCID: PMC3916026 DOI: 10.1155/2014/859817] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 10/20/2013] [Indexed: 12/21/2022] Open
Abstract
Chronic lung diseases, such as cystic fibrosis (CF), asthma, and chronic obstructive pulmonary disease (COPD) are incurable and represent a very high social burden. Stem cell-based treatment may represent a hope for the cure of these diseases. In this paper, we revise the overall knowledge about the plasticity and engraftment of exogenous marrow-derived stem cells into the lung, as well as their usefulness in lung repair and therapy of chronic lung diseases. The lung is easily accessible and the pathophysiology of these diseases is characterized by injury, inflammation, and eventually by remodeling of the airways. Bone marrow-derived stem cells, including hematopoietic stem/progenitor cells (HSPCs) and mesenchymal stromal (stem) cells (MSCs), encompass a wide array of cell subsets with different capacities of engraftment and injured tissue regenerating potential. Proof-of-principle that marrow cells administered locally may engraft and give rise to specialized epithelial cells has been given, but the efficiency of this conversion is too limited to give a therapeutic effect. Besides the identification of plasticity mechanisms, the characterization/isolation of the stem cell subpopulations represents a major challenge to improving the efficacy of transplantation protocols used in regenerative medicine for lung diseases.
Collapse
|
79
|
Bertoncello I, McQualter JL. Endogenous lung stem cells: what is their potential for use in regenerative medicine? Expert Rev Respir Med 2014; 4:349-62. [DOI: 10.1586/ers.10.21] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
80
|
Li J, Huang S, Wu Y, Gu C, Gao D, Feng C, Wu X, Fu X. Paracrine factors from mesenchymal stem cells: a proposed therapeutic tool for acute lung injury and acute respiratory distress syndrome. Int Wound J 2013; 11:114-21. [PMID: 24373614 DOI: 10.1111/iwj.12202] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 10/24/2013] [Accepted: 11/20/2013] [Indexed: 12/22/2022] Open
Abstract
Despite extensive researches in acute lung injury (ALI) and acute respiratory distress syndrome (ARDS), current pharmacological therapies and respiratory support are still the main methods to treat patients with ALI and ARDS and the effects remain limited. Hence, innovative therapies are needed to decrease the morbidity and mortality. Because of the proven therapeutic effects in other fields, mesenchymal stem cells (MSCs) might be considered as a promising alternative to treat ALI and ARDS. Numerous documents demonstrate that MSCs can exert multiple functions, such as engraftment, differentiation and immunoregulation, but now the key researches are concentrated on paracrine factors secreted by MSCs that can mediate endothelial and epithelial permeability, increase alveolar fluid clearance and other potential mechanisms. This review aimed to review the current researches in terms of the effects of MSCs on ALI and ARDS and to analyse these paracrine factors, as well as to predict the potential directions and challenges of the application in this field.
Collapse
Affiliation(s)
- Jiwei Li
- Department of Thoracic and Cardiovascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, P. R. China; Key Laboratory of Wound Repair and Regeneration of PLA, The First Affiliated Hospital, General Hospital of PLA, Trauma Center of Postgraduate Medical College, Beijing, P. R. China
| | | | | | | | | | | | | | | |
Collapse
|
81
|
Sun Z, Gong X, Zhu H, Wang C, Xu X, Cui D, Qian W, Han X. Inhibition of Wnt/β-Catenin Signaling Promotes Engraftment of Mesenchymal Stem Cells to Repair Lung Injury. J Cell Physiol 2013; 229:213-24. [DOI: 10.1002/jcp.24436] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Accepted: 07/15/2013] [Indexed: 11/05/2022]
Affiliation(s)
- Zhaorui Sun
- Immunology and Reproductive Biology Laboratory; Medical College of Nanjing University; Nanjing PR China
- Jiangsu Key Laboratory of Molecular Medicine; Nanjing PR China
- State Key Laboratory of Analytical Chemistry for Life Science; Nanjing University; Nanjing PR China
| | - Xuemin Gong
- Immunology and Reproductive Biology Laboratory; Medical College of Nanjing University; Nanjing PR China
- Jiangsu Key Laboratory of Molecular Medicine; Nanjing PR China
- State Key Laboratory of Analytical Chemistry for Life Science; Nanjing University; Nanjing PR China
| | - Huiming Zhu
- Immunology and Reproductive Biology Laboratory; Medical College of Nanjing University; Nanjing PR China
- Jiangsu Key Laboratory of Molecular Medicine; Nanjing PR China
- State Key Laboratory of Analytical Chemistry for Life Science; Nanjing University; Nanjing PR China
| | - Cong Wang
- Immunology and Reproductive Biology Laboratory; Medical College of Nanjing University; Nanjing PR China
- Jiangsu Key Laboratory of Molecular Medicine; Nanjing PR China
- State Key Laboratory of Analytical Chemistry for Life Science; Nanjing University; Nanjing PR China
| | - Xiaomeng Xu
- Immunology and Reproductive Biology Laboratory; Medical College of Nanjing University; Nanjing PR China
- Jiangsu Key Laboratory of Molecular Medicine; Nanjing PR China
- State Key Laboratory of Analytical Chemistry for Life Science; Nanjing University; Nanjing PR China
| | - Di Cui
- Immunology and Reproductive Biology Laboratory; Medical College of Nanjing University; Nanjing PR China
- Jiangsu Key Laboratory of Molecular Medicine; Nanjing PR China
- State Key Laboratory of Analytical Chemistry for Life Science; Nanjing University; Nanjing PR China
| | - Weiping Qian
- State Key Laboratory of Bioelectronics; Southeast University; Nanjing PR China
| | - Xiaodong Han
- Immunology and Reproductive Biology Laboratory; Medical College of Nanjing University; Nanjing PR China
- Jiangsu Key Laboratory of Molecular Medicine; Nanjing PR China
- State Key Laboratory of Analytical Chemistry for Life Science; Nanjing University; Nanjing PR China
| |
Collapse
|
82
|
N-acetylcysteine-pretreated human embryonic mesenchymal stem cell administration protects against bleomycin-induced lung injury. Am J Med Sci 2013; 346:113-22. [PMID: 23085672 DOI: 10.1097/maj.0b013e318266e8d8] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
INTRODUCTION The transplantation of mesenchymal stem cells (MSCs) has been reported to be a promising approach in the treatment of acute lung injury. However, the poor efficacy of transplanted MSCs is one of the serious handicaps in the progress of MSC-based therapy. Therefore, the purpose of this study was to investigate whether the pretreatment of human embryonic MSCs (hMSCs) with an antioxidant, namely N-acetylcysteine (NAC), can improve the efficacy of hMSC transplantation in lung injury. METHODS In vitro, the antioxidant capacity of NAC-pretreated hMSCs was assessed using intracellular reactive oxygen species (ROS) and glutathione assays and cell adhesion and spreading assays. In vivo, the therapeutic potential of NAC-pretreated hMSCs was assessed in a bleomycin-induced model of lung injury in nude mice. RESULTS The pretreatment of hMSCs with NAC improved antioxidant capacity to defend against redox imbalances through the elimination of cellular ROS, increasing cellular glutathione levels, and the enhancement of cell adhesion and spreading when exposed to oxidative stresses in vitro. In addition, the administration of NAC-pretreated hMSCs to nude mice with bleomycin-induced lung injury decreased the pathological grade of lung inflammation and fibrosis, hydroxyproline content and numbers of neutrophils and inflammatory cytokines in bronchoalveolar lavage fluid and apoptotic cells, while enhancing the retention and proliferation of hMSCs in injured lung tissue and improving the survival rate of mice compared with results from untreated hMSCs. CONCLUSIONS The pretreatment of hMSCs with NAC could be a promising therapeutic approach to improving cell transplantation and, therefore, the treatment of lung injury.
Collapse
|
83
|
Cell therapy with bone marrow mononuclear cells in elastase-induced pulmonary emphysema. Stem Cell Rev Rep 2013; 9:210-8. [PMID: 23242964 DOI: 10.1007/s12015-012-9419-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Emphysema is characterized by destruction of alveolar walls with loss of gas exchange surface and consequent progressive dyspnea. This study aimed to evaluate the efficiency of cell therapy with bone marrow mononuclear cells (BMMC) in an animal model of elastase-induced pulmonary emphysema. Emphysema was induced in C57Bl/J6 female mice by intranasal instillation of elastase. After 21 days, the mice received bone marrow mononuclear cells from EGFP male mice with C57Bl/J6 background. The groups were assessed by comparison and statistically significant differences (p < 0.05) were observed among the groups treated with BMMC and evaluated after 7, 14 and 21 days. Analysis of the mean linear intercept (Lm) values for the different groups allowed to observe that the group treated with BMMC and evaluated after 21 days showed the most significant result. The group that received no treatment showed a statistically significant difference when compared to other groups, except the group treated and evaluated after 21 days, evidencing the efficacy of cell therapy with BMMC in pulmonary emphysema.
Collapse
|
84
|
Toonkel RL, Hare JM, Matthay MA, Glassberg MK. Mesenchymal Stem Cells and Idiopathic Pulmonary Fibrosis. Potential for Clinical Testing. Am J Respir Crit Care Med 2013; 188:133-40. [DOI: 10.1164/rccm.201207-1204pp] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
85
|
Mesenchymal stromal cells augment CD4+ and CD8+ T-cell proliferation through a CCL2 pathway. Cytotherapy 2013; 15:1195-207. [PMID: 23845188 DOI: 10.1016/j.jcyt.2013.05.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 04/10/2013] [Accepted: 05/10/2013] [Indexed: 12/22/2022]
Abstract
BACKGROUND AIMS Mesenchymal stromal cells (MSC) derived from bone marrow are immunosuppressive in vitro and in vivo. Recent evidence, however, has shown that in certain settings, MSC can also be immunostimulatory. The mechanisms involved in this process are largely unknown. METHODS Mouse spleen T cells were stimulated with allogeneic mixed lymphocyte reaction (MLR) or anti-CD3/CD28 beads and treated with autologous bone marrow MSC or MSC-conditioned medium. CD4+ and CD8+ T-cell proliferation was analyzed after treatment. RESULTS We show that MSC have both suppressive and stimulatory functions toward T cells after stimulation with anti-CD3/CD28 beads or in an MLR. This depended on the ratio of MSC to responder T cells, with low numbers of MSC increasing and higher numbers inhibiting T-cell proliferation. Immunostimulatory function was mediated, in part, by soluble factors. MSC immunosuppression of the MLR was indirect and related to inhibition of antigen-presenting cell maturation. Direct effects of MSC-conditioned medium during anti-CD3/CD28 stimulated proliferation were entirely stimulatory and required the presence of the T-cell receptor. MSC supernatant contained both CCL2 and CCL5 at high levels, but only CCL2 level correlated with the ability to augment proliferation. An anti-CCL2 antibody blocked this proliferative activity. CONCLUSIONS CCL2 plays an important role in the immunostimulatory function of MSC, and we further hypothesize that the immunomodulatory role of MSC is determined by a balance between inhibitory and stimulatory factors, suggesting the need for caution when these cells are investigated in clinical protocols.
Collapse
|
86
|
Xu F, Hu Y, Zhou J, Wang X. Mesenchymal stem cells in acute lung injury: are they ready for translational medicine? J Cell Mol Med 2013; 17:927-35. [PMID: 23834470 PMCID: PMC3780529 DOI: 10.1111/jcmm.12063] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 03/11/2013] [Indexed: 12/12/2022] Open
Abstract
Acute lung injury (ALI) is a severe clinical condition responsible for high mortality and the development of multiple organ dysfunctions, because of the lack of specific and effective therapies for ALI. Increasing evidence from pre-clinical studies supports preventive and therapeutic effects of mesenchymal stem cells (MSCs, also called mesenchymal stromal cells) in ALI/ARDS (acute respiratory distress syndrome). Therapeutic effects of MSCs were noticed in various delivery approaches (systemic, local, or other locations), multiple origins (bone marrow or other tissues), or different schedules of administrations (before or after the challenges). MSCs could reduce the over-production of inflammatory mediators, leucocyte infiltration, tissue injury and pulmonary failure, and produce a number of benefit factors through interaction with other cells in the process of lung tissue repair. Thus, it is necessary to establish guidelines, standard operating procedures and evaluation criteria for translating MSC-based therapies into clinical application for patients with ALI.
Collapse
Affiliation(s)
- Feng Xu
- Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | | | | | | |
Collapse
|
87
|
Hashmani K, Branch MJ, Sidney LE, Dhillon PS, Verma M, McIntosh OD, Hopkinson A, Dua HS. Characterization of corneal stromal stem cells with the potential for epithelial transdifferentiation. Stem Cell Res Ther 2013; 4:75. [PMID: 23800436 PMCID: PMC4058700 DOI: 10.1186/scrt226] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Accepted: 06/04/2013] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION The corneal stroma is being increasingly recognized as a repository for stem cells. Like the limbal and endothelial niches, stromal stem cells often reside in the peripheral cornea and limbus. These peripheral and limbal corneal stromal cells (PLCSCs) are known to produce mesenchymal stem cells in vitro. Recently, a common corneal stromal and epithelial progenitor was hinted at. This study aims to examine the stem cell potential of corneal stromal cells and to investigate their epithelial transdifferentiation ability. METHODS PLCSCs were grown in traditional Dulbecco modified Eagle medium (DMEM)-based keratocyte culture medium and an M199-based medium and analyzed for a profile of cell-surface markers by using flow cytometry and differentiated into mesenchymal phenotypes analyzed with quantitative polymerase chain reaction (qPCR) and histologic staining. PLCSCs in M199 were subsequently divided into subpopulations based on CD34 and CD105 expression by using fluorescence- activated cell sorting (FACS). Subpopulations were characterized by marker profile and mesenchymal differentiation ability. Both whole PLCSCs and subpopulations were also cultured for epithelial transdifferentiation. RESULTS Cells cultured in M199 demonstrated a more stem-like cell-surface marker profile, and the keratocyte marker CD34 was retained for several passages but absent in cells cultured in DMEM. Cells cultured in M199 also exhibited a greater mesenchymal differentiation potential, compared with DMEM. PLCSCs could be divided into CD34(+)CD105(+), CD34-CD105(+), and CD34-CD105- subpopulations, of which CD34(+)CD105(+) cells were the most stemlike with regard to marker expression and mesenchymal differentiation potential. Subpopulations of PLCSCs exhibited differing abilities to transdifferentiate into epithelial phenotypes. Cells that were initially CD34(+)CD105(+) showed the greatest differentiation potential, producing CK3(+) and CK19(+) cells, and expressed a range of both epithelial progenitor (HES1, FRZB1, DCT, SOD2, ABCG2, CDH1, KRT19) and terminally differentiated (DSG3, KRT3, KRT12, KRT24) genes. CONCLUSIONS Culture medium has a significant effect on the phenotype and differentiation capacity of PLCSCs. The stroma contains a heterogeneous cell population in which we have identified CD34(+) cells as a stem cell population with a capacity for mesenchymal and epithelial differentiation.
Collapse
|
88
|
Ramachandran S, Suguihara C, Drummond S, Chatzistergos K, Klim J, Torres E, Huang J, Hehre D, Rodrigues CO, McNiece IK, Hare JM, Young KC. Bone marrow-derived c-kit+ cells attenuate neonatal hyperoxia-induced lung injury. Cell Transplant 2013; 24:85-95. [PMID: 23759597 DOI: 10.3727/096368913x667736] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Recent studies suggest that bone marrow (BM)-derived stem cells have therapeutic efficacy in neonatal hyperoxia-induced lung injury (HILI). c-kit, a tyrosine kinase receptor that regulates angiogenesis, is expressed on several populations of BM-derived cells. Preterm infants exposed to hyperoxia have decreased lung angiogenesis. Here we tested the hypothesis that administration of BM-derived c-kit(+) cells would improve angiogenesis in neonatal rats with HILI. To determine whether intratracheal (IT) administration of BM-derived c-kit(+) cells attenuates neonatal HILI, rat pups exposed to either normobaric normoxia (21% O2) or hyperoxia (90% O2) from postnatal day (P) 2 to P15 were randomly assigned to receive either IT BM-derived green fluorescent protein (GFP)(+) c-kit(-) cells (PL) or BM-derived GFP(+) c-kit(+) cells on P8. The effect of cell therapy on lung angiogenesis, alveolarization, pulmonary hypertension, vascular remodeling, cell proliferation, and apoptosis was determined at P15. Cell engraftment was determined by GFP immunostaining. Compared to PL, the IT administration of BM-derived c-kit(+) cells to neonatal rodents with HILI improved alveolarization as evidenced by increased lung septation and decreased mean linear intercept. This was accompanied by an increase in lung vascular density, a decrease in lung apoptosis, and an increase in the secretion of proangiogenic factors. There was no difference in pulmonary vascular remodeling or the degree of pulmonary hypertension. Confocal microscopy demonstrated that 1% of total lung cells were GFP(+) cells. IT administration of BM-derived c-kit(+) cells improves lung alveolarization and angiogenesis in neonatal HILI, and this may be secondary to an improvement in the lung angiogenic milieu.
Collapse
Affiliation(s)
- Shalini Ramachandran
- Department of Pediatrics/Division of Neonatology, University of Miami Miller School of Medicine, Miami, FL, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
89
|
Effects of intratracheal mesenchymal stromal cell therapy during recovery and resolution after ventilator-induced lung injury. Anesthesiology 2013; 118:924-32. [PMID: 23377221 DOI: 10.1097/aln.0b013e318287ba08] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs) have been demonstrated to attenuate acute lung injury when delivered by intravenous or intratracheal routes. The authors aimed to determine the efficacy of and mechanism of action of intratracheal MSC therapy and to compare their efficacy in enhancing lung repair after ventilation-induced lung injury with intravenous MSC therapy. METHODS : After induction of anesthesia, rats were orotracheally intubated and subjected to ventilation-induced lung injury (respiratory rate 18(-1) min, P insp 35 cm H2O,) to produce severe lung injury. After recovery, animals were randomized to receive: (1) no therapy, n = 4; (2) intratracheal vehicle (phosphate-buffered saline, 300 µl, n = 8); (3) intratracheal fibroblasts (4 × 10 cells, n = 8); (4) intratracheal MSCs (4 × 10(6) cells, n = 8); (5) intratracheal conditioned medium (300 µl, n = 8); or (6) intravenous MSCs (4 × 10(6) cells, n = 4). The extent of recovery after acute lung injury and the inflammatory response was assessed after 48 h. RESULTS Intratracheal MSC therapy enhanced repair after ventilation-induced lung injury, improving arterial oxygenation (mean ± SD, 146 ± 3.9 vs. 110.8 ± 21.5 mmHg), restoring lung compliance (1.04 ± 0.11 vs. 0.83 ± 0.06 ml · cm H2O(-1)), reducing total lung water, and decreasing lung inflammation and histologic injury compared with control. Intratracheal MSC therapy attenuated alveolar tumor necrosis factor-α (130 ± 43 vs. 488 ± 211 pg · ml(-1)) and interleukin-6 concentrations (138 ± 18 vs. 260 ± 82 pg · ml(-1)). The efficacy of intratracheal MSCs was comparable with intravenous MSC therapy. Intratracheal MSCs seemed to act via a paracine mechanism, with conditioned MSC medium also enhancing lung repair after injury. CONCLUSIONS Intratracheal MSC therapy enhanced recovery after ventilation-induced lung injury via a paracrine mechanism, and was as effective as intravenous MSC therapy.
Collapse
|
90
|
Knoll A, Brockmeyer T, Chevalier R, Zscheppang K, Nielsen H, Dammann C. Adult Rat Bone Marrow-Derived Stem Cells Promote Late Fetal Type II Cell Differentiation in a Co-Culture Model. Open Respir Med J 2013; 7:46-53. [PMID: 23730368 PMCID: PMC3664445 DOI: 10.2174/1874306401307010046] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2013] [Revised: 03/08/2013] [Accepted: 03/13/2013] [Indexed: 01/08/2023] Open
Abstract
Bronchopulmonary dysplasia develops in preterm infants due to a combination of lung immaturity and lung injury. Cultured pluripotent bone marrow stem cells (BMSC) are known to reduce injury and induce repair in adult and in immature lungs, possibly through paracrine secretion of soluble factors. The paracrine relationship between BMSC and primary fetal lung epithelial type II cells is unknown. We determined the effects of BMSC on type II cell and fibroblast behavior using an in vitro co-culture model. Rat BMSC were isolated and co-cultured with primary fetal E21 rat type II cells or lung fibroblasts in a Transwell® system without direct cell contact. Effects of BMSC conditioned media (CM) on type II cell and fibroblast proliferation and on type II cell surfactant phospholipid (DSPC) synthesis and mRNA expression of surfactant proteins B and C (sftpb and sftpc) were studied. We also determined the effect of fibroblast and type II cell CM on BMSC proliferation and surface marker expression. Co-culture with BMSC significantly decreased type II cell and fibroblast proliferation to 72.5% and 83.7% of controls, respectively. Type II cell DSPC synthesis was significantly increased by 21% and sftpb and sftpc mRNA expressions were significantly induced (2.1 fold and 2.4 fold, respectively). BMSC proliferation was significantly reduced during the co-culture. Flow cytometry confirmed that BMSC retained the expression of undifferentiated stem cell markers despite their exposure to fetal lung cell CM. We conclude that BMSC induce fetal type II cell differentiation through paracrine release of soluble factors. These studies provide clues for how BMSC may act in promoting alveolar repair following injury.
Collapse
Affiliation(s)
- Ab Knoll
- Hannover Medical School, Hannover, Germany ; Division of Newborn Medicine, Floating Hospital for Children at Tufts Medical Center, Boston, MA, USA
| | | | | | | | | | | |
Collapse
|
91
|
Ohba T, Wada H, Yoshino I, Yoshida S, Tagawa T, Shoji F, Yamazaki K, Maehara Y. Increase of bone morphogenetic protein-7 expressing pulmonary resident cells in pneumonectomized rats. Surg Today 2013; 44:324-31. [PMID: 23640479 DOI: 10.1007/s00595-013-0604-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2012] [Accepted: 12/09/2012] [Indexed: 11/28/2022]
Abstract
PURPOSE Compensatory lung growth (CLG) is recognized in rodents subjected to major pulmonary resection; however, the source of cells constituting regenerated tissues during the CLG is still unknown. We investigated the differentiation of lung resident cells and the participation of bone marrow (BM)-derived cells in the remnant lung of pneumonectomized rats. METHODS After left pneumonectomy, the right remnant lung of Wistar rats was subjected to morphologic and molecular experiments at several time points. We studied the expression of bone morphogenic protein 7 (BMP-7), an accelerator of epithelial differentiation, based on the gene expression profile data of the remnant lung. Next, we evaluated the presence of GFP-positive cells in the remnant lung of Wistar rats that had received BM transplantation from green fluorescent protein (GFP) gene-transgenic Wistar rats prior to left pneumonectomy. RESULTS We observed progression of emphysematous change, modulation of gene expression profile, and proliferating cellular nuclear antigen-positive cells in the alveoli of the remnant lungs. BMP-7 protein positive cells were detected in the alveolar septa, which increased significantly over time with the progression of emphysematous change. No bone marrow-derived cells were detected in the right remnant lung of the GFP-BM transferred rats by fluorescence microscopy, immunohistochemistry, or polymerase chain reaction at any time. CONCLUSION Lung resident cells appear to contribute to CLG, possibly via a trans-differentiation pathway.
Collapse
Affiliation(s)
- Taro Ohba
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan,
| | | | | | | | | | | | | | | |
Collapse
|
92
|
Wang YY, Li XZ, Wang LB. Therapeutic implications of mesenchymal stem cells in acute lung injury/acute respiratory distress syndrome. Stem Cell Res Ther 2013; 4:45. [PMID: 23673003 PMCID: PMC3706892 DOI: 10.1186/scrt193] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Acute lung injury (ALI), and its more severe form, acute respiratory distress syndrome (ARDS), are syndromes of acute hypoxemic respiratory failure resulting from a variety of direct and indirect injuries to the gas exchange parenchyma of the lungs. Current treatment of ALI/ARDS is primarily supportive, with lung protective ventilation and fluid conserving strategies. Despite improvement in these strategies, recent data indicate that the mortality of ALI/ARDS is still as high as 30 to 50%. Thus, there is a need for innovative therapies to further improve clinical outcomes of ALI/ARDS. Recent studies involving the administration of mesenchymal stem cells (MSCs) for the treatment of experimental ALI/ARDS have shown promising results. This review focuses on existing studies that have tested the use of MSCs in models of ALI/ARDS, and the potential mechanisms underlying their therapeutic effects.
Collapse
|
93
|
Gao P, Yang X, Mungur L, Kampo S, Wen Q. Adipose tissue-derived stem cells attenuate acute lung injury through eNOS and eNOS-derived NO. Int J Mol Med 2013; 31:1313-8. [PMID: 23563270 DOI: 10.3892/ijmm.2013.1328] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2012] [Accepted: 02/14/2013] [Indexed: 02/04/2023] Open
Abstract
Acute lung injury (ALI) is among the most common causes of mortality in intensive care units. Recent in vivo and in vitro studies have suggested that mesenchymal stem cells (MSCs) attenuate pulmonary edema and inflammatory factors, but the mechanisms of the effects of MSCs on pulmonary vascular function remain unknown. It is believed that nitric oxide (NO) and endothelial nitric oxide synthase (eNOS) play an essential role in the regulation of vascular function and homeostasis. In the present study, we investigated the effect of adipose tissue-derived stem cells (ADSCs) on pulmonary microvascular endothelial cells (PMVECs) and the lung in a lipopolysaccharide (LPS)-induced ALI model in vitro and in vivo. Our results showed that ADSCs were able to attenuate the severity of ALI and pulmonary edema. Increased expression of the eNOS protein was also observed in pulmonary PMVECs and in the lung following treatment with ADSCs. Furthermore, ADSCs increased the concentration of eNOS-derived NO to remodel ALI. The results suggest that ADSCs may be a promising candidate for ALI treatment through interaction with eNOS and eNOS-derived NO.
Collapse
Affiliation(s)
- Peng Gao
- Department of Anesthesiology, Dalian Medical University, Dalian, People's Republic of China
| | | | | | | | | |
Collapse
|
94
|
The rabbit as a model for studying lung disease and stem cell therapy. BIOMED RESEARCH INTERNATIONAL 2013; 2013:691830. [PMID: 23653896 PMCID: PMC3638694 DOI: 10.1155/2013/691830] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 02/28/2013] [Accepted: 02/28/2013] [Indexed: 12/21/2022]
Abstract
No single animal model can reproduce all of the human features of both acute and chronic lung diseases. However, the rabbit is a reliable model and clinically relevant facsimile of human disease. The similarities between rabbits and humans in terms of airway anatomy and responses to inflammatory mediators highlight the value of this species in the investigation of lung disease pathophysiology and in the development of therapeutic agents. The inflammatory responses shown by the rabbit model, especially in the case of asthma, are comparable with those that occur in humans. The allergic rabbit model has been used extensively in drug screening tests, and this model and humans appear to be sensitive to similar drugs. In addition, recent studies have shown that the rabbit serves as a good platform for cell delivery for the purpose of stem-cell-based therapy.
Collapse
|
95
|
Ge X, Bai C, Yang J, Lou G, Li Q, Chen R. Intratracheal transplantation of bone marrow-derived mesenchymal stem cells reduced airway inflammation and up-regulated CD4⁺CD25⁺ regulatory T cells in asthmatic mouse. Cell Biol Int 2013; 37:675-86. [PMID: 23483727 DOI: 10.1002/cbin.10084] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2012] [Accepted: 02/12/2013] [Indexed: 12/18/2022]
Abstract
Mesenchymal stem cells attenuate the severity of lung injury due to their immunomodulatory properties. The effect of bone marrow-derived mesenchymal stem cells on asthma is seldom reported. We have examined the effect of BMSCs on airway inflammation in asthma. Forty female BALB/c mice were equally randomised into PBS group, BMSCs treatment group, BMSCs control group and asthmatic group. Reactivity of the airway to acetylcholine was measured by barometric plethysmography. Cytokine profiles of bronchoalveolar lavage fluid and serum were determined by enzyme-linked immunosorbent assay. Morphometric analysis was done with haematoxylin and periodic-acid Schiff staining. Engraftment of BMSCs in asthmatic mice significantly decreased the number of eosinophils and mononuclear cells in bronchoalveolar lavage fluid and the airway (P < 0.05). Both goblet cell hyperplasia and responsiveness to acetylcholine were significantly reduced in BMSCs treatment groups. Moreover, BMSCs engraftment caused significant increases the ratio of Treg in pulmonary lymph node and interleukin-10 (IL-10) and interleukin-12 levels in BALF and serum. We conclude that BMSCs engraftment ameliorated airway inflammation and improved lung function in asthmatic mouse and the protective effect might be mediated by upregulating Treg and partly involved with increasing IL-10.
Collapse
Affiliation(s)
- Xiahui Ge
- Department of Respiratory Medicine, Changhai Hospital of Second Military Medical University, Shanghai 200433, China
| | | | | | | | | | | |
Collapse
|
96
|
Wang H, Yang YF, Zhao L, Xiao FJ, Zhang QW, Wen ML, Wu CT, Peng RY, Wang LS. Hepatocyte growth factor gene-modified mesenchymal stem cells reduce radiation-induced lung injury. Hum Gene Ther 2013; 24:343-53. [PMID: 23458413 DOI: 10.1089/hum.2012.177] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Abstract Effective therapeutic strategies for radiation-induced lung injury (RILI) are lacking. Mesenchymal stem cells (MSCs), as gene therapy delivery vehicles, possess the ability to repair injured lung. In this study, we conducted MSC-based hepatocyte growth factor (HGF) gene therapy for RILI. Mice received single-dose radiation with 20 Gy of γ rays locally to the lung, and then were administered normal sodium, Ad-HGF-modified MSCs, or Ad-Null-modified MSCs. Ad-HGF-modified MSCs (MSCs-HGF) improved histopathological and biochemical markers of lung injury. MSCs-HGF could reduce secretion and expression of proinflammatory cytokines, including tumor necrosis factor-α, interferon-γ, interleukin (IL)-6, and intercellular adhesion molecule-1, and increase the expression of antiinflammatory cytokine IL-10. It could also decrease expression levels of profibrosis factors transforming growth factor-β, Col1a1 (collagen type 1, α1), and Col3a1, and inhibit fibrosis progress. MSCs-HGF could promote proliferation of lung epithelial cells and protect them from apoptosis, and improve the expression of endogenous HGF and its receptor c-Met significantly. We also found that sphingosine-1-phosphate receptor-1 expression was increased in injured lung. These results suggest MSC-based HGF gene therapy not only reduces inflammation but also inhibits lung fibrosis.
Collapse
Affiliation(s)
- Hua Wang
- Department of Experimental Hematology, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Chamoto K, Gibney BC, Ackermann M, Lee GS, Konerding MA, Tsuda A, Mentzer SJ. Alveolar epithelial dynamics in postpneumonectomy lung growth. Anat Rec (Hoboken) 2013; 296:495-503. [PMID: 23408540 PMCID: PMC3576046 DOI: 10.1002/ar.22659] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 05/14/2012] [Accepted: 07/24/2012] [Indexed: 11/07/2022]
Abstract
The intimate anatomic and functional relationship between epithelial cells and endothelial cells within the alveolus suggests the likelihood of a coordinated response during postpneumonectomy lung growth. To define the population dynamics and potential contribution of alveolar epithelial cells to alveolar angiogenesis, we studied alveolar Type II and I cells during the 21 days after pneumonectomy. Alveolar Type II cells were defined and isolated by flow cytometry using a CD45(-) , MHC class II(+) , phosphine(+) phenotype. These phenotypically defined alveolar Type II cells demonstrated an increase in cell number after pneumonectomy; the increase in cell number preceded the increase in Type I (T1α(+) ) cells. Using a parabiotic wild type/GFP pneumonectomy model, <3% of the Type II cells and 1% of the Type I cells were positive for GFP-a finding consistent with the absence of a blood-borne contribution to alveolar epithelial cells. The CD45(-) , MHC class II(+) , phosphine(+) Type II cells demonstrated the active transcription of angiogenesis-related genes both before and after pneumonectomy. When the Type II cells on Day 7 after pneumonectomy were compared to nonsurgical controls, 10 genes demonstrated significantly increased expression (P<0.05). In contrast to the normal adult Type II cells, there was notable expression of inflammation-associated genes (Ccl2, Cxcl2, Ifng) as well as genes associated with epithelial growth (Ereg, Lep). Together, the data suggest an active contribution of local alveolar Type II cells to alveolar growth.
Collapse
Affiliation(s)
- Kenji Chamoto
- Laboratory of Adaptive and Regenerative Biology, Brigham & Women’s Hospital, Harvard Medical School, Boston MA
| | - Barry C. Gibney
- Laboratory of Adaptive and Regenerative Biology, Brigham & Women’s Hospital, Harvard Medical School, Boston MA
| | - Maximilian Ackermann
- Institute of Functional and Clinical Anatomy, University Medical Center of Johannes Gutenberg-University, Mainz, Germany
| | - Grace S. Lee
- Laboratory of Adaptive and Regenerative Biology, Brigham & Women’s Hospital, Harvard Medical School, Boston MA
| | - Moritz A. Konerding
- Institute of Functional and Clinical Anatomy, University Medical Center of Johannes Gutenberg-University, Mainz, Germany
| | - Akira Tsuda
- Molecular and Integrative Physiological Sciences, Harvard School of Public Health, Boston, MA
| | - Steven J. Mentzer
- Laboratory of Adaptive and Regenerative Biology, Brigham & Women’s Hospital, Harvard Medical School, Boston MA
| |
Collapse
|
98
|
Katsuda T, Kosaka N, Takeshita F, Ochiya T. The therapeutic potential of mesenchymal stem cell-derived extracellular vesicles. Proteomics 2013; 13:1637-53. [PMID: 23335344 DOI: 10.1002/pmic.201200373] [Citation(s) in RCA: 312] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Revised: 10/04/2012] [Accepted: 10/22/2012] [Indexed: 12/13/2022]
Abstract
Extracellular vesicles (EVs), membrane vesicles that are secreted by a variety of mammalian cell types, have been shown to play an important role in intercellular communication. The contents of EVs, including proteins, microRNAs, and mRNAs, vary according to the cell type that secreted them. Accordingly, researchers have demonstrated that EVs derived from various cell types play different roles in biological phenomena. Considering the ubiquitous presence of mesenchymal stem cells (MSCs) in the body, MSC-derived EVs may take part in a wide range of events. In particular, MSCs have recently attracted much attention due to the therapeutic effects of their secretory factors. MSC-derived EVs may therefore provide novel therapeutic approaches. In this review, we first summarize the wide range of functions of EVs released from different cell types, emphasizing that EVs echo the phenotype of their parent cell. Then, we describe the various therapeutic effects of MSCs and pay particular attention to the significance of their paracrine effect. We then survey recent reports on MSC-derived EVs and consider the therapeutic potential of MSC-derived EVs. Finally, we discuss remaining issues that must be addressed before realizing the practical application of MSC-derived EVs, and we provide some suggestions for enhancing their therapeutic efficiency.
Collapse
Affiliation(s)
- Takeshi Katsuda
- Division of Molecular and Cellular Medicine, National Cancer Center Research Institute, Tokyo, Japan
| | | | | | | |
Collapse
|
99
|
Kapetanaki MG, Mora AL, Rojas M. Influence of age on wound healing and fibrosis. J Pathol 2013; 229:310-22. [PMID: 23124998 DOI: 10.1002/path.4122] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Revised: 09/30/2012] [Accepted: 10/02/2012] [Indexed: 12/18/2022]
Abstract
The incidence and severity of fibrotic lung diseases increase with age, but very little is known about how age-related changes affect the mechanisms that underlie disease emergence and progression. Normal ageing includes accumulation of DNA mutations, oxidative and cell stresses, mitochondria dysfunction, increased susceptibility to apoptosis, telomere length dysfunction and differential gene expression as a consequence of epigenetic changes and miR regulation. These inevitable ageing-related phenomena may cause dysfunction and impaired repair capacity of lung epithelial cells, fibroblasts and MSCs. As a consequence, the composition of the extracellular matrix changes and the dynamic interaction between cells and their environment is damaged, resulting ultimately in predisposition for several diseases. This review summarizes what is known about age-related molecular changes that are implicated in the pathobiology of lung fibrosis in lung tissue.
Collapse
Affiliation(s)
- Maria G Kapetanaki
- Dorothy P and Richard P Simmons Center for Interstitial Lung Disease, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | | |
Collapse
|
100
|
Akram KM, Samad S, Spiteri MA, Forsyth NR. Mesenchymal stem cells promote alveolar epithelial cell wound repair in vitro through distinct migratory and paracrine mechanisms. Respir Res 2013; 14:9. [PMID: 23350749 PMCID: PMC3598763 DOI: 10.1186/1465-9921-14-9] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Accepted: 01/22/2013] [Indexed: 01/09/2023] Open
Abstract
Background Mesenchymal stem cells (MSC) are in clinical trials for widespread indications including musculoskeletal, neurological, cardiac and haematological disorders. Furthermore, MSC can ameliorate pulmonary fibrosis in animal models although mechanisms of action remain unclear. One emerging concept is that MSCs may have paracrine, rather than a functional, roles in lung injury repair and regeneration. Methods To investigate the paracrine role of human MSC (hMSC) on pulmonary epithelial repair, hMSC-conditioned media (CM) and a selected cohort of hMSC-secretory proteins (identified by LC-MS/MS mass spectrometry) were tested on human type II alveolar epithelial cell line A549 cells (AEC) and primary human small airway epithelial cells (SAEC) using an in vitro scratch wound repair model. A 3D direct-contact wound repair model was further developed to assess the migratory properties of hMSC. Results We demonstrate that MSC-CM facilitates AEC and SAEC wound repair in serum-dependent and –independent manners respectively via stimulation of cell migration. We also show that the hMSC secretome contains an array of proteins including Fibronectin, Lumican, Periostin, and IGFBP-7; each capable of influencing AEC and SAEC migration and wound repair stimulation. In addition, hMSC also show a strong migratory response to AEC injury as, supported by the observation of rapid and effective AEC wound gap closure by hMSC in the 3D model. Conclusion These findings support the notion for clinical application of hMSCs and/or their secretory factors as a pharmacoregenerative modality for the treatment of idiopathic pulmonary fibrosis (IPF) and other fibrotic lung disorders.
Collapse
Affiliation(s)
- Khondoker M Akram
- Institute for Science and Technology in Medicine, School of Postgraduate Medicine, Keele University, Stoke-on-Trent, Staffordshire ST4 7QB, UK
| | | | | | | |
Collapse
|