1
|
Kor A, Güven SC, Akan S, Eren F, Ecem Konak H, Maraş Y, Orhan K, Neşelioğlu S, Erten Ş. Serum netrin-1 levels are high in Rheumatoid arthritis associated interstitial lung disease. Clin Biochem 2024; 127-128:110760. [PMID: 38556035 DOI: 10.1016/j.clinbiochem.2024.110760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 02/28/2024] [Accepted: 03/28/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND Recent data show that netrin-1 has a role in development of pulmonary fibrosis. This study was aimed to investigate serum netrin-1 level and its relation to interstitial lung disease(ILD) in patients with rheumatoid arthritis (RA). METHOD 42 RA patients with RA-ILD, 58 RA patients without RA-ILD (RA non-ILD group), and 61 healthy volunteers were included in this study. The modified DAS28-ESR score was used to calculate disease activity in RA patients. Using the quantitative immunoassay method, Serum netrin-1 levels were measured with an ELISA kit (Catalog number: E-EL-H2328; lab science, lot number: GZWTKZ5SWK, Texas, USA). RESULTS The median value of netrin-1 was found to be significantly higher in the RA-ILD group (82.9 [59.9-124]) compared to both the RA non-ILD group(52.9 [49.5-73.1])(B = -0.006, OR = 0.994, CI 95 %=0.989-0.999, P = 0.018) and the control group(53.5 [49.5-87.5]) (B: -0.005, OR: 0.994, CI 95 %: 0.990-0.999, p: 0.022). A cut-off value of 61.78 for netrin-1 was found to have a sensitivity of 73.8 % and a specificity of 69 % for the diagnosis of RA-ILD (AUC [95 %Cl] = 0.771 [0.679-0.862], p < 0.0001).It was found that high serum netrin-1 level was strongly associated with the RA-usual interstitial pneumonia(UIP) pattern and poorly related to the RA-nonspecific interstitial pneumonia(NSIP) pattern compared to the RA non-ILD group. CONCLUSIONS Netrin-1 is elevated in the serum of patients with RA-ILD, especially in the UIP pattern. Netrin-1 may be a potential candidate for predicting the development of RA-ILD that should be investigated in the pathophysiological and therapeutic fields..
Collapse
Affiliation(s)
- Ahmet Kor
- Department of Rheumatology, Aksaray Education and Research Hospital, Aksaray, Turkey.
| | - Serdar Can Güven
- Department of Rheumatology, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Selçuk Akan
- Department of Internal Medicine, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Funda Eren
- Department of Medical Biochemistry, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Hatice Ecem Konak
- Department of Rheumatology, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Yüksel Maraş
- Department of Rheumatology, University of Health Sciences, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Kevser Orhan
- Department of Rheumatology, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Salim Neşelioğlu
- Department of Medical Biochemistry, Ankara Yıldırım Beyazıt University, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Şükran Erten
- Department of Rheumatology, Ankara Yıldırım Beyazıt University, Ankara Bilkent City Hospital, Ankara, Turkey
| |
Collapse
|
2
|
Liu C, Hu X, Zhao Y, Huang A, Chen J, Lu T, Wu M, Lu H. High-Glucose-Induced Injury to Proximal Tubules of the Renal System Is Alleviated by Netrin-1 Suppression of Akt/mTOR. J Diabetes Res 2023; 2023:4193309. [PMID: 38033740 PMCID: PMC10684325 DOI: 10.1155/2023/4193309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 08/03/2023] [Accepted: 10/16/2023] [Indexed: 12/02/2023] Open
Abstract
The kidneys have a high level of Netrin-1 expression, which protects against some acute and chronic kidney disorders. However, it is yet unknown how Netrin-1 affects renal proximal tubule cells in diabetic nephropathy (DN) under pathological circumstances. Research has shown that autophagy protects the kidneys in animal models of renal disease. In this study, we looked at the probable autophagy regulation mechanism of Netrin-1 and its function in the pathogenesis of DN. We proved that in HK-2 cell, high blood sugar levels caused Netrin-1 to be downregulated, which then triggered the Akt/mTOR signaling pathway and enhanced cell death and actin cytoskeleton disruption. By adding Netrin-1 or an autophagy activator in vitro, these pathogenic alterations were reverted. Our results indicate that Netrin-1 stimulates autophagy by blocking the Akt/mTOR signaling pathway, which underlies high-glucose-induced malfunction of the renal proximal tubules. After HK-2 cells were incubated with Netrin-1 recombination protein and rapamycin under HG conditions for 24 h, the apoptosis was significantly reduced, as shown by the higher levels of Bcl-2, as well as lower levels of Bax and cleaved caspase-3 (P = 0.012, Cohen's d = 0.489, Glass's delta = 0.23, Hedges' g = 0.641). This study reveals that targeting Netrin-1-related signaling has therapeutic potential for DN and advances our knowledge of the processes operating in renal proximal tubules in DN.
Collapse
Affiliation(s)
- Chenxiao Liu
- Department of Endocrinology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, 242 Guangji Road, Jiangsu 215008, China
| | - Xingna Hu
- Department of Endocrinology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, 242 Guangji Road, Jiangsu 215008, China
| | - Yun Zhao
- Department of Endocrinology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, 242 Guangji Road, Jiangsu 215008, China
| | - Aijie Huang
- Department of Endocrinology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, 242 Guangji Road, Jiangsu 215008, China
| | - Jiaqi Chen
- Department of Endocrinology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, 242 Guangji Road, Jiangsu 215008, China
| | - Ting Lu
- Department of Endocrinology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, 242 Guangji Road, Jiangsu 215008, China
| | - Mian Wu
- Department of Endocrinology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, 242 Guangji Road, Jiangsu 215008, China
| | - Honghong Lu
- Department of Endocrinology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, 242 Guangji Road, Jiangsu 215008, China
| |
Collapse
|
3
|
Newman H, Varghese S. Extracellular adenosine signaling in bone health and disease. Curr Opin Pharmacol 2023; 70:102378. [PMID: 37044008 PMCID: PMC10247430 DOI: 10.1016/j.coph.2023.102378] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/29/2023] [Accepted: 03/13/2023] [Indexed: 04/14/2023]
Abstract
Purinergic signaling is a key molecular pathway in the maintenance of bone health and regeneration. P1 receptor signaling, which is activated by extracellular adenosine, has emerged as a key metabolic pathway that regulates bone tissue formation, function, and homeostasis. Extracellular adenosine is mainly produced by ectonucleotidases, and alterations in the function of these enzymes or compromised adenosine generation can result in bone disorders, such as osteoporosis and impaired fracture healing. This mini review discusses the key role played by adenosine in bone health and how its alterations contribute to bone diseases, as well as potential therapeutic applications of exogenous adenosine to combat bone diseases like osteoporosis and injury.
Collapse
Affiliation(s)
- Hunter Newman
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27710, USA; Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, USA
| | - Shyni Varghese
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, NC 27710, USA; Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC 27710, USA; Department of Biomedical Engineering, Duke University, Durham, NC 27710, USA.
| |
Collapse
|
4
|
Zhu Q, Liu X, Wu H, Yang C, Wang M, Chen F, Cui Y, Hao H, Hill MA, Liu Z. CARD9 deficiency improves the recovery of limb ischemia in mice with ambient fine particulate matter exposure. Front Cardiovasc Med 2023; 10:1125717. [PMID: 36860276 PMCID: PMC9968734 DOI: 10.3389/fcvm.2023.1125717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 01/30/2023] [Indexed: 02/15/2023] Open
Abstract
Background Exposure to fine particulate matter (PM) is a significant risk for cardiovascular diseases largely due to increased reactive oxygen species (ROS) production and inflammation. Caspase recruitment domain (CARD)9 is critically involved in innate immunity and inflammation. The present study was designed to test the hypothesis that CARD9 signaling is critically involved in PM exposure-induced oxidative stress and impaired recovery of limb ischemia. Methods and results Critical limb ischemia (CLI) was created in male wildtype C57BL/6 and age matched CARD9 deficient mice with or without PM (average diameter 2.8 μm) exposure. Mice received intranasal PM exposure for 1 month prior to creation of CLI and continued for the duration of the experiment. Blood flow and mechanical function were evaluated in vivo at baseline and days 3, 7, 14, and 21 post CLI. PM exposure significantly increased ROS production, macrophage infiltration, and CARD9 protein expression in ischemic limbs of C57BL/6 mice in association with decreased recovery of blood flow and mechanical function. CARD9 deficiency effectively prevented PM exposure-induced ROS production and macrophage infiltration and preserved the recovery of ischemic limb with increased capillary density. CARD9 deficiency also significantly attenuated PM exposure-induced increase of circulating CD11b+/F4/80+ macrophages. Conclusion The data indicate that CARD9 signaling plays an important role in PM exposure-induced ROS production and impaired limb recovery following ischemia in mice.
Collapse
Affiliation(s)
- Qiang Zhu
- Center for Precision Medicine, Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, United States
| | - Xuanyou Liu
- Center for Precision Medicine, Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, United States
| | - Hao Wu
- Center for Precision Medicine, Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, United States
| | - Chunlin Yang
- Center for Precision Medicine, Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, United States
| | - Meifang Wang
- Center for Precision Medicine, Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, United States
| | - Feng Chen
- Center for Precision Medicine, Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, United States
| | - Yuqi Cui
- Center for Precision Medicine, Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, United States
| | - Hong Hao
- Center for Precision Medicine, Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, United States
| | - Michael A. Hill
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
| | - Zhenguo Liu
- Center for Precision Medicine, Division of Cardiovascular Medicine, Department of Medicine, University of Missouri School of Medicine, Columbia, MO, United States,*Correspondence: Zhenguo Liu ✉
| |
Collapse
|
5
|
Maraş Y, Kor A, Oğuz EF, Sarı A, Gök K, Akdoğan A. Serum netrin-1 levels in systemic sclerosis patients with capillary abnormalities. THE EGYPTIAN RHEUMATOLOGIST 2023. [DOI: 10.1016/j.ejr.2022.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
6
|
Netrin-1 promotes the vasculogenic capacity of human adipose-derived stem cells. Cell Tissue Bank 2022; 24:357-367. [PMID: 36222969 DOI: 10.1007/s10561-022-10038-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 09/10/2022] [Indexed: 11/02/2022]
Abstract
Adipose derived stem cells (ADSCs) have been increasingly explored for use in cell-based therapy against ischemic diseases. However, unsatisfactory angiogenesis limits the therapeutic efficacy. Netrin-1, a known axon guidance molecule, improves neovascularization in the ischemic region. Thus, our study was performed to evaluate the potential effect of Netrin-1 on the angiogenic behaviors of human ADSCs (hADSCs). hADSCs acquired from human abdominal adipose tissue were modified by liposome transfection of Netrin-1 plasmid, and the proliferation of hADSCs was determined by Cell Counting Kit-8 (CCK-8) assay. The transcript levels of pro-invasive proteins such as matrix metalloproteinase 2 (MMP2) and matrix metalloproteinase 9 (MMP-9), were measured to test migratory and invasive capabilities, and the levels of vascular endothelial growth factors were assayed to monitor angiogenic activity. Our results showed that Netrin-1 overexpression enhanced the proliferation of hADSCs, and promoted the migration and invasion of hADSCs, as indicated by increased levels of MMP-2 and MMP-9. Furthermore, Netrin-1 overexpression increased the expression of vascular endothelial growth factor and placental growth factor in hADSCs. Our results highlighted the possibility that genetic modification of hADSCs by Netrin-1 overexpression might be beneficial for cell transplantation therapy against ischemic diseases.
Collapse
|
7
|
Zeng J, Lu C, Huang H, Huang J. Effect of Recombinant Netrin-1 Protein Combined with Peripheral Blood Mesenchymal Stem Cells on Angiogenesis in Rats with Arteriosclerosis Obliterans. BIOMED RESEARCH INTERNATIONAL 2022; 2022:3361605. [PMID: 35928912 PMCID: PMC9345694 DOI: 10.1155/2022/3361605] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 06/30/2022] [Accepted: 07/02/2022] [Indexed: 01/06/2023]
Abstract
This work was aimed to explore the effect of recombinant netrin-1 protein and peripheral blood mesenchymal stem cells (MSCs) on the angiogenesis ability of atherosclerosis. 28 Sprague Dawley (SD) rats were taken as research models. The arterial occlusion models were created by surgery and then divided into the saline control group (n =7), netrin-1 treatment group (n =7), MSCs treatment group (n =7), and netrin-1 + MSCs combined treatment group (n =7). The peripheral blood MSCs were extracted from the peritoneal cavity of diseased SD rats and cultured alone or in combination with netrin-1. The individually cultured MSCs and netrin-1 were locally injected into the ischemic tissues of SD rats. The Tarlov scoring was performed at the first, second, and third week of treatment, respectively. The expression of vascular endothelial growth factor (VEGF) was also measured by quantitative real-time polymerase chain reaction (qRT-PCR), and the capillary density was measured by immunofluorescence staining. The mean maximum contractility of the gastrocnemius muscle in each group was determined in the third week after treatment. The Tarlov score of the netrin-1 + MSCs group was significantly higher than that of the control group (P < 0.05) at the second week. To the 4th week of treatment, the Tarlov score of the netrin-1 + MSCs group was highly increased compared to the netrin-1 group and the MSCs group (P < 0.05). The expression of VEGF in the treatment groups was greatly increased each week compared to the control group (P < 0.05). Compared with the netrin-1 and the MSCs groups, the VEGF was also notably increased in the netrin-1 + MSCs group (P <0.05). The capillary densities of the treatment groups were observably greater than that of the control group in the second and third weeks (P <0.05), while the capillary density in the netrin-1 + MSCs group was also significantly increased than those in the netrin-1 group and the MSCs group (P < 0.05). The mean maximum contractility of the netrin-1 + MSCs group was remarkably higher than that of the other groups (P < 0.05). The netrin-1 + MSCs group achieved the higher Tarlov score, higher VEGF expression, higher capillary density, and better muscle recovery than netrin-1 and MSCs treatments.
Collapse
Affiliation(s)
- Jie Zeng
- Department of Cardiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, 610072 Sichuan, China
| | - Cong Lu
- Department of Cardiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, 610072 Sichuan, China
| | - Hui Huang
- Department of Cardiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, 610072 Sichuan, China
| | - Jianxin Huang
- Department of Anesthesiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, 610072 Sichuan, China
| |
Collapse
|
8
|
Zhu Q, Liu X, Zhu Q, Liu Z, Yang C, Wu H, Zhang L, Xia X, Wang M, Hao H, Cui Y, Zhang G, Hill MA, Flaker GC, Zhou S, Liu Z. N-Acetylcysteine Enhances the Recovery of Ischemic Limb in Type-2 Diabetic Mice. Antioxidants (Basel) 2022; 11:antiox11061097. [PMID: 35739993 PMCID: PMC9219773 DOI: 10.3390/antiox11061097] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 12/16/2022] Open
Abstract
Critical limb ischemia (CLI) is a severe complication of diabetes mellitus that occurs without effective therapy. Excessive reactive oxygen species (ROS) production and oxidative stress play critical roles in the development of diabetic cardiovascular complications. N-acetylcysteine (NAC) reduces ischemia-induced ROS production. The present study aimed to investigate the effect of NAC on the recovery of ischemic limb in an experimental model of type-2 diabetes. TALLYHO/JngJ diabetic and SWR/J non-diabetic mice were used for developing a CLI model. For NAC treatment, mice received NAC (1 mg/mL) in their drinking water for 24 h before initiating CLI, and continuously for the duration of the experiment. Blood flow, mechanical function, histology, expression of antioxidant enzymes including superoxide dismutase (SOD)-1, SOD-3, glutathione peroxidase (Gpx)-1, catalase, and phosphorylated insulin receptor substrate (IRS)-1, Akt, and eNOS in ischemic limb were evaluated in vivo or ex vivo. Body weight, blood glucose, plasma advanced glycation end-products (AGEs), plasma insulin, insulin resistance index, and plasma TNF-a were also evaluated during the experiment. NAC treatment effectively attenuated ROS production with preserved expressions of SOD-1, Gpx-1, catalase, phosphorylated Akt, and eNOS, and enhanced the recovery of blood flow and function of the diabetic ischemic limb. NAC treatment also significantly decreased the levels of phosphorylated IRS-1 (Ser307) expression and plasma TNF-α in diabetic mice without significant changes in blood glucose and AGEs levels. In conclusion, NAC treatment enhanced the recovery of blood flow and mechanical function in ischemic limbs in T2D mice in association with improved tissue redox/inflammatory status and insulin resistance.
Collapse
Affiliation(s)
- Qiang Zhu
- Center for Precision Medicine, Department of Medicine, Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, MO 65212, USA; (Q.Z.); (X.L.); (Q.Z.); (Z.L.); (C.Y.); (H.W.); (L.Z.); (X.X.); (M.W.); (H.H.); (Y.C.); (G.C.F.)
- Department of Cardiology, Second Xiangya Hospital, Central South University, Changsha 410011, China;
| | - Xuanyou Liu
- Center for Precision Medicine, Department of Medicine, Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, MO 65212, USA; (Q.Z.); (X.L.); (Q.Z.); (Z.L.); (C.Y.); (H.W.); (L.Z.); (X.X.); (M.W.); (H.H.); (Y.C.); (G.C.F.)
| | - Qingyi Zhu
- Center for Precision Medicine, Department of Medicine, Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, MO 65212, USA; (Q.Z.); (X.L.); (Q.Z.); (Z.L.); (C.Y.); (H.W.); (L.Z.); (X.X.); (M.W.); (H.H.); (Y.C.); (G.C.F.)
- Department of Cardiology, Second Xiangya Hospital, Central South University, Changsha 410011, China;
| | - Zehao Liu
- Center for Precision Medicine, Department of Medicine, Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, MO 65212, USA; (Q.Z.); (X.L.); (Q.Z.); (Z.L.); (C.Y.); (H.W.); (L.Z.); (X.X.); (M.W.); (H.H.); (Y.C.); (G.C.F.)
| | - Chunlin Yang
- Center for Precision Medicine, Department of Medicine, Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, MO 65212, USA; (Q.Z.); (X.L.); (Q.Z.); (Z.L.); (C.Y.); (H.W.); (L.Z.); (X.X.); (M.W.); (H.H.); (Y.C.); (G.C.F.)
| | - Hao Wu
- Center for Precision Medicine, Department of Medicine, Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, MO 65212, USA; (Q.Z.); (X.L.); (Q.Z.); (Z.L.); (C.Y.); (H.W.); (L.Z.); (X.X.); (M.W.); (H.H.); (Y.C.); (G.C.F.)
| | - Linfang Zhang
- Center for Precision Medicine, Department of Medicine, Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, MO 65212, USA; (Q.Z.); (X.L.); (Q.Z.); (Z.L.); (C.Y.); (H.W.); (L.Z.); (X.X.); (M.W.); (H.H.); (Y.C.); (G.C.F.)
| | - Xiujuan Xia
- Center for Precision Medicine, Department of Medicine, Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, MO 65212, USA; (Q.Z.); (X.L.); (Q.Z.); (Z.L.); (C.Y.); (H.W.); (L.Z.); (X.X.); (M.W.); (H.H.); (Y.C.); (G.C.F.)
| | - Meifang Wang
- Center for Precision Medicine, Department of Medicine, Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, MO 65212, USA; (Q.Z.); (X.L.); (Q.Z.); (Z.L.); (C.Y.); (H.W.); (L.Z.); (X.X.); (M.W.); (H.H.); (Y.C.); (G.C.F.)
| | - Hong Hao
- Center for Precision Medicine, Department of Medicine, Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, MO 65212, USA; (Q.Z.); (X.L.); (Q.Z.); (Z.L.); (C.Y.); (H.W.); (L.Z.); (X.X.); (M.W.); (H.H.); (Y.C.); (G.C.F.)
| | - Yuqi Cui
- Center for Precision Medicine, Department of Medicine, Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, MO 65212, USA; (Q.Z.); (X.L.); (Q.Z.); (Z.L.); (C.Y.); (H.W.); (L.Z.); (X.X.); (M.W.); (H.H.); (Y.C.); (G.C.F.)
| | - Guangsen Zhang
- Institute of Molecular Hematopathy, Second Xiangya Hospital, Central South University, Changsha 410011, China;
| | - Michael A. Hill
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO 65211, USA;
| | - Gregory C. Flaker
- Center for Precision Medicine, Department of Medicine, Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, MO 65212, USA; (Q.Z.); (X.L.); (Q.Z.); (Z.L.); (C.Y.); (H.W.); (L.Z.); (X.X.); (M.W.); (H.H.); (Y.C.); (G.C.F.)
| | - Shenghua Zhou
- Department of Cardiology, Second Xiangya Hospital, Central South University, Changsha 410011, China;
| | - Zhenguo Liu
- Center for Precision Medicine, Department of Medicine, Division of Cardiovascular Medicine, University of Missouri School of Medicine, Columbia, MO 65212, USA; (Q.Z.); (X.L.); (Q.Z.); (Z.L.); (C.Y.); (H.W.); (L.Z.); (X.X.); (M.W.); (H.H.); (Y.C.); (G.C.F.)
- Correspondence: ; Tel.: +1-573-884-3278; Fax: +1-573-884-7743
| |
Collapse
|
9
|
Modulation of Mesenchymal Stem Cells for Enhanced Therapeutic Utility in Ischemic Vascular Diseases. Int J Mol Sci 2021; 23:ijms23010249. [PMID: 35008675 PMCID: PMC8745455 DOI: 10.3390/ijms23010249] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 12/23/2021] [Accepted: 12/24/2021] [Indexed: 12/12/2022] Open
Abstract
Mesenchymal stem cells are multipotent stem cells isolated from various tissue sources, including but not limited to bone marrow, adipose, umbilical cord, and Wharton Jelly. Although cell-mediated mechanisms have been reported, the therapeutic effect of MSCs is now recognized to be primarily mediated via paracrine effects through the secretion of bioactive molecules, known as the “secretome”. The regenerative benefit of the secretome has been attributed to trophic factors and cytokines that play neuroprotective, anti-angiogenic/pro-angiogenic, anti-inflammatory, and immune-modulatory roles. The advancement of autologous MSCs therapy can be hindered when introduced back into a hostile/disease environment. Barriers include impaired endogenous MSCs function, limited post-transplantation cell viability, and altered immune-modulatory efficiency. Although secretome-based therapeutics have gained popularity, many translational hurdles, including the heterogeneity of MSCs, limited proliferation potential, and the complex nature of the secretome, have impeded the progress. This review will discuss the experimental and clinical impact of restoring the functional capabilities of MSCs prior to transplantation and the progress in secretome therapies involving extracellular vesicles. Modulation and utilization of MSCs–secretome are most likely to serve as an effective strategy for promoting their ultimate success as therapeutic modulators.
Collapse
|
10
|
Zhu Q, Hao H, Xu H, Fichman Y, Cui Y, Yang C, Wang M, Mittler R, Hill MA, Cowan PJ, Zhang G, He X, Zhou S, Liu Z. Combination of Antioxidant Enzyme Overexpression and N-Acetylcysteine Treatment Enhances the Survival of Bone Marrow Mesenchymal Stromal Cells in Ischemic Limb in Mice With Type 2 Diabetes. J Am Heart Assoc 2021; 10:e023491. [PMID: 34569277 PMCID: PMC8649154 DOI: 10.1161/jaha.121.023491] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background Therapy with mesenchymal stem cells remains a promising but challenging approach to critical limb ischemia in diabetes because of the dismal cell survival. Methods and Results Critical limb ischemia in type 2 diabetes mouse model was used to explore the impact of diabetic limb ischemia on the survival of bone marrow mesenchymal stromal cells (bMSCs). Inhibition of intracellular reactive oxygen species was achieved with concomitant overexpression of superoxide dismutase (SOD)‐1 and glutathione peroxidase‐1 in the transplanted bMSCs, and extracellular reactive oxygen species was attenuated using SOD‐3 overexpression and N‐acetylcysteine treatment. In vivo optical fluorescence imaging and laser Doppler perfusion imaging were used to track cell retention and determine blood flow in diabetic ischemic limb, respectively. Survival of the transplanted bMSCs was significantly decreased in diabetic ischemic limb compared with the control. In vitro study indicated that advanced glycation end products, not high glucose, significantly decreased the proliferation of bMSCs and increased their apoptosis associated with increased reactive oxygen species production and selective reduction of SOD‐1 and SOD‐3. In vivo study demonstrated that concomitant overexpression of SOD‐1, SOD‐3, and glutathione peroxidase‐1, or host treatment with N‐acetylcysteine, significantly enhanced in vivo survival of transplanted bMSCs, and improved critical limb ischemia in diabetic mice. Combination of triple antioxidant enzyme overexpression in bMSCs with host N‐acetylcysteine treatment further improved bMSC survival with enhanced circulatory and functional recovery from diabetic critical limb ischemia. Conclusions Simultaneous suppression of reactive oxygen species from transplanted bMSCs and host tissue could additively enhance bMSC survival in diabetic ischemic limb with increased therapeutic efficacy in diabetes.
Collapse
Affiliation(s)
- Qiang Zhu
- Center for Precision Medicine and Division of Cardiovascular Medicine Department of Medicine University of Missouri School of Medicine Columbia MO.,Department of Cardiology Second Xiangya Hospital Central South University Changsha City Hunan Province China
| | - Hong Hao
- Center for Precision Medicine and Division of Cardiovascular Medicine Department of Medicine University of Missouri School of Medicine Columbia MO
| | - Huifang Xu
- Center for Precision Medicine and Division of Cardiovascular Medicine Department of Medicine University of Missouri School of Medicine Columbia MO
| | - Yosef Fichman
- College of Agriculture, Food and Natural Resources University of Missouri Columbia MO.,Dalton Cardiovascular Research Center University of Missouri Columbia MO
| | - Yuqi Cui
- Center for Precision Medicine and Division of Cardiovascular Medicine Department of Medicine University of Missouri School of Medicine Columbia MO
| | - Chunlin Yang
- Center for Precision Medicine and Division of Cardiovascular Medicine Department of Medicine University of Missouri School of Medicine Columbia MO
| | - Meifang Wang
- Center for Precision Medicine and Division of Cardiovascular Medicine Department of Medicine University of Missouri School of Medicine Columbia MO
| | - Ron Mittler
- College of Agriculture, Food and Natural Resources University of Missouri Columbia MO.,Dalton Cardiovascular Research Center University of Missouri Columbia MO
| | - Michael A Hill
- Dalton Cardiovascular Research Center University of Missouri Columbia MO.,Department of Surgery University of Missouri School of MedicineChristopher S. Bond Life Sciences CenterUniversity of Missouri Columbia MO
| | - Peter J Cowan
- Department of Medicine University of Melbourne Australia.,Immunology Research Centre St. Vincent's Hospital Melbourne Australia
| | - Guangsen Zhang
- Institute of Molecular Hematopathy Second Xiangya Hospital Central South University Changsha City Hunan Province China
| | - Xiaoming He
- Fischell Department of Bioengineering University of Maryland College Park MD
| | - Shenghua Zhou
- Department of Cardiology Second Xiangya Hospital Central South University Changsha City Hunan Province China
| | - Zhenguo Liu
- Center for Precision Medicine and Division of Cardiovascular Medicine Department of Medicine University of Missouri School of Medicine Columbia MO
| |
Collapse
|
11
|
Netrin-1 improves adipose-derived stem cell proliferation, migration, and treatment effect in type 2 diabetic mice with sciatic denervation. Stem Cell Res Ther 2018; 9:285. [PMID: 30359296 PMCID: PMC6202825 DOI: 10.1186/s13287-018-1020-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 09/16/2018] [Accepted: 09/27/2018] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Diabetic peripheral neurovascular diseases (DPNVs) are complex, lacking effective treatment. Autologous/allogeneic transplantation of adipose-derived stem cells (ADSCs) is a promising strategy for DPNVs. Nonetheless, the transplanted ADSCs demonstrate unsatisfying viability, migration, adhesion, and differentiation in vivo, which reduce the treatment efficiency. Netrin-1 secreted as an axon guidance molecule and served as an angiogenic factor, demonstrating its ability in enhancing cell proliferation, migration, adhesion, and neovascularization. METHODS ADSCs acquired from adipose tissue were modified by Netrin-1 gene (NTN-1) using the adenovirus method (N-ADSCs) and proliferation, migration, adhesion, and apoptosis examined under high-glucose condition. The sciatic denervated mice (db/db) with type 2 diabetes mellitus (T2DM) were transplanted with N-ADSCs and treatment efficiency assessed based on the laser Doppler perfusion index, immunofluorescence, and histopathological assay. Also, the molecular mechanisms underlying Netrin-1-mediated proliferation, migration, adhesion, differentiation, proangiogenic capacity, and apoptosis of ADSCs were explored. RESULTS N-ADSCs improved the proliferation, migration, and adhesion and inhibited the apoptosis of ADSCs in vitro in the condition of high glucose. The N-ADSCs group demonstrated an elevated laser Doppler perfusion index in the ADSCs and control groups. N-ADSCs analyzed by immunofluorescence and histopathological staining demonstrated the distribution of the cells in the injected limb muscles, indicating chronic ischemia; capillaries and endothelium were formed by differentiation of N-ADSCs. The N-ADSCs group showed a significantly high density of the microvessels than the ADSCs group. The upregulation of AKT/PI3K/eNOS/P-38/NF-κB signaling pathways and secretion of multiple growth factors might explain the positive effects of Netrin-1 on ADSCs. CONCLUSION The overexpression of Netrin-1 in ADSCs improves proliferation, migration, and treatment effect in type 2 diabetic mice with sciatic denervation, which directs the clinical treatment of patients with DPNVs.
Collapse
|
12
|
Huang H, Lin F, Jiang J, Chen Y, Mei A, Zhu P. Effects of intra-arterial transplantation of adipose-derived stem cells on the expression of netrin-1 and its receptor DCC in the peri-infarct cortex after experimental stroke. Stem Cell Res Ther 2017; 8:223. [PMID: 29017609 PMCID: PMC5633888 DOI: 10.1186/s13287-017-0671-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 09/11/2017] [Accepted: 09/13/2017] [Indexed: 12/16/2022] Open
Abstract
Background Stem cell transplantation has been documented to promote functional recovery in animal models of stroke; however, the underlying mechanisms are not yet fully understood. As netrin-1 and its receptor deleted in colorectal cancer (DCC) are important regulators in neuronal and vascular activities, the present study attempted to explore whether netrin-1 and DCC are involved in the neuroprotection of stem cell-based therapies in a rat ischemic stroke model. Methods Adult male Sprague–Dawley rats were subjected to a transient middle cerebral artery occlusion (MCAO) and subsequently received an intra-arterial injection of 2 × 106 PKH26-labeled adipose-derived stem cells (ADSCs) or saline 24 h later. Neurological function was evaluated by behavioral tests before the rats were sacrificed at days 7 and 14 after MCAO. The migration of ADSCs and regeneration of neuronal fibers and blood vessels were determined by immunofluorescence staining. The expression of netrin-1 and DCC was analyzed by Western blot and immunofluorescence staining. Results ADSC transplantation significantly improved the neurological recovery at days 7 and 14, and noticeably promoted the regeneration of neuronal fibers and blood vessels in the peri-infarct cortex at day 14. PKH26-labeled ADSCs located mainly in the peri-infarct area at days 7 and 14. In ADSC-treated rats, the expression of netrin-1 and DCC significantly increased in the peri-infarct cortex at days 7 and 14. Immunofluorescence staining showed that netrin-1 was mainly expressed by neuronal perikaryal in the peri-infarct cortex, and DCC was mainly expressed by neuronal fibers and was present around the blood vessels in the peri-infarct cortex. Conclusions These findings suggest that ADSC transplantation facilitates the regeneration of neuronal fibers and blood vessels in the peri-infarct cortex and improves neurological functions, which may be attributed, at least in part, to the involvement of upregulated netrin-1 and DCC in the remodeling of neuronal and vascular networks in the peri-infarct cortex.
Collapse
Affiliation(s)
- Huan Huang
- Department of Geriatric Medicine, Fujian Provincial Hospital, 134 Dongjie Road, Fuzhou, Fujian, 350001, China.,Provincial Clinical Medical College of Fujian Medical University, 134 Dongjie Road, Fuzhou, Fujian, 350001, China.,Fujian Key Laboratory of Geriatrics, 134 Dongjie Road, Fuzhou, Fujian, 350001, China
| | - Fan Lin
- Department of Geriatric Medicine, Fujian Provincial Hospital, 134 Dongjie Road, Fuzhou, Fujian, 350001, China.,Provincial Clinical Medical College of Fujian Medical University, 134 Dongjie Road, Fuzhou, Fujian, 350001, China.,Fujian Key Laboratory of Geriatrics, 134 Dongjie Road, Fuzhou, Fujian, 350001, China
| | - Jingjing Jiang
- Department of Geriatric Medicine, Fujian Provincial Hospital, 134 Dongjie Road, Fuzhou, Fujian, 350001, China
| | - Yan Chen
- Department of Geriatric Medicine, Fujian Provincial Hospital, 134 Dongjie Road, Fuzhou, Fujian, 350001, China.,Provincial Clinical Medical College of Fujian Medical University, 134 Dongjie Road, Fuzhou, Fujian, 350001, China.,Fujian Key Laboratory of Geriatrics, 134 Dongjie Road, Fuzhou, Fujian, 350001, China
| | - Ainong Mei
- Department of Geriatric Medicine, Fujian Provincial Hospital, 134 Dongjie Road, Fuzhou, Fujian, 350001, China.,Provincial Clinical Medical College of Fujian Medical University, 134 Dongjie Road, Fuzhou, Fujian, 350001, China.,Fujian Key Laboratory of Geriatrics, 134 Dongjie Road, Fuzhou, Fujian, 350001, China
| | - Pengli Zhu
- Department of Geriatric Medicine, Fujian Provincial Hospital, 134 Dongjie Road, Fuzhou, Fujian, 350001, China. .,Provincial Clinical Medical College of Fujian Medical University, 134 Dongjie Road, Fuzhou, Fujian, 350001, China. .,Fujian Key Laboratory of Geriatrics, 134 Dongjie Road, Fuzhou, Fujian, 350001, China.
| |
Collapse
|
13
|
Prieto CP, Ortiz MC, Villanueva A, Villarroel C, Edwards SS, Elliott M, Lattus J, Aedo S, Meza D, Lois P, Palma V. Netrin-1 acts as a non-canonical angiogenic factor produced by human Wharton's jelly mesenchymal stem cells (WJ-MSC). Stem Cell Res Ther 2017; 8:43. [PMID: 28241866 PMCID: PMC5330133 DOI: 10.1186/s13287-017-0494-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 01/16/2017] [Accepted: 02/08/2017] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Angiogenesis, the process in which new blood vessels are formed from preexisting ones, is highly dependent on the presence of classical angiogenic factors. Recent evidence suggests that axonal guidance proteins and their receptors can also act as angiogenic regulators. Netrin, a family of laminin-like proteins, specifically Netrin-1 and 4, act via DCC/Neogenin-1 and UNC5 class of receptors to promote or inhibit angiogenesis, depending on the physiological context. METHODS Mesenchymal stem cells secrete a broad set of classical angiogenic factors. However, little is known about the expression of non-canonical angiogenic factors such as Netrin-1. The aim was to characterize the possible secretion of Netrin ligands by Wharton's jelly-derived mesenchymal stem cells (WJ-MSC). We evaluated if Netrin-1 presence in the conditioned media from these cells was capable of inducing angiogenesis both in vitro and in vivo, using human umbilical vein endothelial cells (HUVEC) and chicken chorioallantoic membrane (CAM), respectively. In addition, we investigated if the RhoA/ROCK pathway is responsible for the integration of Netrin signaling to control vessel formation. RESULTS The paracrine angiogenic effect of the WJ-MSC-conditioned media is mediated at least in part by Netrin-1 given that pharmacological blockage of Netrin-1 in WJ-MSC resulted in diminished angiogenesis on HUVEC. When HUVEC were stimulated with exogenous Netrin-1 assayed at physiological concentrations (10-200 ng/mL), endothelial vascular migration occurred in a concentration-dependent manner. In line with our determination of Netrin-1 present in WJ-MSC-conditioned media we were able to obtain endothelial tubule formation even in the pg/mL range. Through CAM assays we validated that WJ-MSC-secreted Netrin-1 promotes an increased angiogenesis in vivo. Netrin-1, secreted by WJ-MSC, might mediate its angiogenic effect through specific cell surface receptors on the endothelium, such as UNC5b and/or integrin α6β1, expressed in HUVEC. However, the angiogenic response of Netrin-1 seems not to be mediated through the RhoA/ROCK pathway. CONCLUSIONS Thus, here we show that stromal production of Netrin-1 is a critical component of the vascular regulatory machinery. This signaling event may have deep implications in the modulation of several processes related to a number of diseases where angiogenesis plays a key role in vascular homeostasis.
Collapse
Affiliation(s)
- Catalina P. Prieto
- Laboratory of Stem Cells and Developmental Biology, Faculty of Sciences, University of Chile, Santiago de Chile, Chile
| | - María Carolina Ortiz
- Laboratory of Stem Cells and Developmental Biology, Faculty of Sciences, University of Chile, Santiago de Chile, Chile
| | - Andrea Villanueva
- Laboratory of Stem Cells and Developmental Biology, Faculty of Sciences, University of Chile, Santiago de Chile, Chile
| | - Cynthia Villarroel
- Laboratory of Stem Cells and Developmental Biology, Faculty of Sciences, University of Chile, Santiago de Chile, Chile
| | - Sandra S. Edwards
- Laboratory of Stem Cells and Developmental Biology, Faculty of Sciences, University of Chile, Santiago de Chile, Chile
| | - Matías Elliott
- Laboratory of Stem Cells and Developmental Biology, Faculty of Sciences, University of Chile, Santiago de Chile, Chile
| | - José Lattus
- Campus Oriente, Department of Obstetrics and Gynecology, Faculty of Medicine, University of Chile, Santiago de Chile, Chile
| | - Sócrates Aedo
- Campus Oriente, Department of Obstetrics and Gynecology, Faculty of Medicine, University of Chile, Santiago de Chile, Chile
| | - Daniel Meza
- Laboratory of Stem Cells and Developmental Biology, Faculty of Sciences, University of Chile, Santiago de Chile, Chile
| | - Pablo Lois
- Laboratory of Stem Cells and Developmental Biology, Faculty of Sciences, University of Chile, Santiago de Chile, Chile
| | - Verónica Palma
- Laboratory of Stem Cells and Developmental Biology, Faculty of Sciences, University of Chile, Santiago de Chile, Chile
| |
Collapse
|
14
|
Lee SS, Lee SJ, Lee SH, Ryu JM, Lim HS, Kim JS, Song EJ, Jung YH, Lee HJ, Kim CH, Han HJ. Netrin-1-Induced Stem Cell Bioactivity Contributes to the Regeneration of Injured Tissues via the Lipid Raft-Dependent Integrin α6β4 Signaling Pathway. Sci Rep 2016; 6:37526. [PMID: 27881869 PMCID: PMC5121594 DOI: 10.1038/srep37526] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 10/31/2016] [Indexed: 11/17/2022] Open
Abstract
Netrin-1 (Ntn-1) is a multifunctional neuronal signaling molecule; however, its physiological significance, which improves the tissue-regeneration capacity of stem cells, has not been characterized. In the present study, we investigate the mechanism by which Ntn-1 promotes the proliferation of hUCB-MSCs with regard to the regeneration of injured tissues. We found that Ntn-1 induces the proliferation of hUCB-MSCs mainly via Inα6β4 coupled with c-Src. Ntn-1 induced the recruitment of NADPH oxidases and Rac1 into membrane lipid rafts to facilitate ROS production. The Inα6β4 signaling of Ntn-1 through ROS production is uniquely mediated by the activation of SP1 for cell cycle progression and the transcriptional occupancy of SP1 on the VEGF promoter. Moreover, Ntn-1 has the ability to induce the F-actin reorganization of hUCB-MSCs via the Inα6β4 signaling pathway. In an in vivo model, transplantation of hUCB-MSCs pre-treated with Ntn-1 enhanced the skin wound healing process, where relatively more angiogenesis was detected. The potential effect of Ntn-1 on angiogenesis is further verified by the mouse hindlimb ischemia model, where the pre-activation of hUCB-MSCs with Ntn-1 significantly improved vascular regeneration. These results demonstrate that Ntn-1 plays an important role in the tissue regeneration process of hUCB-MSC via the lipid raft-mediated Inα6β4 signaling pathway.
Collapse
Affiliation(s)
- Soo Sang Lee
- Department of plastic and reconstructive surgery, Bundang CHA Medical Center, Yatap-dong, Bundang-gu, Seongnam-si, Gyeonggi-do, Korea.,SKY plastic surgery clinic, 4F, 826-23, Yeoksam-dong, Gangnam-gu, Seoul, Korea
| | - Sei-Jung Lee
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Korea
| | - Sang Hun Lee
- Medical Science Research Institute, Soonchunhyang University Seoul Hospital, Seoul, Korea
| | - Jung Min Ryu
- Department of Veterinary Physiology, College of Veterinary Medicine, Chonnam National University, Gwangju, Republic of Korea
| | - Hyeon Su Lim
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Korea
| | - Jun Sung Kim
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Korea
| | - Eun Ju Song
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Korea
| | - Young Hyun Jung
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Korea
| | - Hyun Jik Lee
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Korea
| | - Chung Hun Kim
- Department of plastic and reconstructive surgery, Bundang CHA Medical Center, Yatap-dong, Bundang-gu, Seongnam-si, Gyeonggi-do, Korea
| | - Ho Jae Han
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science and BK21 PLUS Program for Creative Veterinary Science Research Center, Seoul National University, Seoul 08826, Korea
| |
Collapse
|