1
|
Lin J, Lin HW, Wang YX, Fang Y, Jiang HM, Li T, Huang J, Zhang HD, Chen DZ, Chen YP. FGF4 ameliorates the liver inflammation by reducing M1 macrophage polarization in experimental autoimmune hepatitis. J Transl Med 2024; 22:717. [PMID: 39095789 PMCID: PMC11295337 DOI: 10.1186/s12967-024-05219-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 04/19/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND The global prevalence of autoimmune hepatitis (AIH) is increasing due in part to the lack of effective pharmacotherapies. Growing evidence suggests that fibroblast growth factor 4 (FGF4) is crucial for diverse aspects of liver pathophysiology. However, its role in AIH remains unknown. Therefore, we investigated whether FGF4 can regulate M1 macrophage and thereby help treat liver inflammation in AIH. METHODS We obtained transcriptome-sequencing and clinical data for patients with AIH. Mice were injected with concanavalin A to induce experimental autoimmune hepatitis (EAH). The mechanism of action of FGF4 was examined using macrophage cell lines and bone marrow-derived macrophages. RESULTS We observed higher expression of markers associated with M1 and M2 macrophages in patients with AIH than that in individuals without AIH. EAH mice showed greater M1-macrophage polarization than control mice. The expression of M1-macrophage markers correlated positively with FGF4 expression. The loss of hepatic Fgf4 aggravated hepatic inflammation by increasing the abundance of M1 macrophages. In contrast, the pharmacological administration of FGF4 mitigated hepatic inflammation by reducing M1-macrophage levels. The efficacy of FGF4 treatment was compromised following the in vivo clearance of macrophage populations. Mechanistically, FGF4 treatment activated the phosphatidylinositol 3-kinase (PI3K)-protein kinase B (AKT)-signal pathway in macrophages, which led to reduced M1 macrophages and hepatic inflammation. CONCLUSION We identified FGF4 as a novel M1/M2 macrophage-phenotype regulator that acts through the PI3K-AKT-signaling pathway, suggesting that FGF4 may represent a novel target for treating inflammation in patients with AIH.
Collapse
Affiliation(s)
- Jing Lin
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Hepatology Institute of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
- Department of Infectious Diseases, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| | - Hong-Wei Lin
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Hepatology Institute of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yu-Xing Wang
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Hepatology Institute of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yan Fang
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Hepatology Institute of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Hui-Mian Jiang
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Hepatology Institute of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Ting Li
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Hepatology Institute of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Jia Huang
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Hepatology Institute of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Hua-Dong Zhang
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Hepatology Institute of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Da-Zhi Chen
- Department of Clinical Medicine, Hangzhou Medical College, Hangzhou, 310053, China.
| | - Yong-Ping Chen
- Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Hepatology Institute of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
2
|
Lee SB, Abdal Dayem A, Kmiecik S, Lim KM, Seo DS, Kim HT, Kumar Biswas P, Do M, Kim DH, Cho SG. Efficient improvement of the proliferation, differentiation, and anti-arthritic capacity of mesenchymal stem cells by simply culturing on the immobilized FGF2 derived peptide, 44-ERGVVSIKGV-53. J Adv Res 2024; 62:119-141. [PMID: 37777063 PMCID: PMC11331723 DOI: 10.1016/j.jare.2023.09.041] [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: 09/06/2022] [Revised: 08/23/2023] [Accepted: 09/26/2023] [Indexed: 10/02/2023] Open
Abstract
INTRODUCTION The stem cell microenvironment has been evidenced to robustly affect its biological functions and clinical grade. Natural or synthetic growth factors, especially, are essential for modulating stem cell proliferation, metabolism, and differentiation via the interaction with specific extracellular receptors. Fibroblast growth factor-2 (FGF-2) possesses pleiotropic functions in various tissues and organs. It interacts with the FGF receptor (FGFR) and activates FGFR signaling pathways, which involve numerous biological functions, such as angiogenesis, wound healing, cell proliferation, and differentiation. OBJECTIVES Here, we aim to explore the molecular functions, mode of action, and therapeutic activity of yet undetermined function, FGF-2-derived peptide, FP2 (44-ERGVVSIKGV-53) in promoting the proliferation, differentiation, and therapeutic application of human Wharton's jelly mesenchymal stem cells (hWJ-MSCs) in comparison to other test peptides, canofin1 (FP1), hexafin2 (FP3), and canofin3 (FP4) with known functions. METHODS The immobilization of test peptides that are fused with mussel adhesive proteins (MAP) on the culture plate was carried out via EDC/NHS chemistry. Cell Proliferation assay, colony-forming unit, western blotting analysis, gene expression analysis, RNA-Seq. analysis, osteogenic, and chondrogenic differentiation capacity were applied to test the activity of the test peptides. We additionally utilized three-dimensional (3D) structural analysis and artificial intelligence (AI)-based AlphaFold2 and CABS-dock programs for receptor interaction prediction of the peptide receptor. We also verified the in vivo therapeutic capacity of FP2-cultured hWJ-MSCs using an osteoarthritis mice model. RESULTS Culture of hWJ-MSC onto an FP2-immobilized culture plate showed a significant increase in cell proliferation (n = 3; *p < 0.05, **p < 0.01) and the colony-forming unit (n = 3; *p < 0.05, **p < 0.01) compared with the test peptides. FP2 showed a significantly upregulated phosphorylation of FRS2α and FGFR1 and activated the AKT and ERK signaling pathways (n = 3; *p < 0.05, **p < 0.01, ***p < 0.001). Interestingly, we detected efficient FP2 receptor binding that was predicted using AI-based tools. Treatment with an AKT inhibitor significantly abrogated the FP2-mediated enhancement of cell differentiation (n = 3; *p < 0.05, **p < 0.01, ***p < 0.001). Intra-articular injection of FP2-cultured MSCs significantly mitigated arthritis symptoms in an osteoarthritis mouse model, as shown through the functional tests (n = 10; *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001), modulation of the expression level of the pro-inflammatory and anti-inflammatory genes, and improved osteochondral regeneration as demonstrated by tissue sections. CONCLUSION Our study identified the FGF-2-derived peptide FP2 as a promising candidate peptide to improve the therapeutic potential of hWJ-MSCs, especially in bone and cartilage regeneration.
Collapse
Affiliation(s)
- Soo Bin Lee
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Ahmed Abdal Dayem
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Sebastian Kmiecik
- Biological and Chemical Research Centre, Faculty of Chemistry, University of Warsaw, 02-089 Warsaw, Poland
| | - Kyung Min Lim
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; R&D Team, StemExOne Co., Ltd., 307 KU Technology Innovation Bldg, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Dong Sik Seo
- Stem Cell Research Center of AMOLIFESCIENCE Co., Ltd, 91, Gimpo-daero 1950 Beon-gil, Tongjin-eup, Gimpo-si, Gyeonggi-do 10014, Republic of Korea
| | - Hyeong-Taek Kim
- Stem Cell Research Center of AMOLIFESCIENCE Co., Ltd, 91, Gimpo-daero 1950 Beon-gil, Tongjin-eup, Gimpo-si, Gyeonggi-do 10014, Republic of Korea
| | - Polash Kumar Biswas
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Minjae Do
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205 USA
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21205 USA
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea; R&D Team, StemExOne Co., Ltd., 307 KU Technology Innovation Bldg, 120, Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea.
| |
Collapse
|
3
|
Bao L, Sun Z, Dang L, Zhang Q, Zheng L, Yang F, Zhang J. LncRNA RP11-818O24.3 promotes hair-follicle recovery via FGF2-PI3K/Akt signal pathway. Cytotechnology 2024; 76:425-439. [PMID: 38933868 PMCID: PMC11196536 DOI: 10.1007/s10616-024-00624-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 02/16/2024] [Indexed: 06/28/2024] Open
Abstract
A previous study indicated that patients with androgenic alopecia (AGA) have significantly reduced levels of LncRNA RP11-818O24.3. This study investigates whether LncRNA RP11-818O24.3 promotes hair-follicle recovery and its possible mechanism. Hair alteration and cutaneous histopathological changes induced by testosterone propionate were observed by H&E and bromodeoxyuridinc (BrdU) stain to evaluate the therapeutic effect of LncRNA RP11-818O24.3 in C57BL/6 J mice. The cellular viability was analyzed in LncRNA RP11-818O24.3-transfected human hair-follicle stem cells (HFSCs) in vitro. The signaling pathways and pro-proliferative factors were investigated by transcriptomic gene sequencing and qRT-PCR. LncRNA RP11-818O24.3 transfection successfully recovered hair growth and hair-follicle cells in AGA mice. In a series of HFSC studies in vitro, LncRNA RP11-818O24.3 transfection greatly promoted cellular proliferation and decreased cellular apoptosis. Transcriptome gene sequencing suggested that the phosphatidylinositol 3-kinase (PI3K)-Akt pathway was upregulated by LncRNA RP11-818O24.3. The qRT-PCR results showed that fibroblast growth factor (FGF)-2 was 14-times upregulated after LncRNA RP11-818O24.3 transfection. Hair-follicle recovery activity of LncRNA RP11-818O24.3 may involve the upregulation of FGF2 and PI3K-Akt to promote follicle stem cell survival. These data not only provide a theoretical basis for AGA development but also reveal a novel therapeutic method for AGA patients. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-024-00624-3.
Collapse
Affiliation(s)
- Linlin Bao
- Department of Dermatology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), No. 1017, Dongmen North Road, Luohu District, Shenzhen, 518020 Guangdong China
- Candidate Branch of National Clinical Research Center for Skin Diseases, Shenzhen, 518020 Guangdong China
| | - Zhaojun Sun
- Department of Dermatology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), No. 1017, Dongmen North Road, Luohu District, Shenzhen, 518020 Guangdong China
- Candidate Branch of National Clinical Research Center for Skin Diseases, Shenzhen, 518020 Guangdong China
| | - Lin Dang
- Department of Dermatology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), No. 1017, Dongmen North Road, Luohu District, Shenzhen, 518020 Guangdong China
- Candidate Branch of National Clinical Research Center for Skin Diseases, Shenzhen, 518020 Guangdong China
| | - Qianqian Zhang
- Department of Dermatology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), No. 1017, Dongmen North Road, Luohu District, Shenzhen, 518020 Guangdong China
- Candidate Branch of National Clinical Research Center for Skin Diseases, Shenzhen, 518020 Guangdong China
| | - Lixiong Zheng
- Department of Dermatology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), No. 1017, Dongmen North Road, Luohu District, Shenzhen, 518020 Guangdong China
- Candidate Branch of National Clinical Research Center for Skin Diseases, Shenzhen, 518020 Guangdong China
| | - Fang Yang
- Department of Dermatology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), No. 1017, Dongmen North Road, Luohu District, Shenzhen, 518020 Guangdong China
- Candidate Branch of National Clinical Research Center for Skin Diseases, Shenzhen, 518020 Guangdong China
| | - Jianglin Zhang
- Department of Dermatology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), No. 1017, Dongmen North Road, Luohu District, Shenzhen, 518020 Guangdong China
- Candidate Branch of National Clinical Research Center for Skin Diseases, Shenzhen, 518020 Guangdong China
| |
Collapse
|
4
|
Weston WA, Barr AR. A cell cycle centric view of tumour dormancy. Br J Cancer 2023; 129:1535-1545. [PMID: 37608096 PMCID: PMC10645753 DOI: 10.1038/s41416-023-02401-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 07/31/2023] [Accepted: 08/10/2023] [Indexed: 08/24/2023] Open
Abstract
Tumour dormancy and recurrent metastatic cancer remain the greatest clinical challenge for cancer patients. Dormant tumour cells can evade treatment and detection, while retaining proliferative potential, often for years, before relapsing to tumour outgrowth. Cellular quiescence is one mechanism that promotes and maintains tumour dormancy due to its central role in reducing proliferation, elevating cyto-protective mechanisms, and retaining proliferative potential. Quiescence/proliferation decisions are dictated by intrinsic and extrinsic signals, which regulate the activity of cyclin-dependent kinases (CDKs) to modulate cell cycle gene expression. By clarifying the pathways regulating CDK activity and the signals which activate them, we can better understand how cancer cells enter, maintain, and escape from quiescence throughout the progression of dormancy and metastatic disease. Here we review how CDK activity is regulated to modulate cellular quiescence in the context of tumour dormancy and highlight the therapeutic challenges and opportunities it presents.
Collapse
Affiliation(s)
- William A Weston
- MRC London Institute of Medical Sciences, Du Cane Road, London, W12 0NN, UK
| | - Alexis R Barr
- MRC London Institute of Medical Sciences, Du Cane Road, London, W12 0NN, UK.
- Institute of Clinical Sciences, Imperial College London, Du Cane Rd, London, W12 0NN, UK.
| |
Collapse
|
5
|
Quadri N, Upadhyai P. Primary cilia in skeletal development and disease. Exp Cell Res 2023; 431:113751. [PMID: 37574037 DOI: 10.1016/j.yexcr.2023.113751] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/09/2023] [Accepted: 08/11/2023] [Indexed: 08/15/2023]
Abstract
Primary cilia are non-motile, microtubule-based sensory organelle present in most vertebrate cells with a fundamental role in the modulation of organismal development, morphogenesis, and repair. Here we focus on the role of primary cilia in embryonic and postnatal skeletal development. We examine evidence supporting its involvement in physiochemical and developmental signaling that regulates proliferation, patterning, differentiation and homeostasis of osteoblasts, chondrocytes, and their progenitor cells in the skeleton. We discuss how signaling effectors in mechanotransduction and bone development, such as Hedgehog, Wnt, Fibroblast growth factor and second messenger pathways operate at least in part at the primary cilium. The relevance of primary cilia in bone formation and maintenance is underscored by a growing list of rare genetic skeletal ciliopathies. We collate these findings and summarize the current understanding of molecular factors and mechanisms governing primary ciliogenesis and ciliary function in skeletal development and disease.
Collapse
Affiliation(s)
- Neha Quadri
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Priyanka Upadhyai
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India.
| |
Collapse
|
6
|
Zhao R, Chen Y, Wang D, Zhang C, Song H, Ni G. Role of irisin in bone diseases. Front Endocrinol (Lausanne) 2023; 14:1212892. [PMID: 37600697 PMCID: PMC10436578 DOI: 10.3389/fendo.2023.1212892] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 07/10/2023] [Indexed: 08/22/2023] Open
Abstract
Bone diseases are common among middle-aged and elderly people, and harm to activities of daily living (ADL) and quality of life (QOL) for patients. It is crucial to search for key regulatory factors associated with the development of bone diseases and explore potential therapeutic targets for bone diseases. Irisin is a novel myokine that has been discovered in recent years. Accumulating evidence indicates that irisin has beneficial effects in the treatment of various diseases such as metabolic, cardiovascular and neurological disorders, especially bone-related diseases. Recent studies had shown that irisin plays the role in various bone diseases such as osteoarthritis, osteoporosis and other bone diseases, suggesting that irisin may be a potential molecule for the prevention and treatment of bone diseases. Therefore, in this review, by consulting the related domestic and international literature of irisin and bone diseases, we summarized the specific regulatory mechanisms of irisin in various bone diseases, and provided a systematic theoretical basis for its application in the diagnosis and treatment of the bone diseases.
Collapse
Affiliation(s)
- Ruobing Zhao
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Yan Chen
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Dongxue Wang
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Chunyu Zhang
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Henan Song
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Guoxin Ni
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Xiamen University, Xiamen, China
| |
Collapse
|
7
|
Goh D, Yang Y, Lee EH, Hui JHP, Yang Z. Managing the Heterogeneity of Mesenchymal Stem Cells for Cartilage Regenerative Therapy: A Review. Bioengineering (Basel) 2023; 10:bioengineering10030355. [PMID: 36978745 PMCID: PMC10045936 DOI: 10.3390/bioengineering10030355] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/12/2023] [Accepted: 03/12/2023] [Indexed: 03/18/2023] Open
Abstract
Articular cartilage defects commonly result from trauma and are associated with significant morbidity. Since cartilage is an avascular, aneural, and alymphatic tissue with a poor intrinsic healing ability, the regeneration of functional hyaline cartilage remains a difficult clinical problem. Mesenchymal stem cells (MSCs) are multipotent cells with multilineage differentiation potential, including the ability to differentiate into chondrocytes. Due to their availability and ease of ex vivo expansion, clinicians are increasingly applying MSCs in the treatment of cartilage lesions. However, despite encouraging pre-clinical and clinical data, inconsistencies in MSC proliferative and chondrogenic potential depending on donor, tissue source, cell subset, culture conditions, and handling techniques remain a key barrier to widespread clinical application of MSC therapy in cartilage regeneration. In this review, we highlight the strategies to manage the heterogeneity of MSCs ex vivo for more effective cartilage repair, including reducing the MSC culture expansion period, and selecting MSCs with higher chondrogenic potential through specific genetic markers, surface markers, and biophysical attributes. The accomplishment of a less heterogeneous population of culture-expanded MSCs may improve the scalability, reproducibility, and standardisation of MSC therapy for clinical application in cartilage regeneration.
Collapse
Affiliation(s)
- Doreen Goh
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
| | - Yanmeng Yang
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
- Critical Analytics for Manufacturing Personalised-Medicine, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore
| | - Eng Hin Lee
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
- Critical Analytics for Manufacturing Personalised-Medicine, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore
| | - James Hoi Po Hui
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
| | - Zheng Yang
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower block Level 11, Singapore 119288, Singapore
- NUS Tissue Engineering Program, Life Sciences Institute, National University of Singapore, 27 Medical Drive, DSO (Kent Ridge) Building, Level 4, Singapore 11751, Singapore
- Critical Analytics for Manufacturing Personalised-Medicine, Singapore-MIT Alliance for Research and Technology, Singapore 138602, Singapore
- Correspondence: ; Tel.: +65-6516-5398
| |
Collapse
|
8
|
Effects of Ginsenoside Rg1 on the Biological Behavior of Human Amnion-Derived Mesenchymal Stem/Stromal Cells (hAD-MSCs). Stem Cells Int 2023; 2023:7074703. [PMID: 36845966 PMCID: PMC9946746 DOI: 10.1155/2023/7074703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 11/22/2022] [Accepted: 02/01/2023] [Indexed: 02/17/2023] Open
Abstract
Ginsenoside Rg1 (Rg1) is purified from ginseng with various pharmacological effects, which might facilitate the biological behavior of human amnion-derived mesenchymal stem/stromal cells (hAD-MSCs). This study is aimed at investigating the effects of Rg1 on the biological behavior, such as viability, proliferation, apoptosis, senescence, migration, and paracrine, of hAD-MSCs. hAD-MSCs were isolated from human amnions. The effects of Rg1 on the viability, proliferation, apoptosis, senescence, migration, and paracrine of hAD-MSCs were detected by CCK-8, EdU, flow cytometry, SA-β-Gal staining, wound healing, and ELISA assays, respectively. The protein expression levels were detected by western blot. Cell cycle distribution was evaluated using flow cytometry. We found that Rg1 promoted hAD-MSC cycle progression from G0/G1 to S and G2/M phases and significantly increased hAD-MSC proliferation rate. Rg1 activated PI3K/AKT signaling pathway and significantly upregulated the expressions of cyclin D, cyclin E, CDK4, and CDK2 in hAD-MSCs. Inhibition of PI3K/AKT signaling significantly downregulated the expressions of cyclin D, cyclin E, CDK4, and CDK2, prevented cell cycle progression, and reduced hAD-MSC proliferation induced by Rg1. hAD-MSC senescence rate was significantly increased by D-galactose, while the elevated hAD-MSC senescence rate induced by D-galactose was significantly decreased by Rg1 treatment. D-galactose significantly induced the expressions of senescence markers, p16INK4a, p14ARF, p21CIP1, and p53 in hAD-MSCs, while Rg1 significantly reduced the expressions of those markers induced by D-galactose in hAD-MSCs. Rg1 significantly promoted the secretion of IGF-I in hAD-MSCs. Rg1 reduced the hAD-MSC apoptosis rate. However, the difference was not significant. Rg1 had no influence on hAD-MSC migration. Altogether, our results demonstrate that Rg1 can promote the viability, proliferation, and paracrine and relieve the senescence of hAD-MSCs. PI3K/AKT signaling pathway is involved in the promotive effect of Rg1 on hAD-MSC proliferation. The protective effect of Rg1 on hAD-MSC senescence may be achieved via the downregulation of p16INK4A and p53/p21CIP1 pathway.
Collapse
|
9
|
Owen JS, Clayton A, Pearson HB. Cancer-Associated Fibroblast Heterogeneity, Activation and Function: Implications for Prostate Cancer. Biomolecules 2022; 13:67. [PMID: 36671452 PMCID: PMC9856041 DOI: 10.3390/biom13010067] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/19/2022] [Accepted: 12/27/2022] [Indexed: 01/01/2023] Open
Abstract
The continuous remodeling of the tumor microenvironment (TME) during prostate tumorigenesis is emerging as a critical event that facilitates cancer growth, progression and drug-resistance. Recent advances have identified extensive communication networks that enable tumor-stroma cross-talk, and emphasized the functional importance of diverse, heterogeneous stromal fibroblast populations during malignant growth. Cancer-associated fibroblasts (CAFs) are a vital component of the TME, which mediate key oncogenic events including angiogenesis, immunosuppression, metastatic progression and therapeutic resistance, thus presenting an attractive therapeutic target. Nevertheless, how fibroblast heterogeneity, recruitment, cell-of-origin and differential functions contribute to prostate cancer remains to be fully delineated. Developing our molecular understanding of these processes is fundamental to developing new therapies and biomarkers that can ultimately improve clinical outcomes. In this review, we explore the current challenges surrounding fibroblast identification, discuss new mechanistic insights into fibroblast functions during normal prostate tissue homeostasis and tumorigenesis, and illustrate the diverse nature of fibroblast recruitment and CAF generation. We also highlight the promise of CAF-targeted therapies for the treatment of prostate cancer.
Collapse
Affiliation(s)
- Jasmine S. Owen
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Cardiff CF24 4HQ, UK
| | - Aled Clayton
- Tissue Microenvironment Group, Division of Cancer & Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | - Helen B. Pearson
- The European Cancer Stem Cell Research Institute, School of Biosciences, Cardiff University, Cardiff CF24 4HQ, UK
| |
Collapse
|
10
|
Advance of Mesenchymal Stem Cells in Chronic End-Stage Liver Disease Control. Stem Cells Int 2022; 2022:1526217. [PMID: 36248254 PMCID: PMC9568364 DOI: 10.1155/2022/1526217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 09/18/2022] [Accepted: 09/25/2022] [Indexed: 11/26/2022] Open
Abstract
The chronic liver diseases will slowly develop into liver fibrosis, cirrhosis, and even liver cancer if no proper control is performed with high efficiency. Up to now, the most effective treatment for end-stage liver diseases is liver transplantation. However, liver transplantation has the problems of donor deficiency, low matching rate, surgical complications, high cost, and immune rejection. These problems indicate that novel therapeutic strategies are urgently required. Mesenchymal stem cells (MSCs) are somatic stem cells with multidirectional differentiation potential and self-renewal ability. MSCs can secrete a large number of cytokines, chemokines, immunomodulatory molecules, and hepatotrophic factors, as well as produce extracellular vesicles. They alleviate liver diseases by differentiating to hepatocyte-like cells, immunomodulation, homing to the injured site, regulating cell ferroptosis, regulating cell autophagy, paracrine effects, and MSC-mitochondrial transfer. In this review, we focus on the main resources of MSCs, underlying therapeutic mechanisms, clinical applications, and efforts made to improve MSC-based cell therapy efficiency.
Collapse
|
11
|
Du X, McManus DP, Fogarty CE, Jones MK, You H. Schistosoma mansoni Fibroblast Growth Factor Receptor A Orchestrates Multiple Functions in Schistosome Biology and in the Host-Parasite Interplay. Front Immunol 2022; 13:868077. [PMID: 35812433 PMCID: PMC9257043 DOI: 10.3389/fimmu.2022.868077] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 05/26/2022] [Indexed: 12/02/2022] Open
Abstract
Stem cells play significant roles in driving the complex life cycle of Schistosoma mansoni. Fibroblast growth factor (FGF) receptor A (SmFGFRA) is essential for maintaining the integrity of schistosome stem cells. Using immunolocalization, we demonstrated that SmFGFRA was distributed abundantly in germinal/stem cells of different S. mansoni life stages including eggs, miracidia, cercariae, schistosomula and adult worms. Indeed, SmFGFRA was also localized amply in embryonic cells and in the perinuclear region of immature eggs; von Lichtenberg's layer and the neural mass of mature eggs; the ciliated surface and neural mass of miracidia; the tegument cytosol of cercariae, schistosomula and adult worms; and was present in abundance in the testis and vitellaria of adult worms of S. mansoni. The distribution pattern of SmFGFRA illustrates the importance of this molecule in maintaining stem cells, development of the nervous and reproductive system of schistosomes, and in the host-parasite interplay. We showed SmFGFRA can bind human FGFs, activating the mitogen activated protein kinase (MAPK) pathway of adult worms in vitro. Inhibition of FGF signaling by the specific tyrosine kinase inhibitor BIBF 1120 significantly reduced egg hatching ability and affected the behavior of miracidia hatched from the treated eggs, emphasizing the importance of FGF signaling in driving the life cycle of S. mansoni. Our findings provide increased understanding of the complex schistosome life cycle and host-parasite interactions, indicating components of the FGF signaling pathway may represent promising targets for developing new interventions against schistosomiasis.
Collapse
Affiliation(s)
- Xiaofeng Du
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Donald P. McManus
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Conor E. Fogarty
- Genecology Research Centre, University of the Sunshine Coast, Brisbane, QLD, Australia
| | - Malcolm K. Jones
- School of Veterinary Science, The University of Queensland, Gatton, QLD, Australia
| | - Hong You
- Infection and Inflammation Program, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
12
|
Wang J, Xiao L, Wang W, Zhang D, Ma Y, Zhang Y, Wang X. The Auxiliary Role of Heparin in Bone Regeneration and its Application in Bone Substitute Materials. Front Bioeng Biotechnol 2022; 10:837172. [PMID: 35646879 PMCID: PMC9133562 DOI: 10.3389/fbioe.2022.837172] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 04/13/2022] [Indexed: 11/18/2022] Open
Abstract
Bone regeneration in large segmental defects depends on the action of osteoblasts and the ingrowth of new blood vessels. Therefore, it is important to promote the release of osteogenic/angiogenic growth factors. Since the discovery of heparin, its anticoagulant, anti-inflammatory, and anticancer functions have been extensively studied for over a century. Although the application of heparin is widely used in the orthopedic field, its auxiliary effect on bone regeneration is yet to be unveiled. Specifically, approximately one-third of the transforming growth factor (TGF) superfamily is bound to heparin and heparan sulfate, among which TGF-β1, TGF-β2, and bone morphogenetic protein (BMP) are the most common growth factors used. In addition, heparin can also improve the delivery and retention of BMP-2 in vivo promoting the healing of large bone defects at hyper physiological doses. In blood vessel formation, heparin still plays an integral part of fracture healing by cooperating with the platelet-derived growth factor (PDGF). Importantly, since heparin binds to growth factors and release components in nanomaterials, it can significantly facilitate the controlled release and retention of growth factors [such as fibroblast growth factor (FGF), BMP, and PDGF] in vivo. Consequently, the knowledge of scaffolds or delivery systems composed of heparin and different biomaterials (including organic, inorganic, metal, and natural polymers) is vital for material-guided bone regeneration research. This study systematically reviews the structural properties and auxiliary functions of heparin, with an emphasis on bone regeneration and its application in biomaterials under physiological conditions.
Collapse
Affiliation(s)
- Jing Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Lan Xiao
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, Australia
- Australia−China Centre for Tissue Engineering and Regenerative Medicine, Brisbane, Australia
| | - Weiqun Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Dingmei Zhang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yaping Ma
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yi Zhang
- Department of Hygiene Toxicology, School of Public Health, Zunyi Medical University, Zunyi, China
| | - Xin Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
- Centre for Biomedical Technologies, School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, Australia
- Australia−China Centre for Tissue Engineering and Regenerative Medicine, Brisbane, Australia
| |
Collapse
|
13
|
Cho S, Lee H, Lee HY, Kim SJ, Song W. The effect of fibroblast growth factor receptor inhibition on resistance exercise training-induced adaptation of bone and muscle quality in mice. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY 2022; 26:207-218. [PMID: 35477548 PMCID: PMC9046891 DOI: 10.4196/kjpp.2022.26.3.207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/19/2022] [Accepted: 03/24/2022] [Indexed: 11/15/2022]
Affiliation(s)
- Suhan Cho
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hojun Lee
- Department of Sport and Exercise Science, Seoul Women's University, Seoul 01797, Korea
| | - Ho-Young Lee
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seongnam 13620, Korea
| | - Sung Joon Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul 03080, Korea
- Ischemic/Hypoxic Disease Institute, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Wook Song
- Health and Exercise Science Laboratory, Institute of Sport Science, Seoul 08826, Korea
- Department of Physical Education, Seoul National University, Seoul 08826, Korea
- Institute on Aging, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
14
|
Zhu C, Xu Z, Yuan Y, Wang T, Xu C, Yin C, Xie P, Xu P, Ye H, Patel N, Schaul S, Wang L, Zhu X, Wang S, Gao P, Xi Q, Zhang Y, Shu G, Jiang Q. Heparin impairs skeletal muscle glucose uptake by inhibiting insulin binding to insulin receptor. ENDOCRINOLOGY DIABETES & METABOLISM 2021; 4:e00253. [PMID: 34277977 PMCID: PMC8279624 DOI: 10.1002/edm2.253] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/25/2021] [Accepted: 03/27/2021] [Indexed: 12/24/2022]
Abstract
Aim Heparin, a widely used antithrombotic drug has many other anticoagulant-independent physiological functions. Here, we elucidate a novel role of heparin in glucose homeostasis, suggesting an approach for developing heparin-targeted therapies for diabetes. Methods For serum heparin levels and correlation analysis, 122 volunteer's plasma, DIO (4 weeks HFD) and db/db mice serums were collected and used for spectrophotometric determination. OGTT, ITT, 2-NBDG uptake and muscle GLUT4 immunofluorescence were detected in chronic intraperitoneal injection of heparin or heparinase (16 days) and muscle-specific loss-of-function mice. In 293T cells, the binding of insulin to its receptor was detected by fluorescence resonance energy transfer (FRET), Myc-GLUT4-mCherry plasmid was used in GLUT4 translocation. In vitro, C2C12 cells as mouse myoblast cells were further verified the effects of heparin on glucose homeostasis through 2-NBDG uptake, Western blot and co-immunoprecipitation. Results Serum concentrations of heparin are positively associated with blood glucose levels in humans and are significantly increased in diet-induced and db/db obesity mouse models. Consistently, a chronic intraperitoneal injection of heparin results in hyperglycaemia, glucose intolerance and insulin resistance. These effects are independent of heparin's anticoagulant function and associated with decreases in glucose uptake and translocation of glucose transporter type 4 (GLUT4) in skeletal muscle. By using a muscle-specific loss-of-function mouse model, we further demonstrated that muscle GLUT4 is required for the detrimental effects of heparin on glucose homeostasis. Conclusions Heparin reduced insulin binding to its receptor by interacting with insulin and inhibited insulin-mediated activation of the PI3K/Akt signalling pathway in skeletal muscle, which leads to impaired glucose uptake and hyperglycaemia.
Collapse
Affiliation(s)
- Canjun Zhu
- Guangdong Laboratory of Lingnan Modern Agriculture Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry College of Animal Science South China Agricultural University Guangzhou China
| | | | - Yexian Yuan
- Guangdong Laboratory of Lingnan Modern Agriculture Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry College of Animal Science South China Agricultural University Guangzhou China
| | - Tao Wang
- Guangdong Laboratory of Lingnan Modern Agriculture Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry College of Animal Science South China Agricultural University Guangzhou China
| | - Chang Xu
- Guangdong Laboratory of Lingnan Modern Agriculture Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry College of Animal Science South China Agricultural University Guangzhou China
| | - Cong Yin
- Guangdong Laboratory of Lingnan Modern Agriculture Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry College of Animal Science South China Agricultural University Guangzhou China
| | - Peipei Xie
- Guangdong Laboratory of Lingnan Modern Agriculture Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry College of Animal Science South China Agricultural University Guangzhou China
| | - Pingwen Xu
- Division of Endocrinology, Diabetes and Metabolism Department of Medicine The University of Illinois at Chicago Chicago IL USA
| | - Hui Ye
- Division of Endocrinology, Diabetes and Metabolism Department of Medicine The University of Illinois at Chicago Chicago IL USA
| | - Nirali Patel
- Division of Endocrinology, Diabetes and Metabolism Department of Medicine The University of Illinois at Chicago Chicago IL USA
| | - Sarah Schaul
- Division of Endocrinology, Diabetes and Metabolism Department of Medicine The University of Illinois at Chicago Chicago IL USA
| | - Lina Wang
- Guangdong Laboratory of Lingnan Modern Agriculture Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry College of Animal Science South China Agricultural University Guangzhou China
| | - Xiaotong Zhu
- Guangdong Laboratory of Lingnan Modern Agriculture Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry College of Animal Science South China Agricultural University Guangzhou China
| | - Songbo Wang
- Guangdong Laboratory of Lingnan Modern Agriculture Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry College of Animal Science South China Agricultural University Guangzhou China
| | - Ping Gao
- Guangdong Laboratory of Lingnan Modern Agriculture Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry College of Animal Science South China Agricultural University Guangzhou China
| | - Qianyun Xi
- Guangdong Laboratory of Lingnan Modern Agriculture Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry College of Animal Science South China Agricultural University Guangzhou China
| | - Yongliang Zhang
- Guangdong Laboratory of Lingnan Modern Agriculture Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry College of Animal Science South China Agricultural University Guangzhou China
| | - Gang Shu
- Guangdong Laboratory of Lingnan Modern Agriculture Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry College of Animal Science South China Agricultural University Guangzhou China
| | - Qingyan Jiang
- Guangdong Laboratory of Lingnan Modern Agriculture Guangdong Province Key Laboratory of Animal Nutritional Regulation and National Engineering Research Center for Breeding Swine Industry College of Animal Science South China Agricultural University Guangzhou China
| |
Collapse
|
15
|
Kuwahara Y, Yoshizaki K, Nishida H, Kamishina H, Maeda S, Takano K, Fujita N, Nishimura R, Jo JI, Tabata Y, Akiyoshi H. Extracellular Vesicles Derived From Canine Mesenchymal Stromal Cells in Serum Free Culture Medium Have Anti-inflammatory Effect on Microglial Cells. Front Vet Sci 2021; 8:633426. [PMID: 33996963 PMCID: PMC8113404 DOI: 10.3389/fvets.2021.633426] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 03/24/2021] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) have been used as cell sources for treating dogs with naturally-occurring diseases. Extracellular vesicles (EVs) derived from MSCs are now recognized as pivotal to modulating the immune response and supporting tissue repair. Manufacture of MSC-EVs for clinical application mandates removal of the xeno-proteins, including fetal bovine serum. The objective of this study was to examine whether canine MSCs survived and secreted EVs in serum-free medium (SFM) conditions and to assess the immunomodulatory effect of EVs in vitro. Canine MSCs were found to survive and secrete EVs under SFM conditions. The surface markers of MSCs in the SFM were similar to MSCs in complete culture medium. Canine MSC-EVs had a diameter of ~300 nm and were positive for EV markers. MSC-derived EVs from the serum-free condition reduced the levels of IL-1β by BV-2 cells in response to LPS stimulation. These results warrant further studies of the use of SFM for producing EVs derived from canine MSCs.
Collapse
Affiliation(s)
- Yukina Kuwahara
- Joint Department of Veterinary Medicine, The United Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan
| | - Karin Yoshizaki
- Department of Veterinary Surgery, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Izumisano, Japan
| | - Hidetaka Nishida
- Department of Veterinary Surgery, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Izumisano, Japan
| | - Hiroaki Kamishina
- Joint Department of Veterinary Medicine, The United Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan
| | - Sadatoshi Maeda
- Joint Department of Veterinary Medicine, The United Graduate School of Veterinary Sciences, Gifu University, Gifu, Japan
| | - Katsura Takano
- Department of Integrative Physiology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Izumisano, Japan
| | - Naoki Fujita
- Department of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Ryohei Nishimura
- Department of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Jun-Ichiro Jo
- Laboratory of Biomaterials, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Yasuhiko Tabata
- Laboratory of Biomaterials, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Hideo Akiyoshi
- Department of Veterinary Surgery, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Izumisano, Japan
| |
Collapse
|
16
|
Novais A, Chatzopoulou E, Chaussain C, Gorin C. The Potential of FGF-2 in Craniofacial Bone Tissue Engineering: A Review. Cells 2021; 10:932. [PMID: 33920587 PMCID: PMC8073160 DOI: 10.3390/cells10040932] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/10/2021] [Accepted: 04/15/2021] [Indexed: 12/21/2022] Open
Abstract
Bone is a hard-vascularized tissue, which renews itself continuously to adapt to the mechanical and metabolic demands of the body. The craniofacial area is prone to trauma and pathologies that often result in large bone damage, these leading to both aesthetic and functional complications for patients. The "gold standard" for treating these large defects is autologous bone grafting, which has some drawbacks including the requirement for a second surgical site with quantity of bone limitations, pain and other surgical complications. Indeed, tissue engineering combining a biomaterial with the appropriate cells and molecules of interest would allow a new therapeutic approach to treat large bone defects while avoiding complications associated with a second surgical site. This review first outlines the current knowledge of bone remodeling and the different signaling pathways involved seeking to improve our understanding of the roles of each to be able to stimulate or inhibit them. Secondly, it highlights the interesting characteristics of one growth factor in particular, FGF-2, and its role in bone homeostasis, before then analyzing its potential usefulness in craniofacial bone tissue engineering because of its proliferative, pro-angiogenic and pro-osteogenic effects depending on its spatial-temporal use, dose and mode of administration.
Collapse
Affiliation(s)
- Anita Novais
- Pathologies, Imagerie et Biothérapies Orofaciales, Université de Paris, URP2496, 1 rue Maurice Arnoux, 92120 Montrouge, France; (A.N.); (E.C.); (C.C.)
- AP-HP Département d’Odontologie, Services d’odontologie, GH Pitié Salpêtrière, Henri Mondor, Paris Nord, Hôpital Rothschild, Paris, France
| | - Eirini Chatzopoulou
- Pathologies, Imagerie et Biothérapies Orofaciales, Université de Paris, URP2496, 1 rue Maurice Arnoux, 92120 Montrouge, France; (A.N.); (E.C.); (C.C.)
- AP-HP Département d’Odontologie, Services d’odontologie, GH Pitié Salpêtrière, Henri Mondor, Paris Nord, Hôpital Rothschild, Paris, France
- Département de Parodontologie, Université de Paris, UFR Odontologie-Garancière, 75006 Paris, France
| | - Catherine Chaussain
- Pathologies, Imagerie et Biothérapies Orofaciales, Université de Paris, URP2496, 1 rue Maurice Arnoux, 92120 Montrouge, France; (A.N.); (E.C.); (C.C.)
- AP-HP Département d’Odontologie, Services d’odontologie, GH Pitié Salpêtrière, Henri Mondor, Paris Nord, Hôpital Rothschild, Paris, France
| | - Caroline Gorin
- Pathologies, Imagerie et Biothérapies Orofaciales, Université de Paris, URP2496, 1 rue Maurice Arnoux, 92120 Montrouge, France; (A.N.); (E.C.); (C.C.)
- AP-HP Département d’Odontologie, Services d’odontologie, GH Pitié Salpêtrière, Henri Mondor, Paris Nord, Hôpital Rothschild, Paris, France
| |
Collapse
|
17
|
Proteomic Profiling of the First Human Dental Pulp Mesenchymal Stem/Stromal Cells from Carbonic Anhydrase II Deficiency Osteopetrosis Patients. Int J Mol Sci 2020; 22:ijms22010380. [PMID: 33396517 PMCID: PMC7795265 DOI: 10.3390/ijms22010380] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 12/26/2020] [Accepted: 12/28/2020] [Indexed: 12/30/2022] Open
Abstract
Osteopetrosis is a hereditary disorder characterized by sclerotic, thick, weak, and brittle bone. The biological behavior of mesenchymal cells obtained from osteopetrosis patients has not been well-studied. Isolated mesenchymal stem/stromal cells from dental pulp (DP-MSSCs) of recently extracted deciduous teeth from osteopetrosis (OP) patients and healthy controls (HCs) were compared. We evaluated whether the dental pulp of OP patients has a population of MSSCs with similar multilineage differentiation capability to DP-MSSCs of healthy subjects. Stem/progenitor cells were characterized using immunohistochemistry, flow cytometry, and proteomics. Our DP-MSSCs were strongly positive for CD44, CD73, CD105, and CD90. DP-MSSCs obtained from HC subjects and OP patients showed similar patterns of proliferation and differentiation as well as gene expression. Proteomic analysis identified 1499 unique proteins with 94.3% similarity in global protein fingerprints of HCs and OP patients. Interestingly, we observed subtle differences in expressed proteins of osteopetrosis disease-related in pathways, including MAPK, ERK 1/2, PI3K, and integrin, rather than in the stem cell signaling network. Our findings of similar protein expression signatures in DP-MSSCs of HC and OP patients are of paramount interest, and further in vivo validation study is needed. There is the possibility that OP patients could have their exfoliating deciduous teeth banked for future use in regenerative dentistry.
Collapse
|
18
|
Luo L, Zhang Y, Chen H, Hu F, Wang X, Xing Z, Albashari AA, Xiao J, He Y, Ye Q. Effects and mechanisms of basic fibroblast growth factor on the proliferation and regenerative profiles of cryopreserved dental pulp stem cells. Cell Prolif 2020; 54:e12969. [PMID: 33332682 PMCID: PMC7848956 DOI: 10.1111/cpr.12969] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 11/25/2020] [Accepted: 11/28/2020] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVES Various factors could interfere the biological performance of DPSCs during post-thawed process. Yet, little has been known about optimization of the recovery medium for DPSCs. Thus, our study aimed to explore the effects of adding recombinant bFGF on DPSCs after 3-month cryopreservation as well as the underlying mechanisms. MATERIALS AND METHODS DPSCs were extracted from impacted third molars and purified by MACS. The properties of CD146+ DPSCs (P3) were identified by CCK-8 and flow cytometry. After cryopreservation for 3 months, recovered DPSCs (P4) were immediately supplied with a series of bFGF and analysed cellular proliferation by CCK-8. Then, the optimal dosage of bFGF was determined to further identify apoptosis and TRPC1 channel through Western blot. The succeeding passage (P5) from bFGF pre-treated DPSCs was cultivated in bFGF-free culture medium, cellular proliferation and stemness were verified, and pluripotency was analysed by neurogenic, osteogenic and adipogenic differentiation. RESULTS It is found that adding 20 ng/mL bFGF in culture medium could significantly promote the proliferation of freshly thawed DPSCs (P4) through suppressing apoptosis, activating ERK pathway and up-regulating TRPC1. Such proliferative superiority could be inherited to the succeeding passage (P5) from bFGF pre-stimulated DPSCs, meanwhile, stemness and pluripotency have not been compromised. CONCLUSIONS This study illustrated a safe and feasible cell culture technique to rapidly amplify post-thawed DPSCs with robust regenerative potency, which brightening the future of stem cells banking and tissue engineering.
Collapse
Affiliation(s)
- Lihua Luo
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Yanni Zhang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Hongyu Chen
- Department of Stomatology, Ningbo Women and Children Hospital, Ningbo, China
| | - Fengting Hu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Xiaoyan Wang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | - Zhenjie Xing
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China
| | | | - Jian Xiao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yan He
- Laboratory of Regenerative Medicine, Tianyou Hospital, Wuhan University of Science and Technology, Wuhan, China
| | - Qingsong Ye
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, China.,Center of Regenerative Medicine, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
19
|
Mesenchymal Stem/Progenitor Cells: The Prospect of Human Clinical Translation. Stem Cells Int 2020; 2020:8837654. [PMID: 33953753 PMCID: PMC8063852 DOI: 10.1155/2020/8837654] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/19/2020] [Accepted: 07/20/2020] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem/progenitor cells (MSCs) are key players in regenerative medicine, relying principally on their differentiation/regeneration potential, immunomodulatory properties, paracrine effects, and potent homing ability with minimal if any ethical concerns. Even though multiple preclinical and clinical studies have demonstrated remarkable properties for MSCs, the clinical applicability of MSC-based therapies is still questionable. Several challenges exist that critically hinder a successful clinical translation of MSC-based therapies, including but not limited to heterogeneity of their populations, variability in their quality and quantity, donor-related factors, discrepancies in protocols for isolation, in vitro expansion and premodification, and variability in methods of cell delivery, dosing, and cell homing. Alterations of MSC viability, proliferation, properties, and/or function are also affected by various drugs and chemicals. Moreover, significant safety concerns exist due to possible teratogenic/neoplastic potential and transmission of infectious diseases. Through the current review, we aim to highlight the major challenges facing MSCs' human clinical translation and shed light on the undergoing strategies to overcome them.
Collapse
|
20
|
Hasan MR, Takatalo M, Ma H, Rice R, Mustonen T, Rice DP. RAB23 coordinates early osteogenesis by repressing FGF10-pERK1/2 and GLI1. eLife 2020; 9:55829. [PMID: 32662771 PMCID: PMC7423339 DOI: 10.7554/elife.55829] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 07/13/2020] [Indexed: 12/20/2022] Open
Abstract
Mutations in the gene encoding Ras-associated binding protein 23 (RAB23) cause Carpenter Syndrome, which is characterized by multiple developmental abnormalities including polysyndactyly and defects in skull morphogenesis. To understand how RAB23 regulates skull development, we generated Rab23-deficient mice that survive to an age where skeletal development can be studied. Along with polysyndactyly, these mice exhibit premature fusion of multiple sutures resultant from aberrant osteoprogenitor proliferation and elevated osteogenesis in the suture. FGF10-driven FGFR1 signaling is elevated in Rab23-/-sutures with a consequent imbalance in MAPK, Hedgehog signaling and RUNX2 expression. Inhibition of elevated pERK1/2 signaling results in the normalization of osteoprogenitor proliferation with a concomitant reduction of osteogenic gene expression, and prevention of craniosynostosis. Our results suggest a novel role for RAB23 as an upstream negative regulator of both FGFR and canonical Hh-GLI1 signaling, and additionally in the non-canonical regulation of GLI1 through pERK1/2.
Collapse
Affiliation(s)
- Md Rakibul Hasan
- Craniofacial Development and Malformations research group, Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - Maarit Takatalo
- Craniofacial Development and Malformations research group, Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - Hongqiang Ma
- Craniofacial Development and Malformations research group, Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - Ritva Rice
- Craniofacial Development and Malformations research group, Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - Tuija Mustonen
- Craniofacial Development and Malformations research group, Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland
| | - David Pc Rice
- Craniofacial Development and Malformations research group, Orthodontics, Oral and Maxillofacial Diseases, University of Helsinki, Helsinki, Finland.,Oral and Maxillofacial Diseases, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
21
|
Son J, Tae JY, Min SK, Ko Y, Park JB. Fibroblast growth factor-4 maintains cellular viability while enhancing osteogenic differentiation of stem cell spheroids in part by regulating RUNX2 and BGLAP expression. Exp Ther Med 2020; 20:2013-2020. [PMID: 32782511 PMCID: PMC7401302 DOI: 10.3892/etm.2020.8951] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 04/29/2020] [Indexed: 12/14/2022] Open
Abstract
Fibroblast growth factors (FGFs) are growth factors that were initially identified as proteins that stimulate fibroblast proliferation. The aim of the present study was to examine the effects of FGF-4 on the morphology, cellular viability and osteogenic differentiation of stem cell spheroids. Stem cell spheroids were generated using concave microwells in the presence of FGF-4 at concentrations of 0, 50, 100 and 200 ng/ml. Cellular viability was qualitatively assessed by a fluorometric live/dead assay using a microscope and quantitatively determined by using Cell Counting Kit-8. Furthermore, alkaline phosphatase activity and calcium deposition were determined to assess osteogenic differentiation. Reverse transcription-quantitative PCR (RT-qPCR) was performed to evaluate the mRNA expression levels of Runt-related transcription factor 2 (RUNX2) and bone γ-carboxyglutamate protein (BGLAP). Spheroidal shapes were achieved in the microwells on day 1 and a significant increase in the spheroid diameter was observed in the 200 ng/ml FGF-4 group compared with the control group on day 1 (P<0.05). The results regarding viability using Cell Counting Kit-8 in the presence of FGF-4 at 50, 100 and 200 ng/ml at day 1 were 98.0±2.5, 106.2±17.6 and 99.5±6.0%, respectively, when normalized to the control group (P>0.05). Furthermore, the alkaline phosphatase activity was significantly elevated in the 200 ng/ml group, when compared with the control group. The RT-qPCR results demonstrated that the mRNA expression levels of RUNX2 and BGLAP were significantly increased at 200 ng/ml. Therefore, the present results suggested that the application of FGF-4 maintained cellular viability while enhancing the osteogenic differentiation of stem cell spheroids, at least partially by regulating RUNX2 and BGLAP expression levels.
Collapse
Affiliation(s)
- Juwan Son
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jae-Yong Tae
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sae Kyung Min
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Youngkyung Ko
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jun-Beom Park
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
22
|
Jiang X, Wu F, Xu Y, Yan JX, Wu YD, Li SH, Liao X, Liang JX, Li ZH, Liu HW. A novel role of angiotensin II in epidermal cell lineage determination: Angiotensin II promotes the differentiation of mesenchymal stem cells into keratinocytes through the p38 MAPK, JNK and JAK2 signalling pathways. Exp Dermatol 2020; 28:59-65. [PMID: 30412649 DOI: 10.1111/exd.13837] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 10/14/2018] [Accepted: 10/28/2018] [Indexed: 12/23/2022]
Abstract
BACKGROUND Recent evidence suggests that angiotensin II (Ang II) plays a role in cutaneous wound healing. Mesenchymal stem cells (MSCs) are known as a rich source of cells that re-establish healed skin. However, the potential impact of Ang II on MSC differentiation into keratinocytes is still unknown. OBJECTIVE The present study was conducted to explore the effect of Ang II on the differentiation of bone marrow-derived MSCs (BM-MSCs) into keratinocytes. METHODS Bone marrow-derived MSCs were isolated from rat bone marrow and cultured. The expression of Ang II type 1 (AT1 ) and type 2 (AT2 ) receptors was examined by immunofluorescence staining. The differentiation of BM-MSCs into keratinocytes was investigated by flow cytometry or/and histological observation. RESULTS The BM-MSCs constitutively expressed both AT1 and AT2 receptors. The differentiation of BM-MSCs into keratinocytes was successfully induced. Interestingly, incubation of BM-MSCs with Ang II further promoted the differentiation of BM-MSCs into keratinocyte, which was abolished by pretreament with losartan, an AT1 receptor antagonist, but not by PD123319, an AT2 receptor antagonist. Moreover, the p38 mitogen-activated protein kinase (MAPK) inhibitor SB203580, the c-Jun N-terminal kinase (JNK) inhibitor SP600125 and the Janus-activated kinase (JAK)2 inhibitor AG490 suppressed Ang II-induced differentiation of BM-MSCs into keratinocytes. The phosphoinositide-3 kinase (PI3K) inhibitor wortmannin and MEK1/2 inhibitor U0126 had no effect on BM-MSC differentiation into keratinocytes. CONCLUSIONS Our data demonstrated for the first time that Ang II plays a promotive role in the differentiation of BM-MSC into keratinocytes through the AT1 receptor, and that the p38 MAPK, JNK and JAK2 signalling pathways are involved in this process.
Collapse
Affiliation(s)
- Xiao Jiang
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China.,Innovative Technology Research Institute of Tissue Repair and Regeneration, Key Laboratory of Regenerative Medicine, Ministry of Education, Guangzhou, Guangdong Province, China
| | - Fan Wu
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China.,Innovative Technology Research Institute of Tissue Repair and Regeneration, Key Laboratory of Regenerative Medicine, Ministry of Education, Guangzhou, Guangdong Province, China
| | - Yuan Xu
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China.,Innovative Technology Research Institute of Tissue Repair and Regeneration, Key Laboratory of Regenerative Medicine, Ministry of Education, Guangzhou, Guangdong Province, China
| | - Jian-Xin Yan
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China.,Innovative Technology Research Institute of Tissue Repair and Regeneration, Key Laboratory of Regenerative Medicine, Ministry of Education, Guangzhou, Guangdong Province, China
| | - Yin-Di Wu
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China.,Innovative Technology Research Institute of Tissue Repair and Regeneration, Key Laboratory of Regenerative Medicine, Ministry of Education, Guangzhou, Guangdong Province, China
| | - Sheng-Hong Li
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China.,Innovative Technology Research Institute of Tissue Repair and Regeneration, Key Laboratory of Regenerative Medicine, Ministry of Education, Guangzhou, Guangdong Province, China
| | - Xuan Liao
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China.,Innovative Technology Research Institute of Tissue Repair and Regeneration, Key Laboratory of Regenerative Medicine, Ministry of Education, Guangzhou, Guangdong Province, China
| | - Jun-Xian Liang
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China.,Innovative Technology Research Institute of Tissue Repair and Regeneration, Key Laboratory of Regenerative Medicine, Ministry of Education, Guangzhou, Guangdong Province, China
| | - Ze-Hua Li
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China.,Innovative Technology Research Institute of Tissue Repair and Regeneration, Key Laboratory of Regenerative Medicine, Ministry of Education, Guangzhou, Guangdong Province, China
| | - Hong-Wei Liu
- Department of Plastic Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong Province, China.,Innovative Technology Research Institute of Tissue Repair and Regeneration, Key Laboratory of Regenerative Medicine, Ministry of Education, Guangzhou, Guangdong Province, China
| |
Collapse
|
23
|
Cheng Y, Lin K, Young T, Cheng N. The influence of fibroblast growth factor 2 on the senescence of human adipose-derived mesenchymal stem cells during long-term culture. Stem Cells Transl Med 2020; 9:518-530. [PMID: 31840944 PMCID: PMC7103622 DOI: 10.1002/sctm.19-0234] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 11/25/2019] [Indexed: 12/22/2022] Open
Abstract
Adipose-derived mesenchymal stem cells (ASCs) exhibit great potential in regenerative medicine, and in vitro expansion is frequently necessary to obtain a sufficient number of ASCs for clinical use. Fibroblast growth factor 2 (FGF2) is a common supplement in the ASC culture medium to enhance cell proliferation. To achieve clinical applicability of ASC-based products, prolonged culture of ASCs is sometimes required to obtain sufficient quantity of ASCs. However, the effect of FGF2 on ASCs during prolonged culture has not been previously determined. In this study, ASCs were subjected to prolonged in vitro culture with or without FGF2. FGF2 maintained the small cell morphology and expedited proliferation kinetics in early ASC passages. After prolonged in vitro expansion, FGF2-treated ASCs exhibited increased cell size, arrested cell proliferation, and increased cellular senescence relative to the control ASCs. We observed an upregulation of FGFR1c and enhanced expression of downstream STAT3 in the initial passages of FGF2-treated ASCs. The application of an FGFR1 or STAT3 inhibitor effectively blocked the enhanced proliferation of ASCs induced by FGF2 treatment. FGFR1c upregulation and enhanced STAT3 expression were lost in the later passages of FGF2-treated ASCs, suggesting that the continuous stimulation of FGF2 becomes ineffective because of the refractory downstream FGFR1 and the STAT3 signaling pathway. In addition, no evidence of tumorigenicity was noted in vitro and in vivo after prolonged expansion of FGF2-cultured ASCs. Our data indicate that ASCs have evolved a STAT3-dependent response to continuous FGF2 stimulation which promotes the initial expansion but limits their long-term proliferation.
Collapse
Affiliation(s)
- Yin Cheng
- Department of SurgeryNational Taiwan University Hospital and College of MedicineTaipeiTaiwan
| | - Kai‐Hsuan Lin
- Department of SurgeryNational Taiwan University Hospital and College of MedicineTaipeiTaiwan
| | - Tai‐Horng Young
- Department of Biomedical Engineering, College of Medicine and College of EngineeringNational Taiwan UniversityTaipeiTaiwan
| | - Nai‐Chen Cheng
- Department of SurgeryNational Taiwan University Hospital and College of MedicineTaipeiTaiwan
- Research Center for Developmental Biology and Regenerative MedicineNational Taiwan UniversityTaipeiTaiwan
| |
Collapse
|
24
|
Ji M, Xu K, Zhang D, Chen T, Shen L, Wu W, Zhang J. Adipose-Tissue-Specific Expression of Pig ApoR Protects Mice from Diet-Induced Obesity. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:2256-2262. [PMID: 31927923 DOI: 10.1021/acs.jafc.9b06995] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Fat deposition is one of the most important economic traits of pigs. Decreasing the subcutaneous fat and increasing the intramuscular fat are believed to be an effective way to improve pork quality, which is one of the main goals of pig breeding. Identifying key genes that control porcine lipid metabolism is essential for achieving this goal. Apolipoprotein R (apoR) was identified as the crucial molecule in the process of pig adipose reduction by clenbuterol. In this study, transgenic mice with adipose-tissue-specific overexpression of pig apoR (apoR mice) were constructed. The apoR mice gained less weight than wild-type (WT) mice after 18 weeks of feeding a high-fat diet. A comparison of organs between the two genotypes revealed that the weight of white adipose tissue, including inguinal and epididymal fat tissue, was significantly decreased and the weight of liver tissue was increased in apoR mice compared with WT mice. Glucose and insulin intolerance tests showed that the glucose metabolism of apoR mice was similar to that of WT mice. Histological staining proved that the adipocytes of apoR mice had a reduced average size, and gene expression analysis indicated that lipolysis in the adipose tissue of apoR mice was enhanced. Finally, the primary culture of inguinal adipocytes revealed that apoR promotes lipolysis via the Erk1/2 pathway. Taken together, the results indicate that adipose-tissue-specific expression of pig apoR protects mice from diet-induced obesity by enhancing lipolysis.
Collapse
Affiliation(s)
- Miao Ji
- College of Biological, Chemical Sciences and Engineering , Jiaxing University , Jiaxing 314001 , China
- College of Agronomy and Biotechnology , Hebei Normal University of Science and Technology , Qinhuangdao 066000 , China
| | - Ke Xu
- College of Biological, Chemical Sciences and Engineering , Jiaxing University , Jiaxing 314001 , China
- College of Agronomy and Biotechnology , Hebei Normal University of Science and Technology , Qinhuangdao 066000 , China
| | - Dawei Zhang
- College of Biological, Chemical Sciences and Engineering , Jiaxing University , Jiaxing 314001 , China
| | - Tingting Chen
- Jiaxing Maternal and Child Health Care Hospital , Jiaxing 314001 , China
| | - Liangcai Shen
- College of Agronomy and Biotechnology , Hebei Normal University of Science and Technology , Qinhuangdao 066000 , China
| | - Wenjing Wu
- College of Biological, Chemical Sciences and Engineering , Jiaxing University , Jiaxing 314001 , China
| | - Jin Zhang
- College of Biological, Chemical Sciences and Engineering , Jiaxing University , Jiaxing 314001 , China
| |
Collapse
|
25
|
Kim MH, Jung SY, Song KH, Park JI, Ahn J, Kim EH, Park JK, Hwang SG, Woo HJ, Song JY. A new FGFR inhibitor disrupts the TGF-β1-induced fibrotic process. J Cell Mol Med 2019; 24:830-840. [PMID: 31692229 PMCID: PMC6933341 DOI: 10.1111/jcmm.14793] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/11/2019] [Accepted: 09/23/2019] [Indexed: 01/06/2023] Open
Abstract
Pulmonary fibrosis (PF) is chronic and irreversible damage to the lung characterized by fibroblast activation and matrix deposition. Although recently approved novel anti‐fibrotic agents can improve the lung function and survival of patients with PF, the overall outcomes remain poor. In this study, a novel imidazopurine compound, 3‐(2‐chloro‐6‐fluorobenzyl)‐1,6,7‐trimethyl‐1H‐imidazo[2,1‐f]purine‐2,4(3H,8H)‐dione (IM‐1918), markedly inhibited transforming growth factor (TGF)‐β‐stimulated reporter activity and reduced the expression of representative fibrotic markers, such as connective tissue growth factor, fibronectin, collagen and α‐smooth muscle actin, on human lung fibroblasts. However, IM‐1918 neither decreased Smad‐2 and Smad‐3 nor affected p38MAPK and JNK. Instead, IM‐1918 reduced Akt and extracellular signal‐regulated kinase 1/2 phosphorylation increased by TGF‐β. Additionally, IM‐1918 inhibited the phosphorylation of fibroblast growth factor receptors 1 and 3. In a bleomycin‐induced murine lung fibrosis model, IM‐1918 profoundly reduced fibrotic areas and decreased collagen and α‐smooth muscle actin accumulation. These results suggest that IM‐1918 can be applied to treat lung fibrosis.
Collapse
Affiliation(s)
- Mi-Hyoung Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea.,Laboratory of Immunology, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Seung-Youn Jung
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Kyung-Hee Song
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Jeong-In Park
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Jiyeon Ahn
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Eun-Ho Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Jong Kuk Park
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Sang-Gu Hwang
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| | - Hee-Jong Woo
- Laboratory of Immunology, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | - Jie-Young Song
- Division of Radiation Biomedical Research, Korea Institute of Radiological & Medical Sciences, Seoul, Korea
| |
Collapse
|
26
|
Jung JS, Volk C, Marga C, Navarrete Santos A, Jung M, Rujescu D, Navarrete Santos A. Adipose-Derived Stem/Stromal Cells Recapitulate Aging Biomarkers and Show Reduced Stem Cell Plasticity Affecting Their Adipogenic Differentiation Capacity. Cell Reprogram 2019; 21:187-199. [PMID: 31298565 DOI: 10.1089/cell.2019.0010] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Stromal mesenchymal stem cells (MSCs) have the capability to self-renew and can differentiate into multiple cell types of the mesoderm germ layer, but their properties are affected by molecular aging mechanisms. MSCs can be obtained from adipose tissue termed as adipose-derived stem/stromal cells (ASCs) representing a promising tool for studying age-related diseases in detail. ASCs from young (16 weeks) and old (>108 weeks) rabbits were successfully isolated and propagated. ASCs showed the typical morphology and stained positive for CD105, Vimentin, Collagenase 1A, and negative for CD14, CD90, and CD73, demonstrating their mesenchymal origin. ASCs expressed MSC markers, including MYC, KLF4, CHD1, REST, and KAT6A, whereas pluripotency-related genes, such as NANOG, OCT4, and SOX2, were not expressed. Aged ASCs showed altered protein and mRNA levels of APOE, ATG7, FGF2, PTEN, and SIRT1. Adipogenic differentiation of old visceral ASCs was significantly decreased compared with young visceral ASCs. We successfully established rabbit ASC cultures representing an in vitro model for the analysis of stem cell aging mechanisms. ASCs, obtained from old female rabbits, showed age- and source-specific alteration due to aging of the donor. Stem cell plasticity was altered with age as shown by reduced adipogenic differentiation capacity.
Collapse
Affiliation(s)
- Juliane-Susanne Jung
- 1Department of Anatomy and Cell Biology, Psychotherapy, Psychosomatic Medicine, Martin Luther University Medical Faculty, Halle, Germany
| | - Christin Volk
- 1Department of Anatomy and Cell Biology, Psychotherapy, Psychosomatic Medicine, Martin Luther University Medical Faculty, Halle, Germany
| | - Christina Marga
- 1Department of Anatomy and Cell Biology, Psychotherapy, Psychosomatic Medicine, Martin Luther University Medical Faculty, Halle, Germany
| | - Alexander Navarrete Santos
- 2Center for Medical Basic Research, Psychotherapy, Psychosomatic Medicine, Martin Luther University Medical Faculty, Halle, Germany
| | - Matthias Jung
- 3Department of Psychiatry, Psychotherapy, Psychosomatic Medicine, Martin Luther University Medical Faculty, Halle, Germany
| | - Dan Rujescu
- 3Department of Psychiatry, Psychotherapy, Psychosomatic Medicine, Martin Luther University Medical Faculty, Halle, Germany
| | - Anne Navarrete Santos
- 1Department of Anatomy and Cell Biology, Psychotherapy, Psychosomatic Medicine, Martin Luther University Medical Faculty, Halle, Germany
| |
Collapse
|
27
|
Zhong H, Wu H, Bai H, Wang M, Wen J, Gong J, Miao M, Yuan F. Panax notoginseng saponins promote liver regeneration through activation of the PI3K/AKT/mTOR cell proliferation pathway and upregulation of the AKT/Bad cell survival pathway in mice. Altern Ther Health Med 2019; 19:122. [PMID: 31182089 PMCID: PMC6558887 DOI: 10.1186/s12906-019-2536-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 05/29/2019] [Indexed: 12/15/2022]
Abstract
Backgroud The regenerative capacity of the liver is crucial for the host to survive after serious hepatic injuries, tumor resection, or living donor liver transplantation. Panax notoginseng saponins (PNS) have been reported to exert protective effects during organ injuries. The present study aimed to evaluate the effect of PNS on liver regeneration(LR) and on injuries induced by partial hepatectomy (PH). Methods We performed 70% partial PH on C57BL/6 J mice treated with or without PNS. LR was estimated by liver weight/body weight, serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels and cell proliferation, and the related cellular signals were analyzed by Western blot. Results Different concentrations of PNS promoted hepatocyte proliferation in vitro. Mice in the PNS group showed higher liver/body weight ratios at 2 d and 7 d (P < 0.05) after PH and lower levels of serum ALT and AST (P < 0.05) compared to those of mice in the normal control (NC) group. Histological analysis showed that the expression of proliferating cell nuclear antigen(PCNA) at 2 d and 7 d after PH was significantly higher in the PNS group than in the NC group (P < 0.05). Mechanistically, the AKT/mTOR cell proliferation pathway and AKT/Bad cell survival pathway were activated by PNS, which accelerated hepatocyte proliferation and inhibited apoptosis (P < 0.05). Conclusions PNS promoted liver regeneration through activation of PI3K/AKT/mTOR and upregulated the AKT/Bad cell pathways in mice.
Collapse
|
28
|
Qin X, Li J, Sun J, Liu L, Chen D, Liu Y. Low shear stress induces ERK nuclear localization and YAP activation to control the proliferation of breast cancer cells. Biochem Biophys Res Commun 2019; 510:219-223. [DOI: 10.1016/j.bbrc.2019.01.065] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 01/12/2019] [Indexed: 01/29/2023]
|
29
|
Rajendran AK, Arisaka Y, Iseki S, Yui N. Sulfonated Polyrotaxane Surfaces with Basic Fibroblast Growth Factor Alter the Osteogenic Potential of Human Mesenchymal Stem Cells in Short-Term Culture. ACS Biomater Sci Eng 2019; 5:5652-5659. [DOI: 10.1021/acsbiomaterials.8b01343] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Arun Kumar Rajendran
- Section of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo 113-8549, Japan
| | - Yoshinori Arisaka
- Department of Organic Biomaterials, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062, Japan
| | - Sachiko Iseki
- Section of Molecular Craniofacial Embryology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo, Tokyo 113-8549, Japan
| | - Nobuhiko Yui
- Department of Organic Biomaterials, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda, Tokyo 101-0062, Japan
| |
Collapse
|
30
|
Abstract
Fibroblast growth factors (FGFs) and their receptors (FGFRs) are expressed throughout all stages of skeletal development. In the limb bud and in cranial mesenchyme, FGF signaling is important for formation of mesenchymal condensations that give rise to bone. Once skeletal elements are initiated and patterned, FGFs regulate both endochondral and intramembranous ossification programs. In this chapter, we review functions of the FGF signaling pathway during these critical stages of skeletogenesis, and explore skeletal malformations in humans that are caused by mutations in FGF signaling molecules.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, United States.
| | - Pierre J Marie
- UMR-1132 Inserm (Institut national de la Santé et de la Recherche Médicale) and University Paris Diderot, Sorbonne Paris Cité, Hôpital Lariboisière, Paris, France
| |
Collapse
|
31
|
Vidovic-Zdrilic I, Vining K, Vijaykumar A, Kalajzic I, Mooney D, Mina M. FGF2 Enhances Odontoblast Differentiation by αSMA + Progenitors In Vivo. J Dent Res 2018; 97:1170-1177. [PMID: 29649366 PMCID: PMC6169028 DOI: 10.1177/0022034518769827] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The goal of this study was to examine the effects of early and limited exposure of perivascular cells expressing α (αSMA) to fibroblast growth factor 2 (FGF2) in vivo. We performed in vivo fate mapping by inducible Cre-loxP and experimental pulp injury in molars to induce reparative dentinogenesis. Our results demonstrate that early delivery of exogenous FGF2 to exposed pulp led to proliferative expansion of αSMA-tdTomato+ cells and their accelerated differentiation into odontoblasts. In vivo lineage-tracing experiments showed that the calcified bridge/reparative dentin in FGF2-treated pulps were lined with an increased number of Dspp+ odontoblasts and devoid of BSP+ osteoblasts. The increased number of odontoblasts derived from αSMA-tdTomato+ cells and the formation of reparative dentin devoid of osteoblasts provide in vivo evidence for the stimulatory effects of FGF signaling on odontoblast differentiation from early progenitors in dental pulp.
Collapse
Affiliation(s)
- I. Vidovic-Zdrilic
- Departments of Craniofacial Sciences, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - K.H. Vining
- John A. Paulson School of Engineering and Applied Sciences and Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, USA
| | - A. Vijaykumar
- Departments of Craniofacial Sciences, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - I. Kalajzic
- Departments of Reconstructive Sciences, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT, USA
| | - D.J. Mooney
- John A. Paulson School of Engineering and Applied Sciences and Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, USA
| | - M. Mina
- Departments of Craniofacial Sciences, School of Dental Medicine, University of Connecticut Health Center, Farmington, CT, USA
| |
Collapse
|
32
|
Luo Y, Mohsin A, Xu C, Wang Q, Hang H, Zhuang Y, Chu J, Guo M. Co-culture with TM4 cells enhances the proliferation and migration of rat adipose-derived mesenchymal stem cells with high stemness. Cytotechnology 2018; 70:1409-1422. [PMID: 30032334 DOI: 10.1007/s10616-018-0235-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 07/07/2018] [Indexed: 01/16/2023] Open
Abstract
The proliferation and migration of mesenchymal stem cells (MSCs) are the efficiency determinants in MSCs transplant therapy. Sertoli cells considered as "nurse cell" possesses the ability to enhance the proliferation and migration of umbilical cord mesenchymal stem cells (UCMSCs). However, no reports about TM4 cells' effect on the proliferation and migration of adipose tissue-derived mesenchymal stem cells (ADSCs) have been found until at present research work. Therefore, this study investigates the effect of TM4 cells on the proliferation and migration of ADSCs. We found that the performance of proliferation and migration of ADSCs were improved significantly while maintaining their stemness and reducing their apoptosis rate. After co-culturing with TM4 cells, the co-cultured ADSCs demonstrated higher proportion of synthetic phase (S) cells and colony-forming units-fibroblastic (CFU-F) number, lower proportion of sub-G1 phase cells and enhanced osteogenic and adipogenic differentiation ability. Moreover, results confirmed the higher multiple proteins involved in cell proliferation and migration including expression of the phospho-Akt, mdm2, pho-CDC2, cyclin D1 CXCR4, MMP-2, as well as phospho-p44 MAPK and phospho-p38 MAPK in co-cultured ADSCs. Furthermore, the process of TM4 cells promoting the proliferation of ADSCs was significantly inhibited by the administration of the PI3K/AKT inhibitor LY294002. Obtained results indicated that TM4 cells through MAPK/ERK1/2, MAPK/p-38 and PI3K/Akt pathways influence the proliferation and migration of ADSCs. These findings indicated that TM4 cells were found effective in promoting stemness and migration of ADSCs, that proves adopted co-culturing technique as an efficient approach to obtain ADSCs in transplantation therapy.
Collapse
Affiliation(s)
- Yanxia Luo
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai, 200237, China
| | - Ali Mohsin
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai, 200237, China
| | - Chenze Xu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai, 200237, China
| | - Qizheng Wang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai, 200237, China
| | - Haifeng Hang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai, 200237, China. .,Engineering Research Centre of Processes System, Ministry of Education, East China University of Science and Technology, 130 Meilong Rd., Shanghai, 200237, China.
| | - Yingping Zhuang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai, 200237, China.,Engineering Research Centre of Processes System, Ministry of Education, East China University of Science and Technology, 130 Meilong Rd., Shanghai, 200237, China
| | - Ju Chu
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai, 200237, China
| | - Meijin Guo
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, 130 Meilong Rd., Shanghai, 200237, China. .,Engineering Research Centre of Processes System, Ministry of Education, East China University of Science and Technology, 130 Meilong Rd., Shanghai, 200237, China.
| |
Collapse
|
33
|
Lang S, Herrmann M, Pfeifer C, Brockhoff G, Zellner J, Nerlich M, Angele P, Prantl L, Gehmert S, Loibl M. Leukocyte-reduced platelet-rich plasma stimulates the in vitro proliferation of adipose-tissue derived mesenchymal stem cells depending on PDGF signaling. Clin Hemorheol Microcirc 2018; 67:183-196. [PMID: 28922143 DOI: 10.3233/ch-170246] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND Platelet-rich Plasma (PRP) is suggested as xenoprotein-free cell-culture medium replacement for animal-derived supplements. OBJECTIVE The aim of this study was to investigate PRP-triggered signaling in adipose derived mesenchymal stem cells (ASCs). METHODS PRP was obtained from 4 male patients. We incubated ASCs in α-MEM with different Platelet derived growth factor (PDGF) subtypes or 10% or 20% pooled PRP or 20% fetal calf serum (FCS) prior to determination of the S-phase fraction (SPF). To investigate the influence of PDGF signaling on ASCs, PDGF receptor β inhibitor was added, and protein expression of ASCs was measured. RESULTS ASCs exposed to 20% PRP, PDGF-AB and - BB demonstrated significant higher SPF in comparison to PDGF-AA and 20% FCS after 48 hours (all P < 0.05). PDGF receptor β inhibition diminished the PRP-induced SPF increase of ASCs significantly after 48 hours (P < 0.01). ASCs with PDGF receptor β inhibition showed significant higher PDGF receptor β and significant lower c-MYC expression compared to untreated cells in presence of 20% PRP after 48 hours (both P < 0.05). CONCLUSIONS The proliferation promoting effect of PRP on ASCs is mediated by PDGF signaling and is associated with c-MYC overexpression.
Collapse
Affiliation(s)
- Siegmund Lang
- Department of Trauma Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Marietta Herrmann
- AO Research Institute, Davos, Switzerland.,IZKF Group Tissue Regeneration in Musculoskeletal Diseases, Orthopedic Center for Musculoskeletal Research, University Würzburg, Würzburg, Germany
| | - Christian Pfeifer
- Department of Trauma Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Gero Brockhoff
- Department of Obstetrics and Gynecology, University Medical Center Regensburg, Regensburg, Germany
| | - Johannes Zellner
- Department of Trauma Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Michael Nerlich
- Department of Trauma Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Peter Angele
- Department of Trauma Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Lukas Prantl
- Center of Plastic, Hand and Reconstructive Surgery, University Medical Center Regensburg, Regensburg, Germany
| | - Sebastian Gehmert
- Department of Trauma Surgery, University Medical Center Regensburg, Regensburg, Germany.,Center of Plastic, Hand and Reconstructive Surgery, University Medical Center Regensburg, Regensburg, Germany.,Department of Orthopedic Surgery, University Hospital Basel, Basel, Switzerland
| | - Markus Loibl
- Department of Trauma Surgery, University Medical Center Regensburg, Regensburg, Germany
| |
Collapse
|
34
|
Li J, Li SH, Wu J, Weisel RD, Yao A, Stanford WL, Liu SM, Li RK. Young Bone Marrow Sca-1 Cells Rejuvenate the Aged Heart by Promoting Epithelial-to-Mesenchymal Transition. Am J Cancer Res 2018; 8:1766-1781. [PMID: 29556355 PMCID: PMC5858499 DOI: 10.7150/thno.22788] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 01/13/2018] [Indexed: 01/13/2023] Open
Abstract
Background: To improve the regenerative capacity of aged individuals, we reconstituted bone marrow (BM) of aged mice with young Sca-1 cells, which repopulated cardiac progenitors and prevented cardiac dysfunction after a myocardial infarction (MI). However, the mechanisms involved were incompletely elucidated. This study aimed to investigate whether young, highly regenerative BM Sca-1 cells exert their cardio-protective effects on the aged heart through reactivation of the epithelial-to-mesenchymal transition (EMT) process. Methods:In vitro, BM Sca-1 cells were co-cultured with epicardial-derived cells (EPDCs) under hypoxia condition; mRNA and protein levels of EMT genes were measured along with cellular proliferation and migration. In vivo, BM Sca-1+ or Sca-1- cells from young mice (2-3 months) were transplanted into lethally-irradiated old mice (20-22 months) to generate chimeras. In addition, Sca-1 knockout (KO) mice were reconstituted with wild type (WT) BM Sca-1+ cells. The effects of BM Sca-1 cell on EMT reactivation and improvement of cardiac function after MI were evaluated. Results:In vitro, BM Sca-1+ cells increased EPDC proliferation, migration, and EMT relative to Sca-1- cells and these effects were inhibited by a TGF-β blocker. In vivo, more young BM Sca-1+ than Sca-1- cells homed to the epicardium and induced greater host EPDC proliferation, migration, and EMT after MI. Furthermore, reconstitution of Sca-1 KO mice with WT Sca-1+ cells was associated with the reactivation of EMT and improved cardiac function after MI. Conclusions: Young BM Sca-1+ cells improved cardiac regeneration through promoting EPDC proliferation, migration and reactivation of EMT via the TGF-β signaling pathway.
Collapse
Affiliation(s)
- Jiao Li
- Department of Cardiology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China,Toronto General Research Institute, Division of Cardiovascular Surgery, University Health Network, Toronto, Canada,Division of Cardiac Surgery, Department of Surgery, University of Toronto; Toronto, Canada
| | - Shu-Hong Li
- Toronto General Research Institute, Division of Cardiovascular Surgery, University Health Network, Toronto, Canada
| | - Jun Wu
- Toronto General Research Institute, Division of Cardiovascular Surgery, University Health Network, Toronto, Canada
| | - Richard D. Weisel
- Toronto General Research Institute, Division of Cardiovascular Surgery, University Health Network, Toronto, Canada,Division of Cardiac Surgery, Department of Surgery, University of Toronto; Toronto, Canada
| | - Alina Yao
- Toronto General Research Institute, Division of Cardiovascular Surgery, University Health Network, Toronto, Canada
| | - William L. Stanford
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Department of Cellular and Molecular Medicine, University of Ottawa
| | - Shi-Ming Liu
- Department of Cardiology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China,✉ Corresponding author: Shi-Ming Liu, MD, Department of Cardiology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China. Tel: 86-020-34153522; Fax: 86-20-3415-3709; and Ren-Ke Li, MD, PhD, Toronto Medical Discovery Tower, Room 3-702, 101 College Street, Toronto, Ontario, Canada M5G 1L7. Tel: 1-416-581-7492; Fax: 1-416-581-7493;
| | - Ren-Ke Li
- Toronto General Research Institute, Division of Cardiovascular Surgery, University Health Network, Toronto, Canada,Division of Cardiac Surgery, Department of Surgery, University of Toronto; Toronto, Canada,✉ Corresponding author: Shi-Ming Liu, MD, Department of Cardiology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China. Tel: 86-020-34153522; Fax: 86-20-3415-3709; and Ren-Ke Li, MD, PhD, Toronto Medical Discovery Tower, Room 3-702, 101 College Street, Toronto, Ontario, Canada M5G 1L7. Tel: 1-416-581-7492; Fax: 1-416-581-7493;
| |
Collapse
|
35
|
Ikpegbu E, Basta L, Clements DN, Fleming R, Vincent TL, Buttle DJ, Pitsillides AA, Staines KA, Farquharson C. FGF-2 promotes osteocyte differentiation through increased E11/podoplanin expression. J Cell Physiol 2018; 233:5334-5347. [PMID: 29215722 PMCID: PMC5900964 DOI: 10.1002/jcp.26345] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 11/29/2017] [Indexed: 01/15/2023]
Abstract
E11/podoplanin is critical in the early stages of osteoblast‐to‐osteocyte transitions (osteocytogenesis), however, the upstream events which regulate E11 expression are unknown. The aim of this study was to examine the effects of FGF‐2 on E11‐mediated osteocytogenesis and to reveal the nature of the underlying signaling pathways regulating this process. Exposure of MC3T3 osteoblast‐like cells and murine primary osteoblasts to FGF‐2 (10 ng/ml) increased E11 mRNA and protein expression (p < 0.05) after 4, 6, and 24 hr. FGF‐2 induced changes in E11 expression were also accompanied by significant (p < 0.01) increases in Phex and Dmp1 (osteocyte markers) expression and decreases in Col1a1, Postn, Bglap, and Alpl (osteoblast markers) expression. Immunofluorescent microscopy revealed that FGF‐2 stimulated E11 expression, facilitated the translocation of E11 toward the cell membrane, and subsequently promoted the formation of osteocyte‐like dendrites in MC3T3 and primary osteoblasts. siRNA knock down of E11 expression achieved >70% reduction of basal E11 mRNA expression (p < 0.05) and effectively abrogated FGF‐2‐related changes in E11 expression and dendrite formation. FGF‐2 strongly activated the ERK signaling pathway in osteoblast‐like cells but inhibition of this pathway did not block the ability of FGF‐2 to enhance E11 expression or to promote acquisition of the osteocyte phenotype. The results of this study highlight a novel mechanism by which FGF‐2 can regulate osteoblast differentiation and osteocyte formation. Specifically, the data suggests that FGF‐2 promotes osteocytogenesis through increased E11 expression and further studies will identify if this regulatory pathway is essential for bone development and maintenance in health and disease.
Collapse
Affiliation(s)
- Ekele Ikpegbu
- Roslin Institute and R(D)SVS, The University of Edinburgh, Edinburgh, UK.,Michael Okpara University of Agriculture, Abia, Nigeria
| | - Lena Basta
- Roslin Institute and R(D)SVS, The University of Edinburgh, Edinburgh, UK
| | - Dylan N Clements
- Roslin Institute and R(D)SVS, The University of Edinburgh, Edinburgh, UK
| | - Robert Fleming
- Roslin Institute and R(D)SVS, The University of Edinburgh, Edinburgh, UK
| | - Tonia L Vincent
- Arthritis Research UK Centre for Osteoarthritis Pathogenesis, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - David J Buttle
- Department of Infection, Immunity & Cardiovascular Disease, The University of Sheffield Medical School, Sheffield, UK
| | | | | | - Colin Farquharson
- Roslin Institute and R(D)SVS, The University of Edinburgh, Edinburgh, UK
| |
Collapse
|
36
|
Zheng D, Cui C, Yu M, Li X, Wang L, Chen X, Lin Y. Coenzyme Q10 promotes osteoblast proliferation and differentiation and protects against ovariectomy-induced osteoporosis. Mol Med Rep 2017; 17:400-407. [PMID: 29115467 DOI: 10.3892/mmr.2017.7907] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 09/08/2017] [Indexed: 11/06/2022] Open
Abstract
Coenzyme Q10 (CoQ10) is a fat‑soluble vitamin‑like substance used for the treatment of a variety of disorders, including osteoporosis. The exact mechanism underlying CoQ10‑mediated protection against osteoporosis remains to be elucidated. The present study aimed to evaluate the effect of CoQ10 on osteoblastic cell proliferation and differentiation, and therapeutic effects on a rat model of osteoporosis. Following treatment with different concentrations of CoQ10, cell proliferation and differentiation of rat bone marrow stromal cells (BMSCs), and expression of osteoblastogenic markers, were measured. Rats with osteoporosis subjected to ovariectomy (OVX) were treated with different concentrations of CoQ10. Serum levels of estrogen and bone metabolism markers were measured. Micro computed tomography scans were used to analyze morphological changes in bones. In addition, mRNA and protein levels of phosphatidylinositol 3,4,5‑trisphosphate 3‑phosphatase and dual‑specificity protein phosphatase PTEN (PTEN)/phosphatidylinositol 4,5‑bisphosphate 3‑kinase (PI3K)/RAC‑alpha serine/threonine‑protein kinase(AKT), were determined. CoQ10 significantly increased the proliferation and osteogenic differentiation of BMSCs in a dose‑dependent manner, with an increased expression of osteogenic markers. CoQ10 significantly decreased bone resorption but exhibited no effect on serum E2 levels in vivo. CoQ10 markedly enhanced bone formation. Furthermore, the abundance of p‑PI3K and p‑AKT increased while PTEN levels decreased in a dose‑dependent manner following administration of CoQ10. CoQ10 stimulates the proliferation and differentiation of BMSCs and is effective for the treatment of OVX‑induced osteoporosis in rats. The above effects of CoQ10 may be mediated by activation of the PTEN/PI3K/AKT pathway.
Collapse
Affiliation(s)
- Delu Zheng
- Department of Osteoporosis Diagnostic, Research and Treatment Center, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Chenli Cui
- Department of Gynecology, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Meng Yu
- Department of Osteoporosis Diagnostic, Research and Treatment Center, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Xiang Li
- Department of Osteoporosis Diagnostic, Research and Treatment Center, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Lu Wang
- Department of Osteoporosis Diagnostic, Research and Treatment Center, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Xinyan Chen
- Department of Osteoporosis Diagnostic, Research and Treatment Center, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| | - Yichen Lin
- Department of Osteoporosis Diagnostic, Research and Treatment Center, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning 116001, P.R. China
| |
Collapse
|
37
|
Yin L, Huang D, Liu X, Wang Y, Liu J, Liu F, Yu B. Omentin-1 effects on mesenchymal stem cells: proliferation, apoptosis, and angiogenesis in vitro. Stem Cell Res Ther 2017; 8:224. [PMID: 29017592 PMCID: PMC5633887 DOI: 10.1186/s13287-017-0676-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 07/01/2017] [Accepted: 09/18/2017] [Indexed: 02/07/2023] Open
Abstract
Background Mesenchymal stem cells (MSCs) are emerging as an extremely promising therapeutic agent for tissue repair. However, limitations exist such as the low numbers of MSCs obtained from donors, and the poor survival and function of donor cells. Omentin-1, a new fat depot-specific secretory adipokine, exerts proproliferation, prosurvival, and proangiogenic functions in certain cells via an Akt-dependent mechanism; however, little is known about the influence of omentin-1 on MSCs. Methods MSCs were isolated from 60–80 g donor rats. Cell proliferation was assessed with CCK-8 and EdU assay. Cell cycle, apoptosis ratio, reactive oxygen species concentration, and mitochondrial membrane potential were detected by flow cytometry. Hoechst 33342 dye was used to assess morphological changes of apoptosis. Expression levels of Akt, FoxO3a, GSK-3β, and apoptosis- and cell cycle-associated proteins were detected by Western blotting. Tube formation assay was used to test the angiogenesis role of conditioned medium from MSCs in vitro. The cytokine secretion was assessed by ELISA. Results After treatment with omentin-1 (100–800 ng/ml), MSCs displayed a higher proliferative capacity with an increasing number of cells in the S and G2 phase of the cell cycle. Moreover, omentin-1 preconditioning for 1 h could protect MSCs against H2O2-induced apoptosis in a concentration-dependent manner. Furthermore, omentin-1 pretreatment reduced the excessive reactive oxygen species. Western blots revealed that increased Bcl-2 and decreased Bax appeared in MSCs after omentin-1 incubation, which inhibited the mitochondrial apoptosis pathways with evidence showing inhibition of caspase-3 cleavage and preservation of mitochondrial membrane potential. Omentin-1 could enhance angiogenic growth factor secretion and elevate the ability of MSCs to stimulate tube formation by human umbilical vein endothelial cells (HUVECs). Furthermore, omentin-1 enhanced Akt phosphorylation; however, blockade of the PI3K/Akt pathway with an inhibitor, LY294002 (20 μM), suppressed the above beneficial effects of omentin-1. Conclusion Omentin-1 can exert beneficial effects on MSCs by promoting proliferation, inhibiting apoptosis, increasing secretion of angiogenic cytokines, and enhancing the ability for stimulating tube formation by HUVECs via the PI3K/Akt signaling pathway. Thus, omentin-1 may be considered a candidate for optimizing MSC-based cell therapy.
Collapse
Affiliation(s)
- Li Yin
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China
| | - Dan Huang
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China
| | - Xinxin Liu
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China
| | - Yongshun Wang
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China
| | - Jingjin Liu
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China
| | - Fang Liu
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China.,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China
| | - Bo Yu
- Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China. .,Department of Cardiology, The Second Affiliated Hospital of Harbin Medical University, 148 Baojian Road, Harbin, 150086, People's Republic of China.
| |
Collapse
|
38
|
Ling L, Wei T, He L, Wang Y, Wang Y, Feng X, Zhang W, Xiong Z. Low-intensity pulsed ultrasound activates ERK1/2 and PI3K-Akt signalling pathways and promotes the proliferation of human amnion-derived mesenchymal stem cells. Cell Prolif 2017; 50. [PMID: 28940899 DOI: 10.1111/cpr.12383] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 08/18/2017] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVES This study was to investigate the effect and mechanism of low-intensity pulsed ultrasound (LIPUS) on the proliferation of human amnion-derived mesenchymal stem cells (hAD-MSCs). METHODS Human amnion-derived mesenchymal stem cells were isolated from the amnion of term placentas and identified by flow cytometry and differentiation culture. Proliferation of hAD-MSCs was investigated by Cell Counting Kit-8, cell cycle and EdU assays. Western blotting was used to determine the protein expression levels. RESULTS Human amnion-derived mesenchymal stem cells were successfully isolated from the amnion and identified as multipotent mesenchymal stem cells. Low-intensity pulsed ultrasound promoted the proliferation of hAD-MSCs. Cell cycle analysis showed that LIPUS promoted cells to enter S and G2/M phases from G0/G1 phase. Western blot results showed that LIPUS promoted the phosphorylation and activation of ERK1/2 and Akt and significantly upregulated expression of cyclin D1, cyclin E1, cyclin A2 and cyclin B1. ERK1/2 inhibitor (U0126) and PI3K inhibitor (LY294002) significantly reduced LIPUS-induced phosphorylation of ERK1/2 and Akt, respectively, which in turn reduced the LIPUS-induced proliferation of hAD-MSCs. CONCLUSIONS Low-intensity pulsed ultrasound can promote the proliferation of hAD-MSCs, and ERK1/2 and PI3K-Akt signalling pathways may play important roles in this process.
Collapse
Affiliation(s)
- Li Ling
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Tianqin Wei
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Lianli He
- Department of Obstetrics and Gynecology, Third Affiliated Hospital, Zunyi Medical College, Guizhou, 563000, China
| | - Yaping Wang
- Department of Histology and Embryology, Laboratory of Stem Cell and Tissue Engineering, Chongqing Medical University, Chongqing, 400010, China
| | - Yan Wang
- State Key Laboratory of Ultrasound Engineering in Medicine Co-Founded by Chongqing and the Ministry of Science and Technology, Chongqing Key Laboratory of Biomedical Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, 400010, China
| | - Xiushan Feng
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Wenqian Zhang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Zhengai Xiong
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| |
Collapse
|
39
|
Yu B, Sondag GR, Malcuit C, Kim MH, Safadi FF. Macrophage-Associated Osteoactivin/GPNMB Mediates Mesenchymal Stem Cell Survival, Proliferation, and Migration Via a CD44-Dependent Mechanism. J Cell Biochem 2017; 117:1511-21. [PMID: 26442636 DOI: 10.1002/jcb.25394] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2015] [Accepted: 10/05/2015] [Indexed: 12/23/2022]
Abstract
Although MSCs have been widely recognized to have therapeutic potential in the repair of injured or diseased tissues, it remains unclear how functional activities of mesenchymal stem cells (MSCs) are influenced by the surrounding inflammatory milieu at the site of tissue injury. Macrophages constitute an essential component of innate immunity and have been shown to exhibit a phenotypic plasticity in response to various stimuli, which play a central role in both acute inflammation and wound repair. Osteoactivin (OA)/Glycoprotein non-metastatic melanoma protein B (GPNMB), a transmembrane glycoprotein that plays a role in cell differentiation, survival, and angiogenesis. The objective of this study was to investigate the potential role of OA/GPNMB in macrophage-induced MSC function. We found that reparative M2 macrophages express significantly greater levels of OA/GPNMB than pro-inflammatory M1 macrophages. Furthermore, using loss of function and rescue studies, we demonstrated that M2 macrophages-secreted OA/GPNMB positively regulates the viability, proliferation, and migration of MSCs. More importantly, we demonstrated that OA/GPNMB acts through ERK and AKT signaling pathways in MSCs via CD44, to induce these effects. Taken together, our results provide pivotal insight into the mechanism by which OA/GPNMB contributes to the tissue reparative phenotype of M2 macrophages and positively regulates functional activities of MSCs. J. Cell. Biochem. 117: 1511-1521, 2016. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Bing Yu
- Department of Biological Sciences, Kent State University, Kent, Ohio
| | - Gregory R Sondag
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED), Rootstown, Ohio.,School of Biomedical of Sciences, Kent State University, Kent, OH
| | | | - Min-Ho Kim
- Department of Biological Sciences, Kent State University, Kent, Ohio
| | - Fayez F Safadi
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED), Rootstown, Ohio.,School of Biomedical of Sciences, Kent State University, Kent, OH
| |
Collapse
|
40
|
Schneider AK, Cama G, Ghuman M, Hughes FJ, Gharibi B. Sprouty 2
, an Early Response Gene Regulator of FosB
and Mesenchymal Stem Cell Proliferation During Mechanical Loading and Osteogenic Differentiation. J Cell Biochem 2017; 118:2606-2614. [DOI: 10.1002/jcb.26035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 04/05/2017] [Indexed: 01/26/2023]
Affiliation(s)
- A. Kristin Schneider
- Division of Tissue Engineering and Biophotonics; Dental Institute; King's College London, Tower Wing, Guy's Hospital; London SE1 9RT UK
| | - Giuseppe Cama
- Division of Tissue Engineering and Biophotonics; Dental Institute; King's College London, Tower Wing, Guy's Hospital; London SE1 9RT UK
| | - Mandeep Ghuman
- Division of Tissue Engineering and Biophotonics; Dental Institute; King's College London, Tower Wing, Guy's Hospital; London SE1 9RT UK
| | - Francis J. Hughes
- Division of Tissue Engineering and Biophotonics; Dental Institute; King's College London, Tower Wing, Guy's Hospital; London SE1 9RT UK
| | - Borzo Gharibi
- Division of Tissue Engineering and Biophotonics; Dental Institute; King's College London, Tower Wing, Guy's Hospital; London SE1 9RT UK
| |
Collapse
|
41
|
Leukocyte-Reduced Platelet-Rich Plasma Alters Protein Expression of Adipose Tissue-Derived Mesenchymal Stem Cells. Plast Reconstr Surg 2017; 138:397-408. [PMID: 27064225 DOI: 10.1097/prs.0000000000002388] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Application of platelet-rich plasma and stem cells has become important in regenerative medicine. Recent literature supports the use of platelet-rich plasma as a cell culture media supplement to stimulate proliferation of adipose tissue-derived mesenchymal stem cells. The underlying mechanism of proliferation stimulation by platelet-rich plasma has not been investigated so far. METHODS Adipose tissue-derived mesenchymal stem cells were cultured in α-minimal essential medium supplemented with platelet-rich plasma or fetal calf serum. Cell proliferation was assessed with cell cycle kinetics using flow cytometric analyses after 48 hours. Differences in proteome expression of the adipose tissue-derived mesenchymal stem cells were analyzed using a reverse-phase protein array to quantify 214 proteins. Complementary Ingenuity Pathways Analysis and gene set enrichment analysis were performed using protein data, and confirmed by Western blot analysis. RESULTS A higher percentage of adipose tissue-derived mesenchymal stem cells in the S phase in the presence of platelet-rich plasma advocates the proliferation stimulation. Ingenuity Pathways Analysis and gene set enrichment analysis confirm the involvement of the selected proteins in the process of cell growth and proliferation. Ingenuity Pathways Analysis revealed a participation in the top-ranked canonical pathways PI3K/AKT, PTEN, ILK, and IGF-1. Gene set enrichment analysis identified the authors' protein set as being part of significantly regulated protein sets with the focus on cell cycle, metabolism, and the Kyoto Encyclopedia of Genes and Genomes transforming growth factor-β signaling pathway. CONCLUSIONS The present study provides evidence that platelet-rich plasma stimulates proliferation and induces a unique change in the proteomic profile of adipose tissue-derived mesenchymal stem cells. The interpretation of altered expression of regulatory proteins represents a step forward toward achieving good manufacturing practice-compliant criteria for cell-based strategies.
Collapse
|
42
|
Zhang M, Zhang P, Liu Y, Lv L, Zhang X, Liu H, Zhou Y. RSPO3-LGR4 Regulates Osteogenic Differentiation Of Human Adipose-Derived Stem Cells Via ERK/FGF Signalling. Sci Rep 2017; 7:42841. [PMID: 28220828 PMCID: PMC5318871 DOI: 10.1038/srep42841] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 01/16/2017] [Indexed: 01/09/2023] Open
Abstract
The four R-spondins (RSPOs) and their three related receptors, LGR4, 5 and 6, have emerged as a major ligand-receptor system with critical roles in development and stem cell survival. However, the exact roles of the RSPO-LGR system in osteogenesis remain largely unknown. In the present study, we showed that RSPO3-shRNA increased the osteogenic potential of human adipose-derived stem cells (hASCs) significantly. Mechanistically, we demonstrated that RSPO3 is a negative regulator of ERK/FGF signalling. We confirmed that inhibition of the ERK1/2 signalling pathway blocked osteogenic differentiation in hASCs, and the increased osteogenic capacity observed after RSPO3 knockdown in hASCs was reversed by inhibition of ERK signalling. Further, silencing of LGR4 inhibited the activity of ERK signalling and osteogenic differentiation of hASCs. Most importantly, we found that loss of LGR4 abrogated RSPO3-regulated osteogenesis and RSPO3-induced ERK1/2 signalling inhibition. Collectively, our data show that ERK signalling works downstream of LGR4 and RSPO3 regulates osteoblastic differentiation of hASCs possibly via the LGR4-ERK signalling.
Collapse
Affiliation(s)
- Min Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, China.,National Engineering Lab for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Ping Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, China.,National Engineering Lab for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, China.,National Engineering Lab for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Longwei Lv
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, China.,National Engineering Lab for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Xiao Zhang
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, China.,National Engineering Lab for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Hao Liu
- National Engineering Lab for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, 100081, China.,Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, China.,National Engineering Lab for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing, 100081, China
| |
Collapse
|
43
|
Sardesai VS, Shafiee A, Fisk NM, Pelekanos RA. Avoidance of Maternal Cell Contamination and Overgrowth in Isolating Fetal Chorionic Villi Mesenchymal Stem Cells from Human Term Placenta. Stem Cells Transl Med 2017; 6:1070-1084. [PMID: 28205414 PMCID: PMC5442838 DOI: 10.1002/sctm.15-0327] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 07/05/2016] [Accepted: 07/28/2016] [Indexed: 12/16/2022] Open
Abstract
Human placenta is rich in mesenchymal stem/stromal cells (MSC), with their origin widely presumed fetal. Cultured placental MSCs are confounded by a high frequency of maternal cell contamination. Our recent systematic review concluded that only a small minority of placental MSC publications report fetal/maternal origin, and failed to discern a specific methodology for isolation of fetal MSC from term villi. We determined isolation conditions to yield fetal and separately maternal MSC during ex vivo expansion from human term placenta. MSCs were isolated via a range of methods in combination; selection from various chorionic regions, different commercial media, mononuclear cell digest and/or explant culture. Fetal and maternal cell identities were quantitated in gender‐discordant pregnancies by XY chromosome fluorescence in situ hybridization. We first demonstrated reproducible maternal cell contamination in MSC cultures from all chorionic anatomical locations tested. Cultures in standard media rapidly became composed entirely of maternal cells despite isolation from fetal villi. To isolate pure fetal cells, we validated a novel isolation procedure comprising focal dissection from the cotyledonary core, collagenase/dispase digestion and explant culture in endothelial growth media that selected, and provided a proliferative environment, for fetal MSC. Comparison of MSC populations within the same placenta confirmed fetal to be smaller, more osteogenic and proliferative than maternal MSC. We conclude that in standard media, fetal chorionic villi‐derived MSC (CV‐MSC) do not grow readily, whereas maternal MSC proliferate to result in maternal overgrowth during culture. Instead, fetal CV‐MSCs require isolation under specific conditions, which has implications for clinical trials using placental MSC. Stem Cells Translational Medicine2017;6:1070–1084
Collapse
Affiliation(s)
- Varda S Sardesai
- The University of Queensland, UQ Centre for Clinical Research, Experimental Fetal Medicine Group, Herston, Queensland, Australia
| | - Abbas Shafiee
- The University of Queensland, UQ Centre for Clinical Research, Experimental Fetal Medicine Group, Herston, Queensland, Australia.,Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Queensland, Australia
| | - Nicholas M Fisk
- The University of Queensland, UQ Centre for Clinical Research, Experimental Fetal Medicine Group, Herston, Queensland, Australia.,Centre for Advanced Prenatal Care, Royal Brisbane & Women's Hospital, Brisbane, Australia
| | - Rebecca A Pelekanos
- The University of Queensland, UQ Centre for Clinical Research, Experimental Fetal Medicine Group, Herston, Queensland, Australia
| |
Collapse
|
44
|
Mouse Mesenchymal Progenitor Cells Expressing Adipogenic and Osteogenic Transcription Factors Suppress the Macrophage Inflammatory Response. Stem Cells Int 2017; 2017:5846257. [PMID: 28191017 PMCID: PMC5278224 DOI: 10.1155/2017/5846257] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Revised: 11/22/2016] [Accepted: 12/18/2016] [Indexed: 12/25/2022] Open
Abstract
Mesenchymal progenitor cell characteristics that can identify progenitor populations with specific functions in immunity are actively being investigated. Progenitors from bone marrow and adipose tissue regulate the macrophage (MΦ) inflammatory response by promoting the switch from an inflammatory to an anti-inflammatory phenotype. Conversely, mesenchymal progenitors from the mouse aorta (mAo) support and contribute to the MΦ response under inflammatory conditions. We used cell lines with purported opposing immune-regulatory function, a bone marrow derived mesenchymal progenitor cell line (D1) and a mouse aorta derived mesenchymal progenitor cell line (mAo). Their interaction and regulation of the MΦ cell response to the inflammatory mediator, lipopolysaccharide (LPS), was examined by coculture. As expected, D1 cells suppressed NO, TNF-α, and IL-12p70 production but MΦ phagocytic activity remained unchanged. The mAo cells enhanced NO and TNF-α production in coculture and enhanced MΦ phagocytic activity. Using flow cytometry and PCR array, we then sought to identify sets of MSC-associated genes and markers that are expressed by these progenitor populations. We have determined that immune-supportive mesenchymal progenitors highly express chondrogenic and tenogenic transcription factors while immunosuppressive mesenchymal progenitors highly express adipogenic and osteogenic transcription factors. These data will be useful for the isolation, purification, and modification of mesenchymal progenitors to be used in the treatment of inflammatory diseases.
Collapse
|
45
|
Nowwarote N, Sukarawan W, Pavasant P, Osathanon T. Basic Fibroblast Growth Factor Regulates REX1 Expression Via IL-6 In Stem Cells Isolated From Human Exfoliated Deciduous Teeth. J Cell Biochem 2016; 118:1480-1488. [PMID: 27883224 DOI: 10.1002/jcb.25807] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 11/22/2016] [Indexed: 11/06/2022]
Abstract
Basic fibroblast growth factor (bFGF) regulates pluripotent marker expression and cellular differentiation in various cell types. However, the mechanism by which bFGF regulates REX1 expression in stem cells, isolated from human exfoliated deciduous teeth (SHEDs) remains unclear. The aim of the present study was to investigate the regulation of REX1 expression by bFGF in SHEDs. SHEDs were isolated and characterized. Their mRNA and protein expression levels were determined using real-time polymerase chain reaction and enzyme-linked immunosorbent assay, respectively. In some experiments, chemical inhibitors were added to the culture medium to impede specific signaling pathways. Cells isolated from human exfoliated deciduous tooth dental pulp tissue expressed mesenchymal stem cell surface markers (CD44, CD73, CD90, and CD105). These cells differentiated into osteogenic and adipogenic lineages, when appropriately induced. Treating SHEDs with bFGF induced REX1 mRNA expression and this effect was attenuated by pretreatment with FGFR or Akt inhibitors. Cycloheximide pretreatment also inhibited the bFGF-induced REX1 expression, implying the involvement of intermediate molecule(s). Further, the addition of an IL-6 neutralizing antibody attenuated the bFGF-induced REX1 expression by SHEDs. In conclusion, bFGF enhanced REX1 expression by SHEDs via the FGFR and Akt signaling pathways. Moreover, IL-6 participated in the bFGF-induced REX1 expression in SHEDs. J. Cell. Biochem. 118: 1480-1488, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Nunthawan Nowwarote
- Graduate Program in Oral Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand.,Mineralized Tissue Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Waleerat Sukarawan
- Department of Pediatric Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Prasit Pavasant
- Mineralized Tissue Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand.,Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Thanaphum Osathanon
- Mineralized Tissue Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand.,Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330, Thailand
| |
Collapse
|
46
|
Jo W, Jeong D, Kim J, Park J. Self-Renewal of Bone Marrow Stem Cells by Nanovesicles Engineered from Embryonic Stem Cells. Adv Healthc Mater 2016; 5:3148-3156. [PMID: 27860451 DOI: 10.1002/adhm.201600810] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 09/02/2016] [Indexed: 12/15/2022]
Abstract
Extracellular vesicles can enhance cell proliferation by stimulating signal transduction and delivering genetic materials, and thus may have applications in regenerative medicine and other therapeutic applications. The processes employed to isolate extracellular vesicles, however, are complex and achieve low yield. To overcome these obstacles, a large-scale, micropore device for generating extracellular vesicle-mimetic nanovesicles that have characteristics similar to those of extracellular vesicles is fabricated. The nanovesicles are generated through the self-assembly capability of cell membrane fragments in an aqueous solution. The nanovesicles enhance the proliferation of murine mesenchymal stem cells (MSCs), stimulate the signal pathway related to cell proliferation, and do not influence the characteristics of murine MSCs. Therefore, these nanovesicles could provide stable MSCs for regenerative medicine and other therapeutic applications.
Collapse
Affiliation(s)
- Wonju Jo
- Department of Mechanical Engineering; Pohang University of Science and Technology (POSTECH); 77 Cheongam-Ro, Nam-gu Pohang Gyeong-buk 37673 Republic of Korea
| | - Dayeong Jeong
- School of Interdisciplinary Bioscience and Bioengineering; POSTECH; 77 Cheongam-Ro, Nam-gu Pohang Gyeong-buk 37673 Republic of Korea
| | - Junho Kim
- School of Interdisciplinary Bioscience and Bioengineering; POSTECH; 77 Cheongam-Ro, Nam-gu Pohang Gyeong-buk 37673 Republic of Korea
| | - Jaesung Park
- Department of Mechanical Engineering; Pohang University of Science and Technology (POSTECH); 77 Cheongam-Ro, Nam-gu Pohang Gyeong-buk 37673 Republic of Korea
- School of Interdisciplinary Bioscience and Bioengineering; POSTECH; 77 Cheongam-Ro, Nam-gu Pohang Gyeong-buk 37673 Republic of Korea
| |
Collapse
|
47
|
FGF2 Stimulates COUP-TFII Expression via the MEK1/2 Pathway to Inhibit Osteoblast Differentiation in C3H10T1/2 Cells. PLoS One 2016; 11:e0159234. [PMID: 27404388 PMCID: PMC4942136 DOI: 10.1371/journal.pone.0159234] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 06/29/2016] [Indexed: 11/20/2022] Open
Abstract
Chicken ovalbumin upstream promoter transcription factor II (COUP-TFII) is an orphan nuclear receptor that regulates many key biological processes, including organ development and cell fate determination. Although the biological functions of COUP-TFII have been studied extensively, little is known about what regulates its gene expression, especially the role of inducible extracellular factors in triggering it. Here we report that COUP-TFII expression is regulated specifically by fibroblast growth factor 2 (FGF2), which mediates activation of the MEK1/2 pathway in mesenchymal lineage C3H10T1/2 cells. Although FGF2 treatment increased cell proliferation, the induction of COUP-TFII expression was dispensable. Instead, FGF2-primed cells in which COUP-TFII expression was induced showed a low potential for osteoblast differentiation, as evidenced by decreases in alkaline phosphatase activity and osteogenic marker gene expression. Reducing COUP-TFII by U0126 or siRNA against COUP-TFII prevented the anti-osteogenic effect of FGF2, indicating that COUP-TFII plays a key role in the FGF2-mediated determination of osteoblast differentiation capability. This report is the first to suggest that FGF2 is an extracellular inducer of COUP-TFII expression and may suppress the osteogenic potential of mesenchymal cells by inducing COUP-TFII expression prior to the onset of osteogenic differentiation.
Collapse
|
48
|
Wang Y, Yu X, Chen E, Li L. Liver-derived human mesenchymal stem cells: a novel therapeutic source for liver diseases. Stem Cell Res Ther 2016; 7:71. [PMID: 27176654 PMCID: PMC4866276 DOI: 10.1186/s13287-016-0330-3] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem cells (MSCs) represent an attractive cell type for research and therapy due to their ability to proliferate, differentiate, modulate immune reactions, and secrete trophic factors. MSCs exist in a multitude of tissues, including bone marrow, umbilical cord, and adipose tissues. Moreover, MSCs have recently been isolated from the liver. Compared with other MSC types, liver-derived human MSCs (LHMSCs) possess general morphologies, immune functions, and differentiation capacities. Interestingly, LHMCSs produce higher levels of pro-angiogenic, anti-inflammatory, and anti-apoptotic cytokines than those of bone marrow-derived MSCs. Thus, these cells may be a promising therapeutic source for liver diseases. This paper summarizes the biological characteristics of LHMSCs and their potential benefits and risks for the treatment of liver diseases.
Collapse
Affiliation(s)
- Yini Wang
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Xiaopeng Yu
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Ermei Chen
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Lanuan Li
- State Key Laboratory for the Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for the Diagnosis and Treatment of Infectious Diseases, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China.
| |
Collapse
|
49
|
Zhang GW, Gu TX, Guan XY, Sun XJ, Qi X, Li XY, Wang XB, Yu L, Jiang DQ, Tang R, Li-Ling J. bFGF binding cardiac extracellular matrix promotes the repair potential of bone marrow mesenchymal stem cells in a rabbit model for acute myocardial infarction. ACTA ACUST UNITED AC 2015; 10:065018. [PMID: 26657457 DOI: 10.1088/1748-6041/10/6/065018] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
To assess the effect of basic fibroblast growth factor-binding extracellular matrix (bFGF-ECM) combined with bone marrow mesenchymal stem cells (BMSCs) transplantation on acute myocardial infarction (AMI) and explore the underlying mechenisms. Rabbit hearts were processed by decellularization with sodium dodecyl sulfate (SDS) perfusion, heparin immobilization, bFGF-binding and homogenization, for preparation of bFGF-binding cardiac ECM suspension (bFGF-ECM). Thereafter, the characteristics of bFGF release were analyzed in vitro. Following ligation of the mid-third of the left anterior descending artery, the rabbits were divided into a control group (no treatment), BMSCs group (BMSCs transplantation), bFGF-ECM group (bFGF-ECM implantation), and BMSCs + bFGF-ECM group (BMSCs and bFGF-ECM implantation). Apoptosis and differentiation of implanted BMSCs, and the left ventricular (LV) remodeling and function were assessed. The ex vivo proliferation, apoptosis, migration and differentiation of BMSCs were determined after exposure to bFGF and/or ECM. The ECM could sustainably release bFGF. 24 h and 6 weeks after the operation, improved viability and differentiation of the implanted BMSCs, as well as inhibited dilatation and preserved function of the left ventricle (LV), were significant in the BMSCs + bFGF-ECM group compared with other groups (P < 0.05), although BMSCs and ECM-bFGF groups also showed better results than control group (P < 0.05). Additionally, ECM and bFGF showed a synergistic effect on BMSCs proliferation, viability, migration and differentiation. The combination of bFGF-binding ECM and BMSCs implantation may promote myocardial regeneration and LV function, and become a new strategy for the treatment of AMI.
Collapse
Affiliation(s)
- Guang-Wei Zhang
- Department of Cardiac Surgery, The First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Fibroblast growth factor (FGF) signaling pathways are essential regulators of vertebrate skeletal development. FGF signaling regulates development of the limb bud and formation of the mesenchymal condensation and has key roles in regulating chondrogenesis, osteogenesis, and bone and mineral homeostasis. This review updates our review on FGFs in skeletal development published in Genes & Development in 2002, examines progress made on understanding the functions of the FGF signaling pathway during critical stages of skeletogenesis, and explores the mechanisms by which mutations in FGF signaling molecules cause skeletal malformations in humans. Links between FGF signaling pathways and other interacting pathways that are critical for skeletal development and could be exploited to treat genetic diseases and repair bone are also explored.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Pierre J Marie
- UMR-1132, Institut National de la Santé et de la Recherche Médicale, Hopital Lariboisiere, 75475 Paris Cedex 10, France; Université Paris Diderot, Sorbonne Paris Cité, 75475 Paris Cedex 10, France
| |
Collapse
|