1
|
Gentili C, Palamà MEF, Sexton G, Maybury S, Shanahan M, Omowunmi-Kayode YY, Martin J, Johnson M, Thompson K, Clarkin O, Coleman CM. Sustainably cultured coral scaffold supports human bone marrow mesenchymal stromal cell osteogenesis. Regen Ther 2024; 26:366-381. [PMID: 39050552 PMCID: PMC11267040 DOI: 10.1016/j.reth.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/31/2024] [Accepted: 06/09/2024] [Indexed: 07/27/2024] Open
Abstract
The current gold standard grafting material is autologous bone due to its osteoinductive and osteoconductive properties. Autograft harvesting results in donors site morbidity. Coral scaffolds offer a natural autograft alternative, sharing the density and porosity of human bone. This study investigated the biocompatibility and osteogenic potential of a novel, sustainably grown Pocillopora scaffold with human bone marrow-derived mesenchymal stromal cells (MSCs). The coral-derived scaffold displays a highly textured topography, with concavities of uniform size and a high calcium carbonate content. Large scaffold samples exhibit compressive and diametral tensile strengths in the range of trabecular bone, with strengths likely increasing for smaller particulate samples. Following the in vitro seeding of MSCs adjacent to the scaffold, the MSCs remained viable, continued proliferating and metabolising, demonstrating biocompatibility. The seeded MSCs densely covered the coral scaffold with organized, aligned cultures with a fibroblastic morphology. In vivo coral scaffolds with MSCs supported earlier bone and blood vessel formation as compared to control constructs containing TCP-HA and MSCs. This work characterized a novel, sustainably grown coral scaffold that was biocompatible with MSCs and supports their in vivo osteogenic differentiation, advancing the current repertoire of biomaterials for bone grafting.
Collapse
Affiliation(s)
- Chiara Gentili
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy
| | | | - Gillian Sexton
- College of Medicine, Nursing and Health Science, School of Medicine, Regenerative Medicine Institute (REMEDI), University of Galway, Galway, Ireland
| | - Sophie Maybury
- College of Medicine, Nursing and Health Science, School of Medicine, Regenerative Medicine Institute (REMEDI), University of Galway, Galway, Ireland
| | - Megan Shanahan
- College of Medicine, Nursing and Health Science, School of Medicine, Regenerative Medicine Institute (REMEDI), University of Galway, Galway, Ireland
| | - Yeyetunde Yvonne Omowunmi-Kayode
- DCU Biomaterials Research Group, Centre for Medical Engineering Research, School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland
| | - James Martin
- Zoan Nuáil Teoranta T/A Zoan BioMed, The Hatchery Building, Cloonacarton, Recess, Galway, Ireland
| | - Martin Johnson
- Zoan Nuáil Teoranta T/A Zoan BioMed, The Hatchery Building, Cloonacarton, Recess, Galway, Ireland
- Ecodiversity Ltd, Derryconnell, Schull, Co. Cork, Ireland
| | - Kerry Thompson
- College of Medicine, Nursing and Health Science, School of Medicine, Anatomy Imaging and Microscopy Facility, University of Galway, Galway, Ireland
| | - Owen Clarkin
- DCU Biomaterials Research Group, Centre for Medical Engineering Research, School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland
| | - Cynthia M. Coleman
- College of Medicine, Nursing and Health Science, School of Medicine, Regenerative Medicine Institute (REMEDI), University of Galway, Galway, Ireland
| |
Collapse
|
2
|
Zhu J, Liu Y, Chen C, Chen H, Huang J, Luo Y, Zhao K, Chen D, Xu Z, Li W, Zhang X, Xiong Y, Xu L, Wang B. Cyasterone accelerates fracture healing by promoting MSCs migration and osteogenesis. J Orthop Translat 2021; 28:28-38. [PMID: 33717979 PMCID: PMC7905397 DOI: 10.1016/j.jot.2020.11.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 10/22/2020] [Accepted: 11/05/2020] [Indexed: 12/12/2022] Open
Abstract
Background Mesenchymal Stem Cells (MSCs) therapy has become a new coming focus of clinical research in regenerative medicine. However, only a small number of implanted MSCs could successfully reach the injured areas. The previous studies have shown that fracture healing time is inversely proportional to concentration of MSCs in injured tissue. Methods The migration and osteogenesis of MSCs were assessed by transwell assay and Alizarin Red S staining. Levels of gene and protein expression were checked by qPCR and Western Blot. On the other hand, the enhanced migration ability of MSCs induced by Cyasterone was retarded by CXCR4 siRNA. In addition, the rat model of femoral fracture was established to evaluate the effect of Cyasterone on fracture healing. What's more, we also checked the effect of Cyasterone on mobilisation of MSCs in vivo. Results The results showed that Cyasteron increased the number of MSCs in peripheral blood. The concentrations of SDF-1α in serum at different time points were determined by ELISA assay. Micro-CT and histological analysis were used to evaluate the fractured femurs.Our results showed that Cyasterone could promote the migration and osteogenesis capacities of MSCs. The fractured femurs healed faster with treatment of Cyasterone. Meanwhile, Cyasterone could significantly increase the level of SDF-1α in rats with femur fracture. Conclusion Cyasterone could promote migration and osteogenesis of MSCs, and most importantly, it could accelerate bone fracture healing. Translational Potential statement: These findings provide evidence that Cyasterone could be used as a therapeutic reagent for MSCs mobilisation and osteogenesis. What's more, it could acclerate fracture healing.
Collapse
Affiliation(s)
- Junlang Zhu
- Department of Traumatology, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510240, PR China
| | - Yamei Liu
- College of Basic Medical, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
- Innovative Research & Development Lab. of TCM, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Chen Chen
- College of Basic Medical, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
- Innovative Research & Development Lab. of TCM, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Hongtai Chen
- Department of Orthopaedics and Traumatology, LKS Faculty of Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong, Hong Kong SAR, 999077, PR China
| | - Jiewen Huang
- Department of Traumatology, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510240, PR China
| | - Yiwen Luo
- Department of Traumatology, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510240, PR China
| | - Kewei Zhao
- Department of Laboratory Medicine, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510240, PR China
| | - Dongfeng Chen
- College of Basic Medical, Guangzhou University of Chinese Medicine, Guangzhou, 510006, PR China
| | - Zhiming Xu
- Wuyi Hospital of Traditional Chinese Medicine, Jiangmen, 529000, PR China
| | - Wangyang Li
- The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, 410208, PR China
| | - Xunchao Zhang
- Department of Traumatology, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510240, PR China
| | - Yunpu Xiong
- Department of Orthopaedics, Guangzhou Orthopedic Hospital, Guangzhou, 510030, PR China
| | - Liangliang Xu
- Lingnan Medical Research Center, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510405, PR China
| | - Bin Wang
- Department of Traumatology, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510240, PR China
| |
Collapse
|
3
|
Zhang Q, Cheng X, Zhang H, Zhang T, Wang Z, Zhang W, Yu W. Dissecting molecular mechanisms underlying H 2O 2-induced apoptosis of mouse bone marrow mesenchymal stem cell: role of Mst1 inhibition. Stem Cell Res Ther 2020; 11:526. [PMID: 33298178 PMCID: PMC7724846 DOI: 10.1186/s13287-020-02041-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 11/19/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Bone marrow mesenchymal stem cell (BM-MSC) has been shown to treat pulmonary arterial hypertension (PAH). However, excessive reactive oxygen species (ROS) increases the apoptosis of BM-MSCs, leading to poor survival and engraft efficiency. Thus, improving the ability of BM-MSCs to scavenge ROS may considerably enhance the effectiveness of transplantation therapy. Mammalian Ste20-like kinase 1 (Mst1) is a pro-apoptotic molecule which increases ROS production. The aim of this study is to uncover the underlying mechanisms the effect of Mst1 inhibition on the tolerance of BM-MSCs under H2O2 condition. METHODS Mst1 expression in BM-MSCs was inhibited via transfection with adenoviruses expressing a short hairpin (sh) RNA directed against Mst1 (Ad-sh-Mst1) and exposure to H2O2. Cell viability was detected by Cell Counting Kit 8 (CCK-8) assay, and cell apoptosis was analyzed by Annexin V-FITC/PI, Caspase 3 Activity Assay kits, and pro caspase 3 expression. ROS level was evaluated by the ROS probe DCFH-DA, mitochondrial membrane potential (ΔΨm) assay, SOD1/2, CAT, and GPx expression. Autophagy was assessed using transmission electron microscopy, stubRFP-sensGFP-LC3 lentivirus, and autophagy-related protein expression. The autophagy/Keap1/Nrf2 signal in H2O2-treated BM-MSC/sh-Mst1 was also measured. RESULTS Mst1 inhibition reduced ROS production; increased antioxidant enzyme SOD1/2, CAT, and GPx expression; maintained ΔΨm; and alleviated cell apoptosis in H2O2-treated BM-MSCs. In addition, this phenomenon was closely correlated with the autophagy/Keap1/Nrf2 signal pathway. Moreover, the antioxidant pathway Keap1/Nrf2 was also blocked when autophagy was inhibited by the autophagy inhibitor 3-MA. However, Keap1 or Nrf2 knockout via siRNA had no effect on autophagy activation or suppression. CONCLUSION Mst1 inhibition mediated the cytoprotective action of mBM-MSCs against H2O2-induced oxidative stress injury. The underlying mechanisms involve autophagy activation and the Keap1/Nrf2 signal pathway.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250062, Shandong, China
| | - Xianfeng Cheng
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250062, Shandong, China.,Department of Cardiovascular Surgery, Weifang People's Hospital, Weifang, 261000, Shandong, China
| | - Haizhou Zhang
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250062, Shandong, China
| | - Tao Zhang
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250062, Shandong, China
| | - Zhengjun Wang
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250062, Shandong, China
| | - Wenlong Zhang
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250062, Shandong, China
| | - Wancheng Yu
- Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250062, Shandong, China.
| |
Collapse
|
4
|
Lv Z, Muheremu A, Bai X, Zou X, Lin T, Chen B. PTH(1‑34) activates the migration and adhesion of BMSCs through the rictor/mTORC2 pathway. Int J Mol Med 2020; 46:2089-2101. [PMID: 33125102 PMCID: PMC7595657 DOI: 10.3892/ijmm.2020.4754] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 07/09/2020] [Indexed: 12/23/2022] Open
Abstract
The ability of intermittent parathyroid hormone (1-34) [PTH(1-34)] treatment to enhance bone-implant osseo-integration was recently demonstrated in vivo. However, the mechanisms through which PTH (1-34) regulates bone marrow-derived stromal cells (BMSCs) remain unclear. The present study thus aimed to investigate the effects of PTH(1-34) on the migration and adhesion of, and rictor/mammalian target of rapamycin complex 2 (mTORC2) signaling in BMSCs. In the present study, BMSCs were isolated from Sprague-Dawley rats treated with various concentrations of PTH(1-34) for different periods of time. PTH(1-34) treatment was performed with or without an mTORC1 inhibitor (20 nM rapamycin) and mTORC1/2 inhibitor (10 µM PP242). Cell migration was assessed by Transwell cell migration assays and wound healing assays. Cell adhesion and related mRNA expression were investigated through adhesion assays and reverse transcription-quantitative polymerase chain reaction (RT-qPCR), respectively. The protein expression of chemokine receptors (CXCR4 and CCR2) and adhesion factors [intercellular adhesion molecule 1 (ICAM-1), fibronectin and integrin β1] was examined by western blot analysis. The results revealed that various concentrations (1, 10, 20, 50 and 100 nM) of PTH(1-34) significantly increased the migration and adhesion of BMSCs, as well as the expression of CXCR4, CCR2, ICAM-1, fibronectin and integrin β1. In addition, the p-Akt and p-S6 levels were also upregulated by PTH(1-34). BMSCs subjected to mTORC1/2 signaling pathway inhibition or rictor silencing exhibited a markedly reduced PTH-induced migration and adhesion, while no such effect was observed for the BMSCs subjected to mTORC1 pathway inhibition or raptor silencing. These results indicate that PTH(1-34) promotes BMSC migration and adhesion through rictor/mTORC2 signaling in vitro. Taken together, the results of the present study reveal an important mechanism for the therapeutic effects of PTH(1-34) on bone-implant osseointegration and suggest a potential treatment strategy based on the effect of PTH(1-34) on BMSCs.
Collapse
Affiliation(s)
- Zhong Lv
- Department of Orthopedics, Guangzhou Panyu Central Hospital, Guangzhou, Guangdong 510080, P.R. China
| | | | - Xiaochun Bai
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Xuenong Zou
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Tao Lin
- Department of Orthopedics, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510280, P.R. China
| | - Bailing Chen
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
5
|
Yao X, Ning LJ, He SK, Cui J, Hu RN, Zhang Y, Zhang YJ, Luo JC, Ding W, Qin TW. Stem Cell Extracellular Matrix-Modified Decellularized Tendon Slices Facilitate the Migration of Bone Marrow Mesenchymal Stem Cells. ACS Biomater Sci Eng 2019; 5:4485-4495. [DOI: 10.1021/acsbiomaterials.9b00064] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
6
|
Xiao F, Chen L. Effects of extracorporeal fucosylation of CD44 on the homing ability of rabbit bone marrow mesenchymal stem cells. J Orthop Sci 2019; 24:725-730. [PMID: 30528224 DOI: 10.1016/j.jos.2018.11.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 10/23/2018] [Accepted: 11/16/2018] [Indexed: 02/09/2023]
Abstract
BACKGROUND The aim of the study was to investigate the effects of extracorporeal fucosylation of CD44 on the homing ability of rabbit bone marrow mesenchymal stem cells (BMSCs). METHODS The rabbit BMSCs were extracorporeal fucosylated using alpha-(1,3)-fucosyltransferase VI (FTVI), then the positive rate of sialyl-LewisX (sLeX) and the binding rate of E-selectin were detected by flow cytometry, as well as the fluid adhesion of rabbit BMSCs were detected by the parallel flow chamber adhesion test. Then BMSCs were constructed to stably express enhanced green fluorescent protein (EGFP) and were injected intravenously into the model rabbits with tibial fractures. After 6 weeks of injection, the levels of stromal cell-derived factor (SDF-1) and monocyte chemoattractant protein-1 (MCP-1) in rabbit serum and damaged bone tissues were detected. The positive rate of EGFP expressions was detected by immunohistochemistry staining. RESULTS After fucosylation, the positive rate of sLeX and the binding rate of E-selectin were significantly higher than those in the no fucosylated group. The results of fluorescence microscopy showed that BMSCs with stable expression of EGFP were successfully constructed. The results of ELISA and Western Blot showed that the secretion of SDF-1 and MCP-1 and the expression of SDF-1 and MCP-1 protein in BMSCs treatment group processed by fucosylated were significantly higher than those in BMSCs treatment group processed by no fucosylated. The results of immunohistochemical staining showed that the positive rate of EGFP expression was also significantly increased, which indicated that the BMSCs at the injured bone tissues were significantly increased and helpful in the repair of bone injury. CONCLUSIONS Extracorporeal fucosylation of CD44 molecules can significantly enhance the homing ability of rabbit BMSCs, which may be achieved by SDF-1 and MCP-1 regulation.
Collapse
Affiliation(s)
- Fei Xiao
- Department of Orthopaedics, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430033, China; Department of Orthopaedic Surgery, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, 430071, China
| | - Liaobin Chen
- Department of Orthopaedic Surgery, Zhongnan Hospital, Wuhan University, Wuhan, Hubei, 430071, China.
| |
Collapse
|
7
|
Chen Y, Fu WL, Gan XD, Xing WW, Xia WR, Zou MJ, Liu Q, Wang YY, Zhang C, Xu DG. SAK-HV Promotes RAW264.7 cells Migration Mediated by MCP-1 via JNK and NF-κB Pathways. Int J Biol Sci 2018; 14:1993-2002. [PMID: 30585263 PMCID: PMC6299369 DOI: 10.7150/ijbs.27459] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 09/25/2018] [Indexed: 12/14/2022] Open
Abstract
Macrophage migration plays an essential role in immune system and is also involved in many pathological situations. However, the regulatory mechanism of macrophage migration remains to be elucidated due to its diverse responses to various stimuli. SAK-HV, a multifunctional protein possessing thrombolytic and lipid-lowering activity, can selectively induce the macrophage proliferation. Here, we reported SAK-HV significantly triggered RAW264.7 cells migration through its functional domain of SAK-mutant by activating both c-jun N-terminal kinases (JNK) and nuclear factor-κB (NF-κB) pathways. Meanwhile, SAK-HV upregulated the expression of some effector proteins, among which only the expression of Monocyte chemoattractant protein-1 (MCP-1) was inhibited by the blockade of JNK and NF-κB pathways. Further research showed that MCP-1 promoted migration ultimately by interacting with Chemokine (C-C motif) Receptor 2 (CCR2) in an autocrine manner. In summary, SAK-HV induced RAW264.7 cells migration through its SAK-mutant domain, during which MCP-1 chemokine mediated by JNK and NF-κB pathways played a key role. These results revealed a novel effect of SAK-HV on modulating macrophage migration and also deepened the understanding of its pharmacodynamics.
Collapse
Affiliation(s)
- Yao Chen
- Institute of Military Cognitive and Brain Sciences, Beijing, 100850, China
| | - Wen-Liang Fu
- Institute of Military Cognitive and Brain Sciences, Beijing, 100850, China
| | - Xiang-Dong Gan
- Institute of Military Cognitive and Brain Sciences, Beijing, 100850, China
| | - Wei-Wei Xing
- Institute of Military Cognitive and Brain Sciences, Beijing, 100850, China
| | - Wen-Rong Xia
- Institute of Military Cognitive and Brain Sciences, Beijing, 100850, China
| | - Min-Ji Zou
- Institute of Military Cognitive and Brain Sciences, Beijing, 100850, China
| | - Qing Liu
- Institute of Military Cognitive and Brain Sciences, Beijing, 100850, China
| | - Yuan-Yuan Wang
- Institute of Military Cognitive and Brain Sciences, Beijing, 100850, China
| | - Chao Zhang
- Division of Oncology Research, Mayo Clinic, Rochester, MN 55905, USA
| | - Dong-Gang Xu
- Institute of Military Cognitive and Brain Sciences, Beijing, 100850, China
| |
Collapse
|
8
|
Cui Y, Huang R, Wang Y, Zhu L, Zhang X. Down-regulation of LGR6 promotes bone fracture recovery using bone marrow stromal cells. Biomed Pharmacother 2018; 99:629-637. [PMID: 29625528 DOI: 10.1016/j.biopha.2017.12.109] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 12/08/2017] [Accepted: 12/28/2017] [Indexed: 02/07/2023] Open
Abstract
OBJECTIVE The Leucine-rich repeat-containing G-protein coupled receptor 6 (LGR6) is a well-known marker of stem cells. In present study, we aimed to further explore the effects of LGR6 on promoting osteogenic differentiation of bone marrow stromal cells (BMSCs) and bone healing. METHODS Flow cytometry assay was used to determine the expression of BMSCs surface markers, and western blot was performed to detect the LGR6 protein expression. The osteogenic differentiation of BMSCs was qualified using ALP and ARS staining. Protein expression of osteogenic genes (ALP, Collagen I, Runx2 and OCN) were evaluated using western blot. In vivo, BMSCs transfected with sh-LGR6 or LGR6 cDNA were injected into the fracture site to establish rat fracture healing model. X-ray system and hematoxylin-eosin (HE) staining were conducted to observe the fracture recovery. Biomechanical test was performed to detect the changes of maximum load, elastic modules and bone mineral density. RESULTS In BMSCS, CD90 and CD44 were positively expressed, while CD11b was negatively expressed. Expression level of LGR6 gradually decreased with the osteogenic differentiation of BMSCs. The osteogenic genes expression level during the osteogenic differentiation significantly increased with the down-regulation of LGR6. In vivo, 8 weeks after injection, rats treated with LGR6 knocked-down BMSCs showed increased number of fibroblasts. Maximum load, elastic modulus and the bone mineral density were enhanced with the knocking-down of LGR6. CONCLUSION Inhibition of LGR6 promoted the osteogenic differentiation of BMSCs in vitro. Moreover, transplantation of LGR6-knockout BMSCs in rat models contributes to a better recovery after the fracture.
Collapse
Affiliation(s)
- Yanchao Cui
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China
| | - Renchun Huang
- Emergency Department, Hanzhong Central Hospital, Hanzhong 723000, Shaanxi, China
| | - Yingzhou Wang
- Beijing Meinuoyikang Health Food Co., Ltd., Beijing 100000, China
| | - Li Zhu
- Second Department of Orthopedics, The First Central Hospital of Baoding, No. 320 North Great Wall Street, Baoding 071000, Hebei, China.
| | - Xueliang Zhang
- Department of Orthopedics, The First Hospital of Lanzhou University, No. 1 West Gang Road, East District, Lanzhou 730000, Gansu, China.
| |
Collapse
|
9
|
Rac1 plays a role in CXCL12 but not CCL3-induced chemotaxis and Rac1 GEF inhibitor NSC23766 has off target effects on CXCR4. Cell Signal 2018; 42:88-96. [DOI: 10.1016/j.cellsig.2017.10.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 10/11/2017] [Accepted: 10/13/2017] [Indexed: 12/17/2022]
|
10
|
Castel H, Desrues L, Joubert JE, Tonon MC, Prézeau L, Chabbert M, Morin F, Gandolfo P. The G Protein-Coupled Receptor UT of the Neuropeptide Urotensin II Displays Structural and Functional Chemokine Features. Front Endocrinol (Lausanne) 2017; 8:76. [PMID: 28487672 PMCID: PMC5403833 DOI: 10.3389/fendo.2017.00076] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 03/28/2017] [Indexed: 12/16/2022] Open
Abstract
The urotensinergic system was previously considered as being linked to numerous physiopathological states, including atherosclerosis, heart failure, hypertension, pre-eclampsia, diabetes, renal disease, as well as brain vascular lesions. Thus, it turns out that the actions of the urotensin II (UII)/G protein-coupled receptor UT system in animal models are currently not predictive enough in regard to their effects in human clinical trials and that UII analogs, established to target UT, were not as beneficial as expected in pathological situations. Thus, many questions remain regarding the overall signaling profiles of UT leading to complex involvement in cardiovascular and inflammatory responses as well as cancer. We address the potential UT chemotactic structural and functional definition under an evolutionary angle, by the existence of a common conserved structural feature among chemokine receptorsopioïdergic receptors and UT, i.e., a specific proline position in the transmembrane domain-2 TM2 (P2.58) likely responsible for a kink helical structure that would play a key role in chemokine functions. Even if the last decade was devoted to the elucidation of the cardiovascular control by the urotensinergic system, we also attempt here to discuss the role of UII on inflammation and migration, likely providing a peptide chemokine status for UII. Indeed, our recent work established that activation of UT by a gradient concentration of UII recruits Gαi/o and Gα13 couplings in a spatiotemporal way, controlling key signaling events leading to chemotaxis. We think that this new vision of the urotensinergic system should help considering UT as a chemotactic therapeutic target in pathological situations involving cell chemoattraction.
Collapse
Affiliation(s)
- Hélène Castel
- Normandie University, UNIROUEN, INSERM, DC2N, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
- *Correspondence: Hélène Castel,
| | - Laurence Desrues
- Normandie University, UNIROUEN, INSERM, DC2N, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Jane-Eileen Joubert
- Normandie University, UNIROUEN, INSERM, DC2N, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Marie-Christine Tonon
- Normandie University, UNIROUEN, INSERM, DC2N, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Laurent Prézeau
- CNRS UMR 5203, INSERM U661, Institute of Functional Genomic (IGF), University of Montpellier 1 and 2, Montpellier, France
| | - Marie Chabbert
- UMR CNRS 6214, INSERM 1083, Faculté de Médecine 3, Angers, France
| | - Fabrice Morin
- Normandie University, UNIROUEN, INSERM, DC2N, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Pierrick Gandolfo
- Normandie University, UNIROUEN, INSERM, DC2N, Rouen, France
- Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| |
Collapse
|
11
|
Human umbilical cord mesenchymal stem cells delivering sTRAIL home to lung cancer mediated by MCP-1/CCR2 axis and exhibit antitumor effects. Tumour Biol 2016; 37:8425-35. [PMID: 26733169 DOI: 10.1007/s13277-015-4746-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 12/27/2015] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are believed to be a potential vehicle delivering antitumor agents for their tumor-homing capacity, while the underlying mechanism is yet to be explored. The apoptotic ligand TNF-related apoptosis-inducing ligand (TRAIL) has been suggested as a promising candidate for cancer gene therapy owing to its advantage of selectively inducing apoptosis in cancer cells while sparing normal cells. An isoleucine zipper (ISZ) added to the N-terminal of secretable soluble TRAIL (sTRAIL) can generate the trimeric form of TRAIL (ISZ-sTRAIL) and increase its antitumor potential. However, the inefficient delivery and toxicity are still obstacles for its use. In this study, the migration of human umbilical cord mesenchymal stem cells (HUMSCs) to lung cancer was observed through transwell migration assay and animal bioluminescent imaging both in vitro and in vivo. We found that the homing ability of HUMSCs was suppressed after either knocking down the expression of monocyte chemoattractant protein-1(MCP-1) in lung cancer cells or blocking CCR2 expressed on the surface of HUMSCs, indicating the important role of MCP-1/CCR2 axis in the tropism of HUMSCs to lung cancer. Furthermore, we genetically modified HUMSCs to deliver ISZ-sTRAIL to tumor sites specifically. This targeted therapeutic system exhibited promising apoptotic induction and antitumor potential in a xenograft mouse model without obvious side effects. In conclusion, HUMSCs expressing ISZ-sTRAIL might be an efficient therapeutic approach against lung cancer and MCP-1/CCR2 axis is essential for the tumor tropism of HUMSCs.
Collapse
|