1
|
Han D, Ding L, Zheng X, Li S, Yan H, Liu J, Wang H. Proteomics and lipidomics of human umbilical cord mesenchymal stem cells exposed to ionizing radiation. Eur J Med Res 2025; 30:340. [PMID: 40296080 PMCID: PMC12036141 DOI: 10.1186/s40001-025-02578-4] [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: 03/08/2025] [Accepted: 04/10/2025] [Indexed: 04/30/2025] Open
Abstract
OBJECTIVES Mesenchymal stem cell (MSC)-based therapies exhibit beneficial effects on various forms of tissue damage, including ionizing radiation-induced lesions. However, whether ionizing radiation affects the functions of human umbilical cord mesenchymal stem cells (hucMSCs) remains unclear. This study aimed to investigate the effect and possible mechanisms of ionizing radiation on the proliferation and differentiation of hucMSCs. METHODS The hucMSCs were divided into the 1 Gy group (exposure to a single dose (1 Gy) of X-ray radiation (1 Gy/min) for 14 days) and control (without radiation treatment) group. The proliferation, apoptosis, and adipogenic and osteogenic differentiation abilities of hucMSCs in the two groups were evaluated. Moreover, the lipidomics and proteomics analyses were conducted to explore crucial lipids and proteins by which ionizing radiation affected the functions of hucMSCs. In addition, the effects of BYSL on radiation-treated hucMSCs were explore, as well as the involved potential mechanisms. RESULTS X-ray radiation treatment inhibited proliferation, promoted apoptosis, and decreased adipogenic and osteogenic differentiation abilities of hucMSCs. Key lipids, such as triglyceride (TG) and phosphatidylcholine (PC), and hub proteins (BYSL, MRTO4, and RRP9) exhibited significant differences between the 1 Gy group and control group. Moreover, BYSL, MRTO4, and RRP9 were significantly correlated with TG and PC. BYSL overexpression evidently promoted the cell proliferation, adipogenic and osteogenic differentiation abilities of radiation-treated hucMSCs, as well as the protein expression levels of p-GSK-3β/GSK-3β and β-catenin, while suppressed cell apoptosis. However, the GSK-3β inhibitor (1-Az) treatment reversed the protein expression levels of p-GSK-3β/GSK-3β, β-catenin and BYSL, as well as the cell proliferation, apoptosis, adipogenic and osteogenic differentiation abilities of radiation-treated hucMSCs. CONCLUSIONS Our findings reveal that the proliferation and differentiation of hucMSCs are suppressed by radiation, which may be associated with the changes of key lipids (TG and PC) and proteins (BYSL, MRTO4, and RRP9). Furthermore, BYSL promotes adipogenic and osteogenic differentiation abilities of radiation-treated hucMSCs via GSK-3β/β-catenin pathway. These findings help explain the response of hucMSCs to radiation and have clinical implications for improving the outcomes of MSC-based therapies after radiotherapy.
Collapse
Affiliation(s)
- Dongmei Han
- Department of Hematology, Air Force Medical University, Air Force Medical Center, PLA, No.30, Fucheng Road, Beijing, 100142, China
| | - Li Ding
- Department of Hematology, Air Force Medical University, Air Force Medical Center, PLA, No.30, Fucheng Road, Beijing, 100142, China
| | - Xiaoli Zheng
- Department of Hematology, Air Force Medical University, Air Force Medical Center, PLA, No.30, Fucheng Road, Beijing, 100142, China
| | - Sheng Li
- Department of Hematology, Air Force Medical University, Air Force Medical Center, PLA, No.30, Fucheng Road, Beijing, 100142, China
| | - Hongmin Yan
- Department of Hematology, Air Force Medical University, Air Force Medical Center, PLA, No.30, Fucheng Road, Beijing, 100142, China
| | - Jing Liu
- Department of Hematology, Air Force Medical University, Air Force Medical Center, PLA, No.30, Fucheng Road, Beijing, 100142, China
| | - Hengxiang Wang
- Department of Hematology, Air Force Medical University, Air Force Medical Center, PLA, No.30, Fucheng Road, Beijing, 100142, China.
| |
Collapse
|
2
|
Xiang L, Li F, Xiang Y, Zhang W, Shi D, Zhang X, Chen L, Ran Q, Li Z. CR6-Interacting Factor-1 Promotes Osteoclastogenesis Through the NF-κB Signaling Pathway after Irradiation. Radiat Res 2023; 200:489-502. [PMID: 37815199 DOI: 10.1667/rade-22-00066.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 09/11/2023] [Indexed: 10/11/2023]
Abstract
Radiation exposure arising from radiotherapy may induce rapid bone loss and an increase in the extent of bone resorption. Reactive oxygen species (ROS) caused by radiation exposure play a crucial role during the process of osteoclastogenesis. However, the pathological mechanisms underlying radiation-induced osteoclastogenesis have yet to be fully elucidated. CR6-interacting factor-1 (Crif1) as a multifunctional protein is involved in regulating multiple biological functions in cells. Here, we investigated the role of Crif1 in radiation-induced osteoclastogenesis and found that radiation exposure induced an increase in the expression level of Crif1 and enhanced osteoclastogenesis in osteoclast progenitors. Crif1 and NF-κB p65 co-localized in the cytoplasm after radiation exposure. Crif1 knockdown did not affect the phosphorylation and total protein levels of extracellular signal-regulated kinases (ERK), c-Jun amino (N)-terminal kinases (JNK), p38, and IκB-α before and after irradiation. However, Crif1 knockdown did lead to the reduced phosphorylation and nuclear translocation of NF-κB p65 after irradiation and resulted in a reduced level of osteoclastogenesis in RAW264.7 cells after irradiation. In vivo studies involving Lyz2Cre;Crif1fl/fl mice possessing the myeloid-specific deletion of Crif1 demonstrated the alleviation of bone loss after irradiation when compared with Crif1fl/fl mice. Our findings demonstrate that Crif1 mediated the phosphorylation and nuclear translocation of NF-κB p65 and promoted osteoclastogenesis via the NF-κB signaling pathway after radiation exposure. Thus, our analysis revealed a specific role for Crif1 in the mediation of radiation-induced bone loss and may provide new insight into potential therapeutic strategies for radiation-induced bone loss.
Collapse
Affiliation(s)
- Lixin Xiang
- Basic Research Innovation Center for Acute Radiation Syndrome, Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Fengjie Li
- Basic Research Innovation Center for Acute Radiation Syndrome, Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Yang Xiang
- Basic Research Innovation Center for Acute Radiation Syndrome, Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Weiwei Zhang
- Basic Research Innovation Center for Acute Radiation Syndrome, Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Dongling Shi
- Basic Research Innovation Center for Acute Radiation Syndrome, Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Xiaomei Zhang
- Basic Research Innovation Center for Acute Radiation Syndrome, Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Li Chen
- Basic Research Innovation Center for Acute Radiation Syndrome, Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Qian Ran
- Basic Research Innovation Center for Acute Radiation Syndrome, Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China
| | - Zhongjun Li
- Basic Research Innovation Center for Acute Radiation Syndrome, Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Army Military Medical University, Chongqing, 400037, China
| |
Collapse
|
3
|
Xu X, Zhao L, Terry PD, Chen J. Reciprocal Effect of Environmental Stimuli to Regulate the Adipogenesis and Osteogenesis Fate Decision in Bone Marrow-Derived Mesenchymal Stem Cells (BM-MSCs). Cells 2023; 12:1400. [PMID: 37408234 PMCID: PMC10216952 DOI: 10.3390/cells12101400] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 05/02/2023] [Accepted: 05/12/2023] [Indexed: 07/07/2023] Open
Abstract
Mesenchymal stem cells derived from bone marrow (BM-MSCs) can differentiate into adipocytes and osteoblasts. Various external stimuli, including environmental contaminants, heavy metals, dietary, and physical factors, are shown to influence the fate decision of BM-MSCs toward adipogenesis or osteogenesis. The balance of osteogenesis and adipogenesis is critical for the maintenance of bone homeostasis, and the interruption of BM-MSCs lineage commitment is associated with human health issues, such as fracture, osteoporosis, osteopenia, and osteonecrosis. This review focuses on how external stimuli shift the fate of BM-MSCs towards adipogenesis or osteogenesis. Future studies are needed to understand the impact of these external stimuli on bone health and elucidate the underlying mechanisms of BM-MSCs differentiation. This knowledge will inform efforts to prevent bone-related diseases and develop therapeutic approaches to treat bone disorders associated with various pathological conditions.
Collapse
Affiliation(s)
- Xinyun Xu
- Department of Nutrition, The University of Tennessee, Knoxville, TN 37996, USA
| | - Ling Zhao
- Department of Nutrition, The University of Tennessee, Knoxville, TN 37996, USA
| | - Paul D. Terry
- Department of Medicine, Graduate School of Medicine, The University of Tennessee, Knoxville, TN 37920, USA;
| | - Jiangang Chen
- Department of Public Health, The University of Tennessee, Knoxville, TN 37996, USA
| |
Collapse
|
4
|
Jiang Y, Xiang Y, Lin C, Zhang W, Yang Z, Xiang L, Xiao Y, Chen L, Ran Q, Li Z. Multifunctions of CRIF1 in cancers and mitochondrial dysfunction. Front Oncol 2022; 12:1009948. [PMID: 36263222 PMCID: PMC9574215 DOI: 10.3389/fonc.2022.1009948] [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: 08/02/2022] [Accepted: 09/14/2022] [Indexed: 11/23/2022] Open
Abstract
Sustaining proliferative signaling and enabling replicative immortality are two important hallmarks of cancer. The complex of cyclin-dependent kinase (CDK) and its cyclin plays a decisive role in the transformation of the cell cycle and is also critical in the initiation and progression of cancer. CRIF1, a multifunctional factor, plays a pivotal role in a series of cell biological progresses such as cell cycle, cell proliferation, and energy metabolism. CRIF1 is best known as a negative regulator of the cell cycle, on account of directly binding to Gadd45 family proteins or CDK2. In addition, CRIF1 acts as a regulator of several transcription factors such as Nur77 and STAT3 and partly determines the proliferation of cancer cells. Many studies showed that the expression of CRIF1 is significantly altered in cancers and potentially regarded as a tumor suppressor. This suggests that targeting CRIF1 would enhance the selectivity and sensitivity of cancer treatment. Moreover, CRIF1 might be an indispensable part of mitoribosome and is involved in the regulation of OXPHOS capacity. Further, CRIF1 is thought to be a novel target for the underlying mechanism of diseases with mitochondrial dysfunctions. In summary, this review would conclude the latest aspects of studies about CRIF1 in cancers and mitochondria-related diseases, shed new light on targeted therapy, and provide a more comprehensive holistic view.
Collapse
Affiliation(s)
- Yangzhou Jiang
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing, China
| | - Yang Xiang
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing, China
| | - Chuanchuan Lin
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing, China
| | - Weiwei Zhang
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing, China
| | - Zhenxing Yang
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing, China
| | - Lixin Xiang
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing, China
| | - Yanni Xiao
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing, China
| | - Li Chen
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing, China
| | - Qian Ran
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing, China
| | - Zhongjun Li
- Laboratory of Radiation Biology, Laboratory Medicine Center, Department of Blood Transfusion, The Second Affiliated Hospital, Army Military Medical University, Chongqing, China
- State Key Laboratory of Trauma, Burn and Combined Injuries, The Second Affiliated Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
5
|
Dai S, Wen Y, Luo P, Ma L, Liu Y, Ai J, Shi C. Therapeutic implications of exosomes in the treatment of radiation injury. BURNS & TRAUMA 2022; 10:tkab043. [PMID: 35071650 PMCID: PMC8778593 DOI: 10.1093/burnst/tkab043] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 11/11/2021] [Indexed: 12/28/2022]
Abstract
Radiotherapy is one of the main cancer treatments, but it may damage normal tissue and cause various side effects. At present, radioprotective agents used in clinics have side effects such as nausea, vomiting, diarrhea and hypotension, which limit their clinical application. It has been found that exosomes play an indispensable role in radiation injury. Exosomes are lipid bilayer vesicles that carry various bioactive substances, such as proteins, lipids and microRNA (miRNA), that play a key role in cell-to-cell communication and affect tissue injury and repair. In addition, studies have shown that radiation can increase the uptake of exosomes in cells and affect the composition and secretion of exosomes. Here, we review the existing studies and discuss the effects of radiation on exosomes and the role of exosomes in radiation injury, aiming to provide new insights for the treatment of radiation injury.
Collapse
Affiliation(s)
| | | | | | | | | | - Junhua Ai
- Correspondence. Junhua Ai, ; Chunmeng Shi,
| | | |
Collapse
|
6
|
Li F, Zhang R, Hu C, Ran Q, Xiang Y, Xiang L, Chen L, Yang Y, Li SC, Zhang G, Li Z. Irradiation Haematopoiesis Recovery Orchestrated by IL-12/IL-12Rβ1/TYK2/STAT3-Initiated Osteogenic Differentiation of Mouse Bone Marrow-Derived Mesenchymal Stem Cells. Front Cell Dev Biol 2021; 9:729293. [PMID: 34540843 PMCID: PMC8446663 DOI: 10.3389/fcell.2021.729293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 08/03/2021] [Indexed: 11/13/2022] Open
Abstract
PURPOSE Repairing the irradiation-induced osteogenic differentiation injury of bone marrow mesenchymal stem cells (BM-MSCs) is beneficial to recovering haematopoiesis injury in radiotherapy; however, its mechanism is elusive. Our study aimed to help meet the needs of understanding the effects of radiotherapy on BM-MSC osteogenic potential. METHODS AND MATERIALS Balb/c mice and the BM-MSCs were used to evaluate the irradiation-induced osteogenic differentiation injury in vivo. The cellular and molecular characterization were applied to determine the mechanism for recovery of irradiation-derived haematopoiesis injuries. RESULTS We report a functional role of IL-12 in acute irradiation hematopoietic injury recovery and intend to dissect the possible mechanisms through BM-MSC, other than the direct effect of IL-12 on hematopoietic stem and progenitor cells (HSPCs). Specifically, we show that early use of IL-12 enhanced the osteogenic differentiation of BM-MSCs through IL-12Rβ1/TYK2/STAT3 signaling; furthermore, IL-12 induced osteogenesis facilitated bone formation and irradiation hematopoiesis recovery when transplanted BM-MSCs in the femur of Balb/c mice. For the mechanism of action, we found that IL-12 receptor beta 1 (IL-12Rβ1) expression of irradiated BM-MSCs was upregulated rapidly, coincidentally consistent with early use of IL-12 induced osteogenic differentiation enhancement. IL-12Rβ1 and tyrosine kinase 2 gene (Tyk2) silencing experiments and phosphotyrosine of signal transducer and activator of transcription 3 (p-STAT3) suppression experiments indicated the IL-12Rβ1/TYK2/STAT3 signaling was essential in IL-12-induced osteogenic differentiation enhancement of BM-MSCs. CONCLUSION These findings suggested that IL-12 may exert BM-MSCs-based hematopoietic recovery by repairing osteogenic differentiation abilities damages through IL-12Rβ1/TYK2/STAT3 signaling pathway post-irradiation.
Collapse
Affiliation(s)
- Fengjie Li
- Department of Blood Transfusion, The Irradiation Biology Laboratory, The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Rong Zhang
- Department of Pharmacy, The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Changpeng Hu
- Department of Pharmacy, The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Qian Ran
- Department of Blood Transfusion, The Irradiation Biology Laboratory, The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Yang Xiang
- Department of Blood Transfusion, The Irradiation Biology Laboratory, The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Lixin Xiang
- Department of Blood Transfusion, The Irradiation Biology Laboratory, The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Li Chen
- Department of Blood Transfusion, The Irradiation Biology Laboratory, The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Yang Yang
- Department of Blood Transfusion, The Irradiation Biology Laboratory, The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Shengwen Calvin Li
- CHOC Children’s Research Institute, Children’s Hospital of Orange County, University of California, Irvine, Irvine, CA, United States
| | - Gang Zhang
- Department of Oral and Maxillofacial Surgery, The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| | - Zhongjun Li
- Department of Blood Transfusion, The Irradiation Biology Laboratory, The Second Affiliated Hospital, The Third Military Medical University, Chongqing, China
| |
Collapse
|
7
|
Xiang LX, Ran Q, Chen L, Xiang Y, Li FJ, Zhang XM, Xiao YN, Zou LY, Zhong JF, Li SC, Li ZJ. CR6-interacting factor-1 contributes to osteoclastogenesis by inducing receptor activator of nuclear factor κB ligand after radiation. World J Stem Cells 2020; 12:222-240. [PMID: 32266053 PMCID: PMC7118287 DOI: 10.4252/wjsc.v12.i3.222] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 03/09/2020] [Accepted: 03/15/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Radiation induces rapid bone loss and enhances bone resorption and adipogenesis, leading to an increased risk of bone fracture. There is still a lack of effective preventive or therapeutic method for irradiation-induced bone injury. Receptor activator of nuclear factor κB ligand (RANKL) provides the crucial signal to induce osteoclast differentiation and plays an important role in bone resorption. However, the mechanisms of radiation-induced osteoporosis are not fully understood. AIM To investigate the role of CR6-interacting factor-1 (Crif1) in osteoclastogenesis after radiation and its possible mechanism. METHODS C57BL/6 mice were exposed to Co-60 gamma rays and received 5 Gy of whole-body sublethal irradiation at a rate of 0.69 Gy/min. For in vitro study, mouse bone marrow mesenchymal stem/stromal cells (BM-MSCs) were irradiated with Co-60 at a single dose of 9 Gy. For osteoclast induction, monocyte-macrophage RAW264.7 cells were cocultured with mouse BM-MSCs for 7 d. ClusPro and InterProSurf were used to investigate the interaction interface in Crif1 and protein kinase cyclic adenosine monophosphate (cAMP)-activited catalytic subunit alpha complex. Virtual screening using 462608 compounds from the Life Chemicals database around His120 of Crif1 was carried out using the program Autodock_vina. A tetrazolium salt (WST-8) assay was carried out to study the toxicity of compounds to different cells, including human BM-MSCs, mouse BM-MSCs, and Vero cells. RESULTS Crif1 expression increased in bone marrow cells after radiation in mice. Overexpression of Crif1 in mouse BM-MSCs and radiation exposure could increase RANKL secretion and promote osteoclastogenesis in vitro. Deletion of Crif1 in BM-MSCs could reduce both adipogenesis and RANKL expression, resulting in the inhibition of osteoclastogenesis. Deletion of Crif1 in RAW264.7 cells did not affect the receptor activator of nuclear factor κB expression or osteoclast differentiation. Following treatment with protein kinase A (PKA) agonist (forskolin) and inhibitor (H-89) in mouse BM-MSCs, Crif1 induced RANKL secretion via the cAMP/PKA pathway. Moreover, we identified the Crif1-protein kinase cyclic adenosine monophosphate-activited catalytic subunit alpha interaction interface by in silico studies and shortlisted interface inhibitors through virtual screening on Crif1. Five compounds dramatically suppressed RANKL secretion and adipogenesis by inhibiting the cAMP/PKA pathway. CONCLUSION Crif1 promotes RANKL expression via the cAMP/PKA pathway, which induces osteoclastogenesis by binding to receptor activator of nuclear factor κB on monocytes-macrophages in the mouse model. These results suggest a role for Crif1 in modulating osteoclastogenesis and provide insights into potential therapeutic strategies targeting the balance between osteogenesis and adipogenesis for radiation-induced bone injury.
Collapse
Affiliation(s)
- Li-Xin Xiang
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Qian Ran
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Li Chen
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Yang Xiang
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Feng-Jie Li
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Xiao-Mei Zhang
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Yan-Ni Xiao
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Ling-Yun Zou
- Bioinformatics Center, College of Basic Medical Sciences, Third Military Medical University, Chongqing 400038, China
| | - Jiang F Zhong
- Department of Otolaryngology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, United States
| | - Shengwen Calvin Li
- CHOC Children's Research Institute, Children's Hospital of Orange County, University of California, Irvine, CA 92868, United States
| | - Zhong-Jun Li
- Laboratory Medicine Center, Department of Blood Transfusion, Lab of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, Chongqing, 400037, China.
| |
Collapse
|
8
|
Increased BMPR1A Expression Enhances the Adipogenic Differentiation of Mesenchymal Stem Cells in Patients with Ankylosing Spondylitis. Stem Cells Int 2019; 2019:4143167. [PMID: 31827527 PMCID: PMC6885782 DOI: 10.1155/2019/4143167] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Revised: 09/02/2019] [Accepted: 09/10/2019] [Indexed: 11/18/2022] Open
Abstract
Objective To investigate the adipogenic differentiation capacity of mesenchymal stem cells (MSCs) from ankylosing spondylitis (AS) patients and explore the mechanism of abnormal MSC adipogenesis in AS. Methods MSCs from patients with AS (ASMSCs) and healthy donors (HDMSCs) were cultured in adipogenic differentiation medium for up to 21 days. Adipogenic differentiation was determined using oil red O (ORO) staining and quantification and was confirmed by assessing adipogenic marker expression (PPAR-γ, FABP4, and adiponectin). Gene expression of adipogenic markers was detected using qRT-PCR. Protein levels of adipogenic markers and signaling pathway-related molecules were assessed via Western blotting. Levels of bone morphogenetic proteins 4, 6, 7, and 9 were determined using enzyme-linked immunosorbent assays. Lentiviruses encoding short hairpin RNAs (shRNAs) were constructed to reverse abnormal bone morphogenetic protein receptor 1A (BMPR1A) expression and evaluate its role in abnormal ASMSC adipogenic differentiation. Bone marrow fat content was assessed using hematoxylin and eosin (HE) staining. BMPR1A expression in bone marrow MSCs was measured using immunofluorescence staining. Results ASMSCs exhibited a greater adipogenic differentiation capacity than HDMSCs. During adipogenesis, ASMSCs expressed BMPR1A at higher levels, which activated the BMP-pSmad1/5/8 signaling pathway and increased adipogenesis. BMPR1A silencing using an shRNA eliminated the difference in adipogenic differentiation between HDMSCs and ASMSCs. Moreover, HE and immunofluorescence staining showed higher bone marrow fat content and BMPR1A expression in patients with AS than in healthy donors. Conclusion Increased BMPR1A expression induces abnormal ASMSC adipogenic differentiation, potentially contributing to fat metaplasia and thus new bone formation in patients with AS.
Collapse
|
9
|
Ran Q, Jin F, Xiang Y, Xiang L, Wang Q, Li F, Chen L, Zhang Y, Wu C, Zhou L, Xiao Y, Chen L, Wu J, Zhong JF, Li SC, Li Z. CRIF1 as a potential target to improve the radiosensitivity of osteosarcoma. Proc Natl Acad Sci U S A 2019; 116:20511-20516. [PMID: 31548420 PMCID: PMC6789918 DOI: 10.1073/pnas.1906578116] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Resistance to ionizing radiation (IR), which is a conventional treatment for osteosarcoma that cannot be resected, undermines the efficacy of this therapy. However, the mechanism by which IR induces radioresistance in osteosarcoma is not defined. Here, we report that CR6-interacting factor-1 (CRIF1) is highly expressed in osteosarcoma and undergoes nuclear-cytoplasmic shuttling of cyclin-dependent kinase 2 (CDK2) after IR. Osteosarcoma cells lacking CRIF1 show increased sensitivity to IR, which is associated with delayed DNA damage repair, inactivated G1/S checkpoint, and mitochondrial dysfunction. CRIF1 interacts with the DNA damage checkpoint regulator CDK2, and CRIF1 and CDK2 colocalize in the nucleus after IR. Nuclear localization of CDK2 is associated with phosphorylation changes that promote DNA repair and activation of the G1/S checkpoint. CRIF1 knockdown synergized with IR in an in vivo osteosarcoma model, leading to tumor regression. Based on these findings, we identify CRIF1 as a potential therapeutic target in osteosarcoma that can increase the efficacy of radiotherapy. More broadly, our findings may provide insights into the mechanism for other types of radioresistant cancers and be exploited for therapeutic ends.
Collapse
Affiliation(s)
- Qian Ran
- Department of Blood Transfusion, Laboratory of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, 400037 Chongqing, China
- Division of Periodontology, Diagnostic Sciences and Dental Hygiene and Biomedical Sciences, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90089
| | - Feng Jin
- Cancer Center, Daping Hospital and Research Institute of Surgery, Third Military Medical University, 400042 Chongqing, China
| | - Yang Xiang
- Department of Blood Transfusion, Laboratory of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, 400037 Chongqing, China
- Division of Periodontology, Diagnostic Sciences and Dental Hygiene and Biomedical Sciences, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90089
| | - Lixin Xiang
- Department of Blood Transfusion, Laboratory of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, 400037 Chongqing, China
| | - Qiushi Wang
- Department of Pathology and Clinical Biobank, Daping Hospital and Research Institute of Surgery, Third Military Medical University, 400042 Chongqing, China
| | - Fengjie Li
- Department of Blood Transfusion, Laboratory of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, 400037 Chongqing, China
| | - Li Chen
- Department of Blood Transfusion, Laboratory of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, 400037 Chongqing, China
| | - Yuan Zhang
- Department of Orthopedics, The Second Affiliated Hospital, Third Military Medical University, 400037 Chongqing, China
| | - Chun Wu
- Department of Blood Transfusion, Laboratory of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, 400037 Chongqing, China
| | - Luping Zhou
- Department of Blood Transfusion, Laboratory of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, 400037 Chongqing, China
| | - Yanni Xiao
- Department of Blood Transfusion, Laboratory of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, 400037 Chongqing, China
| | - Lili Chen
- Department of Blood Transfusion, Laboratory of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, 400037 Chongqing, China
| | - Jiang Wu
- Department of Blood Transfusion, Laboratory of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, 400037 Chongqing, China
| | - Jiang F Zhong
- Division of Periodontology, Diagnostic Sciences and Dental Hygiene and Biomedical Sciences, Ostrow School of Dentistry, University of Southern California, Los Angeles, CA 90089
| | - Shengwen Calvin Li
- Children's Hospital of Orange County, University of California-Irvine School of Medicine, Orange, CA 92868
| | - Zhongjun Li
- Department of Blood Transfusion, Laboratory of Radiation Biology, The Second Affiliated Hospital, Third Military Medical University, 400037 Chongqing, China;
| |
Collapse
|
10
|
Zhang W, Chen L, Wu J, Li J, Zhang X, Xiang Y, Li F, Wu C, Xiang L, Ran Q, Li Z. Long noncoding RNA TUG1 inhibits osteogenesis of bone marrow mesenchymal stem cells via Smad5 after irradiation. Am J Cancer Res 2019; 9:2198-2208. [PMID: 31149038 PMCID: PMC6531293 DOI: 10.7150/thno.30798] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 02/18/2019] [Indexed: 12/11/2022] Open
Abstract
Irradiation can greatly inhibit osteogenesis of bone marrow mesenchymal stem cells (BM-MSCs). However, the mechanism remains unclear. Methods: We analyzed the expression profile of long noncoding RNAs (lncRNAs) in BM-MSCs using microarray data. LncRNA TUG1 (Taurine Upregulated Gene 1) was selected and tested in radiated BM-MSCs and non-radiated BM-MSCs. Functional analyses (in vitro) were performed to confirm the role of TUG1 in the osteogenic inhibition induced by irradiation. A RIP (RNA immunoprecipitation) assay was performed to detect the interaction of TUG1 and Smad5. Smad5 and the phosphorylated Smad5 (p-Smad5) were tested by western blot. The nuclear translocation of p-Smad5 were tested by immunofluorescence analysis. Furthermore, a series of Smad5 deletions was constructed to identify the TUG1 binding site of Smad5. Results: We found that numerous lncRNAs, including TUG1, exhibit significant expression differences after irradiation. After irradiation TUG1 was significantly increased in BM-MSCs and inhibited osteogenesis. Furthermore, TUG1 directly bound to Smad5, an osteogenic enhancer. Although the phosphorylation level of Smad5 was increased following irradiation, osteogenesis of BM-MSCs was decreased. Mechanistically, TUG1 interacting with the 50-90 aa region of Smad5 and blocks the nuclear translocation of p-Smad5, abolishing osteogenic signalling after irradiation. Conclusion: These results indicate that TUG1 is a negative regulator of Smad5 signalling and suppresses osteogenesis of BM-MSCs after irradiation.
Collapse
|
11
|
Xiang Y, Wu C, Wu J, Quan W, Cheng C, Zhou J, Chen L, Xiang L, Li F, Zhang K, Ran Q, Zhang Y, Li Z. In vitro expansion affects the response of human bone marrow stromal cells to irradiation. Stem Cell Res Ther 2019; 10:82. [PMID: 30850008 PMCID: PMC6408817 DOI: 10.1186/s13287-019-1191-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 01/25/2019] [Accepted: 02/25/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Bone marrow stromal cells (BMSCs) are extensively used in regeneration therapy and cytology experiments simulate how BMSCs respond to radiation. Due to the small number and the heterogeneity of primary isolated BMSCs, extensive in vitro expansion is usually required before application, which affects the cellular characteristics and gene expression of BMSCs. However, whether the radiation response of BMSCs changes during in vitro expansion is unclear. METHODS In this study, BMSCs were passaged in vitro and irradiated at passage 6 (P6) and passage 10 (P10). Then, apoptosis, the cell cycle, senescence, the cytokine secretion and the gene expression profile were analysed for the P6, P10, and non-irradiated (control) BMSCs at different post-irradiation time points. RESULTS The P6 BMSCs had a lower percentage of apoptotic cells than the P10 BMSCs at 24 and 48 h post-irradiation but not compared to that of the controls at 2 and 8 h post-irradiation. The P6 BMSCs had a lower percentage of cells in S phase and a higher percentage in G1 phase than the P10 BMSCs at 2 and 8 h post-irradiation. The radiation had similar effects on the senescent cell level and impaired immunomodulation capacity of the P6 and P10 BMSCs. Regardless of whether they were irradiated, the P6 and P10 BMSCs always expressed a distinctive set of genes. The upregulated genes were enriched in pathways including the cell cycle, DNA replication and oocyte meiosis. Then, a subset of conserved irradiation response genes across the BMSCs was identified, comprising 12 differentially upregulated genes and 5 differentially downregulated genes. These genes were especially associated with the p53 signaling pathway, DNA damage and DNA repair. Furthermore, validation experiments revealed that the mRNA and protein levels of these conserved genes were different between the P6 and P10 BMSCs after irradiation. Weighted gene co-expression network analysis supported these findings and further revealed the effects of cell passage on the irradiation response in BMSCs. CONCLUSION The results indicated that cell passage in vitro affected the irradiation response of BMSCs via molecular mechanisms that mediated differences in apoptosis, the cell cycle, senescence and the cytokine secretion. Thus, accurate cell passage information is not only important for transplantation therapy but also for future studies on the radiation response in BMSCs.
Collapse
Affiliation(s)
- Yang Xiang
- Department of Blood Transfusion, Irradiation biology laboratory, The Second Affiliated Hospital, Army Medical University, Xinqiao Road, Shapingba, Chongqing, 400037 China
| | - Chun Wu
- Department of Blood Transfusion, Irradiation biology laboratory, The Second Affiliated Hospital, Army Medical University, Xinqiao Road, Shapingba, Chongqing, 400037 China
- Central Laboratory, The Second Affiliated Hospital, Army Medical University, Xinqiao Road, Shapingba, Chongqing, 400037 China
| | - Jiang Wu
- Department of Blood Transfusion, Irradiation biology laboratory, The Second Affiliated Hospital, Army Medical University, Xinqiao Road, Shapingba, Chongqing, 400037 China
| | - Weili Quan
- Center for Genome Analysis, ABLife Inc., Wuhan, 430075 Hubei China
| | - Chao Cheng
- Center for Genome Analysis, ABLife Inc., Wuhan, 430075 Hubei China
| | - Jian Zhou
- Center for Genome Analysis, ABLife Inc., Wuhan, 430075 Hubei China
| | - Li Chen
- Department of Blood Transfusion, Irradiation biology laboratory, The Second Affiliated Hospital, Army Medical University, Xinqiao Road, Shapingba, Chongqing, 400037 China
| | - Lixin Xiang
- Department of Blood Transfusion, Irradiation biology laboratory, The Second Affiliated Hospital, Army Medical University, Xinqiao Road, Shapingba, Chongqing, 400037 China
| | - Fengjie Li
- Department of Blood Transfusion, Irradiation biology laboratory, The Second Affiliated Hospital, Army Medical University, Xinqiao Road, Shapingba, Chongqing, 400037 China
| | - Kebin Zhang
- Central Laboratory, The Second Affiliated Hospital, Army Medical University, Xinqiao Road, Shapingba, Chongqing, 400037 China
| | - Qian Ran
- Department of Blood Transfusion, Irradiation biology laboratory, The Second Affiliated Hospital, Army Medical University, Xinqiao Road, Shapingba, Chongqing, 400037 China
| | - Yi Zhang
- Center for Genome Analysis, ABLife Inc., Wuhan, 430075 Hubei China
| | - Zhongjun Li
- Department of Blood Transfusion, Irradiation biology laboratory, The Second Affiliated Hospital, Army Medical University, Xinqiao Road, Shapingba, Chongqing, 400037 China
| |
Collapse
|
12
|
Zuo R, Liu M, Wang Y, Li J, Wang W, Wu J, Sun C, Li B, Wang Z, Lan W, Zhang C, Shi C, Zhou Y. BM-MSC-derived exosomes alleviate radiation-induced bone loss by restoring the function of recipient BM-MSCs and activating Wnt/β-catenin signaling. Stem Cell Res Ther 2019; 10:30. [PMID: 30646958 PMCID: PMC6334443 DOI: 10.1186/s13287-018-1121-9] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 12/12/2018] [Accepted: 12/20/2018] [Indexed: 02/07/2023] Open
Abstract
Background Radiotherapy to cancer patients is inevitably accompanied by normal tissue injury, and the bone is one of the most commonly damaged tissues. Damage to bone marrow mesenchymal stem cells (BM-MSCs) induced by radiation is thought to be a major cause of radiation-induced bone loss. Exosomes exhibit great therapeutic potential in the treatment of osteoporosis, but whether exosomes are involved in radiation-induced bone loss has not been thoroughly elucidated to date. The main purpose of this study is to investigate the role of exosomes derived from BM-MSCs in restoring recipient BM-MSC function and alleviating radiation-induced bone loss. Methods BM-MSC-derived exosomes were intravenously injected to rats immediately after irradiation. After 28 days, the left tibiae were harvested for micro-CT and histomorphometric analysis. The effects of exosomes on antioxidant capacity, DNA damage repair, proliferation, and cell senescence of recipient BM-MSCs were determined. Osteogenic and adipogenic differentiation assays were used to detect the effects of exosomes on the differentiation potential of recipient BM-MSCs, and related genes were measured by qRT-PCR and Western blot analysis. β-Catenin expression was detected at histological and cytological levels. Results BM-MSC-derived exosomes can attenuate radiation-induced bone loss in a rat model that is similar to mesenchymal stem cell transplantation. Exosome-treated BM-MSCs exhibit reduced oxidative stress, accelerated DNA damage repair, and reduced proliferation inhibition and cell senescence-associate protein expression compared with BM-MSCs that exclusively received irradiation. Following irradiation, exosomes promote β-catenin expression in BM-MSCs and restore the balance between adipogenic and osteogenic differentiation. Conclusions Our findings indicate that BM-MSC-derived exosomes take effects by restoring the function of recipient BM-MSCs. Therefore, exosomes may represent a promising cell-free therapeutic approach for the treatment of radiation-induced bone loss. Electronic supplementary material The online version of this article (10.1186/s13287-018-1121-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rui Zuo
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Minghan Liu
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Yanqiu Wang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Jie Li
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Wenkai Wang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Junlong Wu
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Chao Sun
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Bin Li
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Ziwen Wang
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University(Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Weiren Lan
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Chao Zhang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Chunmeng Shi
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical University(Third Military Medical University), Chongqing, 400038, People's Republic of China.
| | - Yue Zhou
- Department of Orthopedics, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China.
| |
Collapse
|
13
|
Zhang Y, Ding H, Wang X, Ye SD. Modulation of STAT3 phosphorylation by PTPN2 inhibits naïve pluripotency of embryonic stem cells. FEBS Lett 2018; 592:2227-2237. [PMID: 29797458 DOI: 10.1002/1873-3468.13112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 04/12/2018] [Accepted: 04/27/2018] [Indexed: 01/18/2023]
Abstract
STAT3 phosphorylation at tyrosine 705 (STAT3pY705 ), triggered by the addition of the leukemia inhibitory factor (LIF), can maintain mouse embryonic stem cell (mESC) self-renewal and reprogram mouse epiblast stem cells (EpiSCs) to enter a naïve pluripotent state. The activation of STAT3pY705 occurs mainly through Janus kinases. However, it remains unclear how STAT3pY705 levels are decreased in mESCs. Our study shows that upregulation of the protein tyrosine phosphatase (PTPN2) inhibits STAT3 activity by reducing its phosphorylation level and promotes mESC differentiation, whereas PTPN2 knockout by CRISPR/CAS9 delays mESC differentiation. Consistently, PTPN2 knockdown facilitates the generation of mESC-like colonies in STAT3-overexpressing EpiSCs. PTPN2-mediated STAT3 activity, thus, contributes to the exit of ESCs from the pluripotent ground state. These findings expand the current understanding of the regulatory network of naïve pluripotency.
Collapse
Affiliation(s)
- Yan Zhang
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei, China
| | - Huiwen Ding
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei, China
| | - Xiaohu Wang
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei, China
| | - Shou-Dong Ye
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei, China
| |
Collapse
|
14
|
Li M, Xie Z, Wang P, Li J, Liu W, Tang S, Liu Z, Wu X, Wu Y, Shen H. The long noncoding RNA GAS5 negatively regulates the adipogenic differentiation of MSCs by modulating the miR-18a/CTGF axis as a ceRNA. Cell Death Dis 2018; 9:554. [PMID: 29748618 PMCID: PMC5945827 DOI: 10.1038/s41419-018-0627-5] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/19/2018] [Accepted: 04/23/2018] [Indexed: 12/21/2022]
Abstract
Mesenchymal stem cells (MSCs) are important pluripotent stem cells and a major source of adipocytes in the body. However, the mechanism of adipogenic differentiation has not yet been completely elucidated. In this study, the long noncoding RNA GAS5 was found to be negatively correlated with MSC adipogenic differentiation. GAS5 overexpression negatively regulated adipocyte formation, whereas GAS5 knockdown had the opposite effect. Further mechanistic analyses using luciferase reporter assays revealed that GAS5 regulates the adipogenic differentiation of MSCs by acting as competing endogenous RNA (ceRNA) to sponge miR-18a, which promotes adipogenic differentiation. Mutation of the binding sites for GAS5 in miR-18a abolished the effect of the interaction. The miR-18a mimic and inhibitor reversed the negative regulatory effect of GAS5 on MSCs adipogenic differentiation. In addition, GAS5 inhibited miR-18a, which downregulates connective tissue growth factor (CTGF) expression, to negatively regulate the adipogenic differentiation of MSCs. Taken together, the results show that GAS5 serves as a sponge for miR-18a, inhibiting its capability to suppress CTGF protein translation and ultimately decreasing the adipogenic differentiation of MSCs. GAS5 is an important molecule involved in the adipogenic differentiation of MSCs and may contribute to the functional regulation and clinical applications of MSCs.
Collapse
Affiliation(s)
- Ming Li
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, People's Republic of China
| | - Zhongyu Xie
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, People's Republic of China
| | - Peng Wang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, People's Republic of China
| | - Jinteng Li
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, People's Republic of China
| | - Wenjie Liu
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, People's Republic of China
| | - Su'an Tang
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, People's Republic of China
| | - Zhenhua Liu
- Department of Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, 510120, People's Republic of China
| | - Xiaohua Wu
- Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, People's Republic of China
| | - Yanfeng Wu
- Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, People's Republic of China.
| | - Huiyong Shen
- Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, People's Republic of China.
| |
Collapse
|
15
|
Chen L, Ran Q, Xiang Y, Xiang L, Chen L, Li F, Wu J, Wu C, Li Z. Co-Activation of PKC-δ by CRIF1 Modulates Oxidative Stress in Bone Marrow Multipotent Mesenchymal Stromal Cells after Irradiation by Phosphorylating NRF2 Ser40. Theranostics 2017; 7:2634-2648. [PMID: 28819452 PMCID: PMC5558558 DOI: 10.7150/thno.17853] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Accepted: 04/19/2017] [Indexed: 12/26/2022] Open
Abstract
The high mortality associated with pancytopenia and multi-organ failure resulting from hematopoietic disorders of acute radiation syndrome (h-ARS) creates an urgent need for developing more effective treatment strategies. Here, we showed that bone marrow multipotent mesenchymal stromal cells (BMMSCs) effectively regulate oxidative stress following radiative injury, which might be on account of irradiation-induced elevation of protein levels of CR6-interacting factor 1(CRIF1) and nuclear factor E2-related factor 2(NRF2). Crif1-knockdown BMMSCs presented increased oxidative stress and apoptosis after irradiation, which were partially due to a suppressed antioxidant response mediated by decreased NRF2 nuclear translocation. Co-immunoprecipitation (Co-IP) experiments indicated that CRIF1 interacted with protein kinase C-δ (PKC-δ). NRF2 Ser40 phosphorylation was inhibited in Crif1-deficient BMMSCs even in the presence of three kinds of PKC agonists, suggesting that CRIF1 might co-activate PKC-δ to phosphorylate NRF2 Ser40. After radiative injury, the supporting effect of BMMSCs for the colony forming ability of HSCs in vitro was reduced, and the deficiency of CRIF1 aggravated such damage. Thus, CRIF1 plays an essential role in PKC-δ/NRF2 pathway modulation to alleviate oxidative stress in BMMSCs after irradiative injury, and at some level it may maintain the HSCs-supporting effect of BMMSCs after radiative injuries.
Collapse
|
16
|
Yan Q, Ahn SH, Medie FM, Sharma-Kuinkel BK, Park LP, Scott WK, Deshmukh H, Tsalik EL, Cyr DD, Woods CW, Yu CHA, Adams C, Qi R, Hansen B, Fowler VG. Candidate genes on murine chromosome 8 are associated with susceptibility to Staphylococcus aureus infection in mice and are involved with Staphylococcus aureus septicemia in humans. PLoS One 2017; 12:e0179033. [PMID: 28594911 PMCID: PMC5464679 DOI: 10.1371/journal.pone.0179033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 05/23/2017] [Indexed: 02/06/2023] Open
Abstract
We previously showed that chromosome 8 of A/J mice was associated with susceptibility to S. aureus infection. However, the specific genes responsible for this susceptibility are unknown. Chromosome substitution strain 8 (CSS8) mice, which have chromosome 8 from A/J but an otherwise C57BL/6J genome, were used to identify the genetic determinants of susceptibility to S. aureus on chromosome 8. Quantitative trait loci (QTL) mapping of S. aureus-infected N2 backcross mice (F1 [C8A] × C57BL/6J) identified a locus 83180780–88103009 (GRCm38/mm10) on A/J chromosome 8 that was linked to S. aureus susceptibility. All genes on the QTL (n~ 102) were further analyzed by three different strategies: 1) different expression in susceptible (A/J) and resistant (C57BL/6J) mice only in response to S. aureus, 2) consistently different expression in both uninfected and infected states between the two strains, and 3) damaging non-synonymous SNPs in either strain. Eleven candidate genes from the QTL region were significantly differently expressed in patients with S. aureus infection vs healthy human subjects. Four of these 11 genes also exhibited significantly different expression in S. aureus-challenged human neutrophils: Ier2, Crif1, Cd97 and Lyl1. CD97 ligand binding was evaluated within peritoneal neutrophils from A/J and C57BL/6J. CD97 from A/J had stronger CD55 but weaker integrin α5β1 ligand binding as compared with C57BL/6J. Because CD55/CD97 binding regulates immune cell activation and cytokine production, and integrin α5β1 is a membrane receptor for fibronectin, which is also bound by S. aureus, strain-specific differences could contribute to susceptibility to S. aureus. Down-regulation of Crif1 with siRNA was associated with increased host cell apoptosis among both naïve and S. aureus-infected bone marrow-derived macrophages. Specific genes in A/J chromosome 8, including Cd97 and Crif1, may play important roles in host defense against S. aureus.
Collapse
Affiliation(s)
- Qin Yan
- Division of Infectious Diseases & International Health, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Sun Hee Ahn
- Department of Biochemistry School of Dentistry, Chonnam National University, Bukgu, Gwangju, Korea
| | - Felix Mba Medie
- Division of Infectious Diseases & International Health, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Batu K. Sharma-Kuinkel
- Division of Infectious Diseases & International Health, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Lawrence P. Park
- Duke Global Health Institute, Duke University, Durham, North Carolina, United States of America
| | - William K. Scott
- Dr. John T. Macdonald Foundation Department of Human Genetics and John P. Hussman Institute for Human Genomics, University of Miami, Miami, Florida, United States of America
| | - Hitesh Deshmukh
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Ephraim L. Tsalik
- Division of Infectious Diseases & International Health, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
- Emergency Medicine Service, Durham Veteran’s Affairs Medical Center, Durham, North Carolina, United States of America
- Duke Clinical Research Institute, Durham, North Carolina, United States of America
| | - Derek D. Cyr
- Duke Clinical Research Institute, Durham, North Carolina, United States of America
| | - Christopher W. Woods
- Division of Infectious Diseases & International Health, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Clinical Research Institute, Durham, North Carolina, United States of America
- Section on Infectious Diseases, Durham Veteran’s Affairs Medical Center, Durham, North Carolina, United States of America
| | - Chen-Hsin Albert Yu
- Division of Infectious Diseases & International Health, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Carlton Adams
- Division of Infectious Diseases & International Health, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Robert Qi
- Division of Infectious Diseases & International Health, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Brenda Hansen
- Division of Infectious Diseases & International Health, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Vance G. Fowler
- Division of Infectious Diseases & International Health, Department of Medicine, Duke University School of Medicine, Durham, North Carolina, United States of America
- Duke Clinical Research Institute, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
17
|
Yan HX, Zhang YJ, Zhang Y, Ren X, Shen YF, Cheng MB, Zhang Y. CRIF1 enhances p53 activity via the chromatin remodeler SNF5 in the HCT116 colon cancer cell lines. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2017; 1860:516-522. [DOI: 10.1016/j.bbagrm.2017.02.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 02/17/2017] [Accepted: 02/18/2017] [Indexed: 12/28/2022]
|
18
|
Misra J, Mohanty ST, Madan S, Fernandes JA, Hal Ebetino F, Russell RGG, Bellantuono I. Zoledronate Attenuates Accumulation of DNA Damage in Mesenchymal Stem Cells and Protects Their Function. Stem Cells 2015; 34:756-67. [PMID: 26679354 PMCID: PMC4832316 DOI: 10.1002/stem.2255] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 09/18/2015] [Accepted: 10/05/2015] [Indexed: 11/18/2022]
Abstract
Mesenchymal stem cells (MSCs) undergo a decline in function following ex vivo expansion and exposure to irradiation. This has been associated with accumulation of DNA damage and has important implications for tissue engineering approaches or in patients receiving radiotherapy. Therefore, interventions, which limit accumulation of DNA damage in MSC, are of clinical significance. We were intrigued by findings showing that zoledronate (ZOL), an anti‐resorptive nitrogen containing bisphosphonate, significantly extended survival in patients affected by osteoporosis. The effect was too large to be simply due to the prevention of fractures. Moreover, in combination with statins, it extended the lifespan in a mouse model of Hutchinson Gilford Progeria Syndrome. Therefore, we asked whether ZOL was able to extend the lifespan of human MSC and whether this was due to reduced accumulation of DNA damage, one of the important mechanisms of aging. Here, we show that this was the case both following expansion and irradiation, preserving their ability to proliferate and differentiate in vitro. In addition, administration of ZOL before irradiation protected the survival of mesenchymal progenitors in mice. Through mechanistic studies, we were able to show that inhibition of mTOR signaling, a pathway involved in longevity and cancer, was responsible for these effects. Our data open up new opportunities to protect MSC from the side effects of radiotherapy in cancer patients and during ex vivo expansion for regenerative medicine approaches. Given that ZOL is already in clinical use with a good safety profile, these opportunities can be readily translated for patient benefit. Stem Cells2016;34:756–767
Collapse
Affiliation(s)
- Juhi Misra
- Mellanby Centre for Bone Research and MRC-Arthritis Research Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Department of Human Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Sindhu T Mohanty
- Mellanby Centre for Bone Research and MRC-Arthritis Research Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Department of Human Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Sanjeev Madan
- Department of Paediatric Orthopaedic and Trauma Surgery, Sheffield Children's Hospital, Sheffield, United Kingdom
| | - James A Fernandes
- Department of Paediatric Orthopaedic and Trauma Surgery, Sheffield Children's Hospital, Sheffield, United Kingdom
| | - F Hal Ebetino
- Department of Chemistry, University of Rochester, Rochester, New York, USA.,Structural Genomics Consortium, University of Oxford, United Kingdom
| | - R Graham G Russell
- Mellanby Centre for Bone Research and MRC-Arthritis Research Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Department of Human Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom.,Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Oxford NIHR Biomedical Research Unit, The Oxford University Institute of Musculoskeletal Sciences, The Botnar Research Centre, Headington, Oxford, United Kingdom
| | - Ilaria Bellantuono
- Mellanby Centre for Bone Research and MRC-Arthritis Research Centre for Integrated Research into Musculoskeletal Ageing (CIMA), Department of Human Metabolism, Medical School, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|