1
|
Jin B, Su G, Zhou X, Xu L, Wang W, Zhou T, Tan Y, Wang S, Li G. Basic Fibroblast Growth Factor Supports the Function of Limbal Niche Cells via the Wnt/β-Catenin Pathway. J Ocul Pharmacol Ther 2024. [PMID: 39083404 DOI: 10.1089/jop.2024.0042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2024] Open
Abstract
Purpose: To test the effects and underlying mechanisms of basic fibroblast growth factor (bFGF) on the limbal niche cell (LNC) function ex vivo. Methods: By using different concentrations of bFGF (0, 4, 8, 12, and 16 ng/mL) and fibroblast growth factor receptor (FGFR) inhibitors, the effects of bFGF on LNC proliferation, expression of stem cell markers, and transcription levels of the β-catenin were investigated. Single-cell RNA sequencing (scRNA-seq) was used to analyze the action and mechanisms of FGFR subtypes and the Wnt/β-catenin pathway during LNC culture. An mature corneal epithelial cell (MCEC)/LNC three-dimensional model was constructed to verify whether bFGF activates the Wnt/β-catenin pathway in LNC by inhibiting FGFR or β-catenin targets. Results: scRNA-seq showed that FGFR1 is the main receptor in LNC, along with the molecules in the Wnt pathway, including WNT2, FZD7, LRP5, LRP6, and β-catenin. The 12 ng/mL bFGF treatment group showed higher LNC proliferation rate and transcription levels of OCT4, SOX2, NANOG, and β-catenin than any other groups (P < 0.001). In the MCEC/LNC co-culture model, MCEC/LNC treated with 12 ng/mL bFGF promoted the aggregation of the spheres than other groups, associated with increased transcription levels of P63α, WNT2, β-catenin, and a decreased transcription level of CK12 (P < 0.001). Wnt/β-catenin inhibitor LF3 treatment reversed the abovementioned effect of bFGF. Conclusions: bFGF could maintain and promote the stemness of LNC via the FGFR1/Wnt2/FZD7/LRP6 axis in a concentration-dependent manner.
Collapse
Affiliation(s)
- Bihui Jin
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guanyu Su
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao Zhou
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lingjuan Xu
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Wang
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tianyu Zhou
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongyao Tan
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shusheng Wang
- Department of Cell and Molecular Biology & Ophthalmology, Tulane University, New Orleans, Louisiana, USA
| | - Guigang Li
- Department of Ophthalmology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
2
|
He J, Jiang P, Ma L, Liu F, Fu P, Du X, Xu Z, Xu J, Cheng L, Wang Z, Li C, Liu D. Intravenous immunoglobulin protects the integrity of the intestinal epithelial barrier and inhibits ferroptosis induced by radiation exposure by activating the mTOR pathway. Int Immunopharmacol 2024; 131:111908. [PMID: 38518594 DOI: 10.1016/j.intimp.2024.111908] [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: 01/24/2024] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 03/24/2024]
Abstract
Radiation exposure often leads to serious health problems in humans. The intestinal epithelium is sensitive to radiation damage, and radiation causes destruction of the intestinal epithelial barrier, which leads to radiation enteritis (RE), the loss of fluids, and the translocation of intestinal bacteria and toxins; radiation can even threaten survival. In this study, we aimed to explore the influence of IVIg on the integrity of the intestinal epithelial barrier after RE. Using a RE mouse model, we investigated the protective effects of intravenous immunoglobulin (IVIg) on the epithelial junctions of RE mice and validated these findings with intestinal organoids cultured in vitro. In addition, transmission electron microscopy (TEM), western blotting (WB) and immunostaining were used to further investigate changes in intestinal epithelial ferroptosis and related signaling pathways. When RE occurs, the intestinal epithelial barrier is severely damaged. IVIg treatment significantly ameliorated this damage to epithelial tight junctions both in vivo and in vitro. Notably, IVIg alleviated RE by inhibiting intestinal epithelial ferroptosis in RE mice. Mechanistically, IVIg promoted activation of the mTOR pathway and inhibited ferroptosis in the intestinal epithelium of mice. Rapamycin, which is a potent inhibitor of the mTOR protein, significantly abolished the protective effect of IVIg against radiation-induced damage to intestinal epithelial tight junctions. Overall, IVIg can prevent RE-induced damage to the intestinal epithelial barrier and inhibit ferroptosis by activating the mTOR pathway; this study provides a new treatment strategy for patients with RE caused by radiotherapy or accidental nuclear exposure.
Collapse
Affiliation(s)
- Jia He
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, Sichuan Province 610052, China
| | - Peng Jiang
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, Sichuan Province 610052, China
| | - Li Ma
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, Sichuan Province 610052, China
| | - Fengjuan Liu
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, Sichuan Province 610052, China
| | - Ping Fu
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, Sichuan Province 610052, China
| | - Xi Du
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, Sichuan Province 610052, China
| | - Zhenni Xu
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital & Institute, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan Province 610041, China
| | - Jun Xu
- Shanghai RAAS Blood Products Co., Ltd., Shanghai 201401, China
| | - Lu Cheng
- Shanghai RAAS Blood Products Co., Ltd., Shanghai 201401, China
| | - Zongkui Wang
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, Sichuan Province 610052, China.
| | - Changqing Li
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, Sichuan Province 610052, China.
| | - Dengqun Liu
- Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Cancer Hospital & Institute, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan Province 610041, China.
| |
Collapse
|
3
|
Park HY, Yu JH. X-ray radiation-induced intestinal barrier dysfunction in human epithelial Caco-2 cell monolayers. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 264:115404. [PMID: 37625335 DOI: 10.1016/j.ecoenv.2023.115404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 08/18/2023] [Accepted: 08/21/2023] [Indexed: 08/27/2023]
Abstract
Radiation therapy and unwanted radiological or nuclear exposure, such as nuclear plant accidents, terrorist attacks, and military conflicts, pose serious health issues to humans. Dysfunction of the intestinal epithelial barrier and the leakage of luminal antigens and bacteria across the barrier have been linked to various human diseases. Intestinal permeability is regulated by intercellular structures, termed tight junctions (TJs), which are disrupted after radiation exposure. In this study, we investigated radiation-induced alterations in TJ-related proteins in an intestinal epithelial cell model. Caco-2 cells were irradiated with 2, 5, and 10 Gy and harvested 1 and 24 h after X-ray exposure. The trypan blue assay revealed that cell viability was reduced in a dose-dependent manner 24 h after X-ray exposure compared to that of non-irradiated cells. However, the WST-8 assay revealed that cell proliferation was significantly reduced only 24 h after radiation exposure to 10 Gy compared to that of non-irradiated cells. In addition, a decreased growth rate and increased doubling time were observed in cells irradiated with X-rays. Intestinal permeability was significantly increased, and transepithelial electrical resistance values were remarkably reduced in Caco-2 cell monolayers irradiated with X-rays compared to non-irradiated cells. X-ray irradiation significantly decreased the mRNA and protein levels of ZO-1, occludin, claudin-3, and claudin-4, with ZO-1 and claudin-3 protein levels decreasing in a dose-dependent manner. Overall, the present study reveals that exposure to X-ray induces dysfunction of the human epithelial intestinal barrier and integrity via the downregulation of TJ-related genes, which may be a key factor contributing to intestinal barrier damage and increased intestinal permeability.
Collapse
Affiliation(s)
- Ha-Young Park
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup 56212, Republic of Korea.
| | - Jin-Hee Yu
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup 56212, Republic of Korea
| |
Collapse
|
4
|
Zheng T, Shi X, Nie S, Yin L, Zhu J, Yu E, Shen H, Mo F. Effects of Chinese herbal diet on hematopoiesis, immunity, and intestines of mice exposed to different doses of radiation. Heliyon 2023; 9:e15473. [PMID: 37131450 PMCID: PMC10149268 DOI: 10.1016/j.heliyon.2023.e15473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 03/20/2023] [Accepted: 04/11/2023] [Indexed: 05/04/2023] Open
Abstract
Radiotherapy causes a series of side effects in patients with malignant tumors. Polygonati Rhizoma, Achyranthis Bidentatae Radix, and Epimedii Folium are all traditional Chinese herbs with varieties of functions such as anti-radiation and immune regulation. In this study, the above three herbs were used as a herbal diet to study their effects on the hematopoietic, immune, and intestinal systems of mice exposed to three doses of radiation. Our study showed that the diet had no radiation-protective effect on the hematopoietic and immune systems. However, at the radiation dose of 4 Gy and 8 Gy, the diet showed an obvious radiation-protective effect on intestinal crypts. At the dose of 8 Gy, we also found that the Chinese herbal diet had an anti-radiation effect on reducing the loss of the inhibitory nNOS+ neurons in the intestine. That provides a new diet for relieving the symptoms of hyperperistalsis and diarrhea in patients after radiotherapy.
Collapse
Affiliation(s)
- Tianyu Zheng
- Department of Naval Nutrition and Food Hygiene, Faculty of Naval Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai 200433, China
| | - Xiaohui Shi
- Department of Colorectal Surgery, The First Affiliated Hospital of Naval Medical University, 800 Xiangyin Road, Shanghai 200433, China
| | - Shuang Nie
- Department of Naval Nutrition and Food Hygiene, Faculty of Naval Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai 200433, China
| | - Lifeng Yin
- Department of Naval Nutrition and Food Hygiene, Faculty of Naval Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai 200433, China
| | - Jian Zhu
- Department of Naval Nutrition and Food Hygiene, Faculty of Naval Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai 200433, China
| | - Enda Yu
- Department of Colorectal Surgery, The First Affiliated Hospital of Naval Medical University, 800 Xiangyin Road, Shanghai 200433, China
| | - Hui Shen
- Department of Naval Nutrition and Food Hygiene, Faculty of Naval Medicine, Naval Medical University, 800 Xiangyin Road, Shanghai 200433, China
- Corresponding author. Department of Naval Nutrition and Food Hygiene, Faculty of Naval Medicine, Naval Medical University, 800 Xiangyin Rd, Shanghai 200433, China.
| | - Fengfeng Mo
- Department of Colorectal Surgery, The First Affiliated Hospital of Naval Medical University, 800 Xiangyin Road, Shanghai 200433, China
- Corresponding author. Department of Naval Nutrition and Food Hygiene, Faculty of Naval Medicine, Naval Medical University, 800 Xiangyin Rd, Shanghai 200433, China.
| |
Collapse
|
5
|
Wang Y, Ren K, Tan J, Mao Y. Alginate oligosaccharide alleviates aging-related intestinal mucosal barrier dysfunction by blocking FGF1-mediated TLR4/NF-κB p65 pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 116:154806. [PMID: 37236046 DOI: 10.1016/j.phymed.2023.154806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 03/30/2023] [Accepted: 04/04/2023] [Indexed: 05/28/2023]
Abstract
BACKGROUND Alginate oligosaccharide (AOS) has been reported to exert a crucial role in maintaining the intestinal mucosal barrier (IMB) function. The current study aimed at ascertaining the protective effects of AOS on aging-induced IMB dysfunction and to elucidate the underlying molecular mechanisms. METHODS An aging mouse model and a senescent NCM460 cell model were established using d-galactose. AOS was administered to aging mice and senescent cells, and IMB permeability, inflammatory response and tight junction proteins were assessed. In silico analysis was conducted to identify factors regulated by AOS. Using gain- and loss-of-function approaches, we evaluated the roles of FGF1, TLR4 and NF-κB p65 in the aging-induced IMB dysfunction and NCM460 cell senescence. RESULTS AOS protected the IMB function of aging mice and NCM460 cells by reducing permeability and increasing tight junction proteins. In addition, AOS up-regulated FGF1, which blocked the TLR4/NF-κB p65 pathway, and identified as the mechanism responsible for the protective effect of AOS. CONCLUSION AOS blocks the TLR4/NF-κB p65 pathway via inducing FGF1, ultimately reducing the risk of IMB dysfunction in aging mice. This study highlights the potential of AOS as a protective agent against aging-induced IMB disorder and provides insight into the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Yanting Wang
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, Shandong 266000, China
| | - Keyu Ren
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, Shandong 266000, China
| | - Junying Tan
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, Shandong 266000, China
| | - Yongjun Mao
- Department of Geriatric Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| |
Collapse
|
6
|
Yang Q, Qin B, Hou W, Qin H, Yin F. Pathogenesis and therapy of radiation enteritis with gut microbiota. Front Pharmacol 2023; 14:1116558. [PMID: 37063268 PMCID: PMC10102376 DOI: 10.3389/fphar.2023.1116558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 03/07/2023] [Indexed: 04/03/2023] Open
Abstract
Radiotherapy is widely used in clinic due to its good effect for cancer treatment. But radiotherapy of malignant tumors in the abdomen and pelvis is easy to cause radiation enteritis complications. Gastrointestinal tract contains numerous microbes, most of which are mutualistic relationship with the host. Abdominal radiation results in gut microbiota dysbiosis. Microbial therapy can directly target gut microbiota to reverse microbiota dysbiosis, hence relieving intestinal inflammation. In this review, we mainly summarized pathogenesis and novel therapy of the radiation-induced intestinal injury with gut microbiota dysbiosis and envision the opportunities and challenges of radiation enteritis therapy.
Collapse
Affiliation(s)
- Qilin Yang
- Research Institute of Intestinal Diseases, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- School of Clinical Medicine of Nanjing Medical University, Nanjing, China
| | - Bingzhi Qin
- Research Institute of Intestinal Diseases, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
| | - Weiliang Hou
- Research Institute of Intestinal Diseases, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- Shanghai Cancer Institute, Renji Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Weiliang Hou, ; Huanlong Qin, ; Fang Yin,
| | - Huanlong Qin
- Research Institute of Intestinal Diseases, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- *Correspondence: Weiliang Hou, ; Huanlong Qin, ; Fang Yin,
| | - Fang Yin
- Research Institute of Intestinal Diseases, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- *Correspondence: Weiliang Hou, ; Huanlong Qin, ; Fang Yin,
| |
Collapse
|
7
|
Liu Y, Liu Z, Hu L, He L, Yang L, Qin Z, Xie Y, Peng X, Dai L. Function of stem cells in radiation-induced damage. Int J Radiat Biol 2023; 99:1483-1494. [PMID: 36912588 DOI: 10.1080/09553002.2023.2188935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 02/27/2023] [Indexed: 03/14/2023]
Abstract
PURPOSE The aim of this review is to discuss previous studies on the function of stem cells in radiation-induced damage, and the factors affecting these processes, in the hope of improving our understanding of the different stem cells and the communication networks surrounding them. This is essential for the development of effective stem cell-based therapies to regenerate or replace normal tissues damaged by radiation. CONCLUSION In salivary glands, senescence-associated cytokines and inflammation-associated cells have a greater effect on stem cells. In the intestinal glands, Paneth cells strongly affect stem cell-mediated tissue regeneration after radiation treatment. In the pancreas, β-cells as well as protein C receptor positive (Procr) cells are expected to be key cells in the treatment of diabetes. In the bone marrow, a variety of cytokines such as CXC-chemokine ligand 12 (CXCL12) and stem cell factor (SCF), contribute to the functional recovery of hematopoietic stem cells after irradiation.
Collapse
Affiliation(s)
- Yingtong Liu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, and Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Zheran Liu
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Liqiang Hu
- West China-California Research Center for Predictive Intervention Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Ling He
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Lianlian Yang
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Zijian Qin
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yuping Xie
- Department of Oncology, Chengdu First People's Hospital, Chengdu, Sichuan, China
| | - Xingchen Peng
- Department of Biotherapy, Cancer Center, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Lei Dai
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
8
|
Dong Y, Zhang Y, Wang X, Li W, Zhang J, Lu L, Dong H, Fan S, Meng A, Li D. The protective effects of Xuebijing injection on intestinal injuries of mice exposed to irradiation. Animal Model Exp Med 2022; 5:565-574. [PMID: 36376997 PMCID: PMC9773304 DOI: 10.1002/ame2.12285] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 10/06/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Gastrointestinal (GI) injury is one of the most common side effects of radiotherapy. However, there is no ideal therapy method except for symptomatic treatment in the clinic. Xuebijing (XBJ) is a traditional Chinese medicine, used to treat sepsis by injection. In this study, the protective effects of XBJ on radiation-induced intestinal injury (RIII) and its mechanism were explored. METHODS The effect of XBJ on survival of irradiated C57BL/6 mice was monitored. Histological changes including the number of crypts and the length of villi were evaluated by H&E. The expression of Lgr5+ intestinal stem cells (ISCs), Ki67+ cells, villin and lysozymes were examined by immunohistochemistry. The expression of cytokines in the intestinal crypt was detected by RT-PCR. DNA damage and apoptosis rates in the small intestine were also evaluated by immunofluorescence. RESULTS In the present study, XBJ improved the survival rate of the mice after 8.0 and 9.0 Gy total body irradiation (TBI). XBJ attenuated structural damage of the small intestine, maintained regenerative ability and promoted proliferation and differentiation of crypt cells, decreased apoptosis rate and reduced DNA damage in the intestine. Elevation of IL-6 and TNF-α was limited, but IL-1, TNF-𝛽 and IL-10 levels were increased in XBJ-treated group after irradiation. The expression of Bax and p53 were decreased after XBJ treatment. CONCLUSIONS Taken together, XBJ provides a protective effect on RIII by inhibiting inflammation and blocking p53-related apoptosis pathway.
Collapse
Affiliation(s)
- Yinping Dong
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Science & Peking Union Medical CollegeTianjinChina
| | - YuanYang Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Science & Peking Union Medical CollegeTianjinChina
| | - Xinyue Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Science & Peking Union Medical CollegeTianjinChina
| | - Wenxuan Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Science & Peking Union Medical CollegeTianjinChina
| | - Junling Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Science & Peking Union Medical CollegeTianjinChina
| | - Lu Lu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Science & Peking Union Medical CollegeTianjinChina
| | - Hui Dong
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Science & Peking Union Medical CollegeTianjinChina
| | - Saijun Fan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Science & Peking Union Medical CollegeTianjinChina
| | - Aimin Meng
- Key Laboratory of Human Disease Comparative Medicine, Ministry of Health, Beijing Engineering Research Center for Laboratory Animal Models of Human Critical Diseases, National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal SciencesChinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC)BeijingChina
| | - Deguan Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation MedicineChinese Academy of Medical Science & Peking Union Medical CollegeTianjinChina
| |
Collapse
|
9
|
Zhou S, Li Y, He L, Chen M, Li W, Xiao T, Guan J, Qi Z, Wang Q, Li S, Zhou P, Wang Z. Haptoglobin is an early indicator of survival after radiation-induced severe injury and bone marrow transplantation in mice. Stem Cell Res Ther 2022; 13:461. [PMID: 36068556 PMCID: PMC9450283 DOI: 10.1186/s13287-022-03162-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 05/23/2022] [Indexed: 11/15/2022] Open
Abstract
Background Hematopoietic stem cell transplantation (HSCT) is the main treatment for acute radiation sickness, especially after fatal radiation. The determination of HSCT for radiation patients is mainly based on radiation dose, hemogram and bone marrow injury severity. This study aims to explore a better biomarker of acute radiation injury from the perspective of systemic immune response.
Methods C57BL/6J female mice were exposed to total body irradiation (TBI) and partial body irradiation (PBI). Changes in haptoglobin (Hp) level in plasma were shown at different doses and time points after the exposure and treatment with amifostine or bone marrow transplantation. Student’s t-test/two tailed test were used in two groups. To decide the Hp levels as a predictor of the radiation dose in TBI and PBI, multiple linear regression analysis were performed. The ability of biomarkers to identify two groups of different samples was determined by the receiver operating characteristic (ROC) curve. The results were expressed as mean ± standard deviation (SD). Significance was set at P value < 0.05, and P value < 0.01 was set as highly significant. Survival distribution was determined by log-rank test. Results In this study, we found that Hp was elevated dose-dependently in plasma in the early post-irradiation period and decreased on the second day, which can be used as a molecular indicator for early dose assessment. Moreover, we detected the second increase of Hp on the 3rd and 5th days after the lethal irradiation at 10 Gy, which was eliminated by amifostine, a radiation protection drug, while protected mice from death. Most importantly, bone marrow transplantation (BMT) on the 3rd and 5th day after 10 Gy radiation improved the 30-days survival rate, and effectively accelerated the regression of secondary increased Hp level. Conclusions Our study suggests that Hp can be used not only as an early molecule marker of radiation injury, but also as an important indicator of bone marrow transplantation therapy for radiation injury, bringing new scientific discoveries in the diagnosis and treatment of acute radiation injury from the perspective of systemic immunity. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03162-x.
Collapse
Affiliation(s)
- Shixiang Zhou
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Yaqiong Li
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Lexin He
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Min Chen
- Department of Radiotherapy, Nanfang Hospital, Southern Medical University, Guangzhou, 510080, China
| | - Weihong Li
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, China.,Graduate Collaborative Training Base of Academy of Military Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Ting Xiao
- Department of Radiotherapy, Nanfang Hospital, Southern Medical University, Guangzhou, 510080, China
| | - Jian Guan
- Department of Radiotherapy, Nanfang Hospital, Southern Medical University, Guangzhou, 510080, China
| | - Zhenhua Qi
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Qi Wang
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, China
| | - Siyuan Li
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, China.,Graduate Collaborative Training Base of Academy of Military Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Pingkun Zhou
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, China.
| | - Zhidong Wang
- Department of Radiobiology, Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing, 100850, China. .,Graduate Collaborative Training Base of Academy of Military Sciences, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
10
|
Forsberg MH, Kink JA, Thickens AS, Lewis BM, Childs CJ, Hematti P, Capitini CM. Exosomes from primed MSCs can educate monocytes as a cellular therapy for hematopoietic acute radiation syndrome. Stem Cell Res Ther 2021; 12:459. [PMID: 34407878 PMCID: PMC8371870 DOI: 10.1186/s13287-021-02491-7] [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: 02/17/2021] [Accepted: 07/04/2021] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Acute radiation syndrome (ARS) is caused by acute exposure to ionizing radiation that damages multiple organ systems but especially the bone marrow (BM). We have previously shown that human macrophages educated with exosomes from human BM-derived mesenchymal stromal cells (MSCs) primed with lipopolysaccharide (LPS) prolonged survival in a xenogeneic lethal ARS model. The purpose of this study was to determine if exosomes from LPS-primed MSCs could directly educate human monocytes (LPS-EEMos) for the treatment of ARS. METHODS Human monocytes were educated by exosomes from LPS-primed MSCs and compared to monocytes educated by unprimed MSCs (EEMos) and uneducated monocytes to assess survival and clinical improvement in a xenogeneic mouse model of ARS. Changes in surface molecule expression of exosomes and monocytes after education were determined by flow cytometry, while gene expression was determined by qPCR. Irradiated human CD34+ hematopoietic stem cells (HSCs) were co-cultured with LPS-EEMos, EEMos, or uneducated monocytes to assess effects on HSC survival and proliferation. RESULTS LPS priming of MSCs led to the production of exosomes with increased expression of CD9, CD29, CD44, CD146, and MCSP. LPS-EEMos showed increases in gene expression of IL-6, IL-10, IL-15, IDO, and FGF-2 as compared to EEMos generated from unprimed MSCs. Generation of LPS-EEMos induced a lower percentage of CD14+ monocyte subsets that were CD16+, CD73+, CD86+, or CD206+ but a higher percentage of PD-L1+ cells. LPS-EEMos infused 4 h after lethal irradiation significantly prolonged survival, reducing clinical scores and weight loss as compared to controls. Complete blood counts from LPS-EEMo-treated mice showed enhanced hematopoietic recovery post-nadir. IL-6 receptor blockade completely abrogated the radioprotective survival benefit of LPS-EEMos in vivo in female NSG mice, but only loss of hematopoietic recovery was noted in male NSG mice. PD-1 blockade had no effect on survival. Furthermore, LPS-EEMos also showed benefits in vivo when administered 24 h, but not 48 h, after lethal irradiation. Co-culture of unprimed EEMos or LPS-EEMos with irradiated human CD34+ HSCs led to increased CD34+ proliferation and survival, suggesting hematopoietic recovery may be seen clinically. CONCLUSION LPS-EEMos are a potential counter-measure for hematopoietic ARS, with a reduced biomanufacturing time that facilitates hematopoiesis.
Collapse
Affiliation(s)
- Matthew H Forsberg
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, 1111 Highland Ave, WIMR 4137, Madison, WI, 53705, USA
| | - John A Kink
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA.,Department of Medicine, University of Wisconsin School of Medicine and Public Health, 1111 Highland Ave, WIMR 4033, Madison, WI, 53705, USA
| | - Anna S Thickens
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, 1111 Highland Ave, WIMR 4033, Madison, WI, 53705, USA
| | - Bryson M Lewis
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, 1111 Highland Ave, WIMR 4033, Madison, WI, 53705, USA
| | - Charlie J Childs
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, 1111 Highland Ave, WIMR 4033, Madison, WI, 53705, USA
| | - Peiman Hematti
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA. .,Department of Medicine, University of Wisconsin School of Medicine and Public Health, 1111 Highland Ave, WIMR 4033, Madison, WI, 53705, USA.
| | - Christian M Capitini
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, 1111 Highland Ave, WIMR 4137, Madison, WI, 53705, USA. .,University of Wisconsin Carbone Cancer Center, Madison, WI, USA.
| |
Collapse
|
11
|
Wu T, Gao J, Liu W, Cui J, Yang M, Guo W, Wang FY. NLRP3 protects mice from radiation-induced colon and skin damage via attenuating cGAS-STING signaling. Toxicol Appl Pharmacol 2021; 418:115495. [PMID: 33741346 DOI: 10.1016/j.taap.2021.115495] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 03/12/2021] [Accepted: 03/14/2021] [Indexed: 10/21/2022]
Abstract
In the present study, the effects of NLRP3 on radiation-induced tissue damage, including colon and skin damage in mice, and the possible mechanisms were explored in vivo and in vitro. The mice were subjected to whole abdomen radiation by timed exposure to X-ray at a cumulative dose of 14 Gy. The survival rate showed that NLRP3 deficiency increased the mortality rate in mice. Furthermore, colon damage, evaluated by H&E staining and barrier function analysis, were significantly aggravated by NLRP3 deficiency. Enhanced phosphorylation of p-TBK1 and p-IRF3 in colonic tissue as well as elevated IFN-β levels in the serum indicated hyperactivation of cGAS-STING signaling. Moreover, radiation-induced expression of p-TBK1, p-IRF3, and IFN-β in BMDMs increased in vitro after NLRP3 knockout. Thus, our study outcomes suggest that NLRP3 may protect mice from radiation-induced tissue damage via attenuating cGAS-STING signaling.
Collapse
Affiliation(s)
- Tiancong Wu
- Department of Gastroenterology and Hepatology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing 210002, China; Jinling Hospital, Department of Radiation Oncology, Nanjing University, School Medicine, Nanjing 210002, China
| | - Jianhua Gao
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing 210093, China
| | - Wen Liu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing 210093, China
| | - Jian Cui
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing 210093, China
| | - Miaofang Yang
- Department of Gastroenterology and Hepatology, Jinling Hospital, Medical School of Nanjing University, 305 Zhongshan East Road, Nanjing 210002, Jiangsu Province, China
| | - Wenjie Guo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Science, Nanjing University, Nanjing 210093, China.
| | - Fang-Yu Wang
- Department of Gastroenterology and Hepatology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing 210002, China; Department of Gastroenterology and Hepatology, Jinling Hospital, Medical School of Nanjing University, 305 Zhongshan East Road, Nanjing 210002, Jiangsu Province, China.
| |
Collapse
|
12
|
Sadeghi H, Bagheri H, Shekarchi B, Javadi A, Najafi M. Mitigation of Radiation-Induced Gastrointestinal System Injury by Melatonin: A Histopathological Study. Curr Drug Res Rev 2020; 12:72-79. [PMID: 32578524 DOI: 10.2174/2589977511666191031094625] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/04/2019] [Accepted: 10/15/2019] [Indexed: 06/11/2023]
Abstract
AIMS The current study aimed to investigate the potential role of melatonin in the mitigation of radiation-induced gastrointestinal injury. BACKGROUND Organs of the gastrointestinal system such as the intestines, colon, duodenum, ileum etc. are sensitive to ionizing radiation. Mitigation of radiation-induced gastrointestinal injury is an interesting topic in radiobiology and a life-saving approach for exposed persons after a radiation event or improving the quality of life of radiotherapy patients. OBJECTIVE The study aimed to find the possible mitigation effect of melatonin on radiation-induced damage to the small and large intestines. METHODS 40 male mice were randomly assigned into four groups namely G1: control, G2: melatonin treatment, G3: whole-body irradiation, and G4: melatonin treatment after whole-body irradiation. A cobalt-60 gamma-ray source was used to deliver 7 Gy to the whole body. 100 mg/kg melatonin was administered orally 24 h after irradiation and continued for 5 days. Thirty days after irradiation, histopathological evaluations were performed. RESULTS The whole-body irradiation led to remarkable inflammation, villi shortening, apoptosis and damage to goblet cells of the small intestine. Furthermore, moderate to severe inflammation, apoptosis, congestion, crypt injury and goblet cell damage were reported for the colon. Treatment with melatonin after whole-body irradiation led to significant mitigation of radiation toxicity in both small and large intestines. CONCLUSION Melatonin could mitigate intestinal injury following whole-body exposure to radiation. Treatment with melatonin after an accidental exposure to radiation may increase survival via mitigation of damages to radiosensitive organs, including the gastrointestinal system.
Collapse
Affiliation(s)
- Hossein Sadeghi
- AJA Radiation Sciences Radiation Sciences (ARSRC), Tehran, Iran
| | - Hamed Bagheri
- AJA Radiation Sciences Radiation Sciences (ARSRC), Tehran, Iran
| | - Babak Shekarchi
- AJA Radiation Sciences Radiation Sciences (ARSRC), Tehran, Iran
| | - Abdolreza Javadi
- Department of Pathology, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
13
|
Xiao HW, Cui M, Li Y, Dong JL, Zhang SQ, Zhu CC, Jiang M, Zhu T, Wang B, Wang HC, Fan SJ. Gut microbiota-derived indole 3-propionic acid protects against radiation toxicity via retaining acyl-CoA-binding protein. MICROBIOME 2020; 8:69. [PMID: 32434586 PMCID: PMC7241002 DOI: 10.1186/s40168-020-00845-6] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 04/26/2020] [Indexed: 05/07/2023]
Abstract
BACKGROUND We have proved fecal microbiota transplantation (FMT) is an efficacious remedy to mitigate acute radiation syndrome (ARS); however, the mechanisms remain incompletely characterized. Here, we aimed to tease apart the gut microbiota-produced metabolites, underpin the therapeutic effects of FMT to radiation injuries, and elucidate the underlying molecular mechanisms. RESULTS FMT elevated the level of microbial-derived indole 3-propionic acid (IPA) in fecal pellets from irradiated mice. IPA replenishment via oral route attenuated hematopoietic system and gastrointestinal (GI) tract injuries intertwined with radiation exposure without precipitating tumor growth in male and female mice. Specifically, IPA-treated mice represented a lower system inflammatory level, recuperative hematogenic organs, catabatic myelosuppression, improved GI function, and epithelial integrity following irradiation. 16S rRNA gene sequencing and subsequent analyses showed that irradiated mice harbored a disordered enteric bacterial pattern, which was preserved after IPA administration. Notably, iTRAQ analysis presented that IPA replenishment retained radiation-reprogrammed protein expression profile in the small intestine. Importantly, shRNA interference and hydrodynamic-based gene delivery assays further validated that pregnane X receptor (PXR)/acyl-CoA-binding protein (ACBP) signaling played pivotal roles in IPA-favored radioprotection in vitro and in vivo. CONCLUSIONS These evidences highlight that IPA is a key intestinal microbiota metabolite corroborating the therapeutic effects of FMT to radiation toxicity. Owing to the potential pitfalls of FMT, IPA might be employed as a safe and effective succedaneum to fight against accidental or iatrogenic ionizing ARS in clinical settings. Our findings also provide a novel insight into microbiome-based remedies toward radioactive diseases. Video abstract.
Collapse
Affiliation(s)
- Hui-Wen Xiao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Baidi Road, Tianjin, 300192, China
| | - Ming Cui
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Baidi Road, Tianjin, 300192, China.
| | - Yuan Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Baidi Road, Tianjin, 300192, China
| | - Jia-Li Dong
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Baidi Road, Tianjin, 300192, China
| | - Shu-Qin Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Baidi Road, Tianjin, 300192, China
| | - Chang-Chun Zhu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Baidi Road, Tianjin, 300192, China
| | - Mian Jiang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Baidi Road, Tianjin, 300192, China
| | - Tong Zhu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Baidi Road, Tianjin, 300192, China
| | - Bin Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Baidi Road, Tianjin, 300192, China
| | - Hai-Chao Wang
- Laboratory of Emergency Medicine, Feinstein Institute for Medical Research, Manhasset, NY, USA
| | - Sai-Jun Fan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Baidi Road, Tianjin, 300192, China.
| |
Collapse
|
14
|
Farhood B, Hassanzadeh G, Amini P, Shabeeb D, Musa AE, Khodamoradi E, Mohseni M, Aliasgharzadeh A, Moradi H, Najafi M. Mitigation of Radiation-induced Gastrointestinal System Injury using Resveratrol or Alpha-lipoic Acid: A Pilot Histopathological Study. Antiinflamm Antiallergy Agents Med Chem 2020; 19:413-424. [PMID: 31713500 DOI: 10.2174/1871523018666191111124028] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/05/2019] [Accepted: 10/20/2019] [Indexed: 06/10/2023]
Abstract
AIM In this study, we aimed to determine possible mitigation of radiationinduced toxicities in the duodenum, jejunum and colon using post-exposure treatment with resveratrol and alpha-lipoic acid. BACKGROUND After the bone marrow, gastrointestinal system toxicity is the second critical cause of death following whole-body exposure to radiation. Its side effects reduce the quality of life of patients who have undergone radiotherapy. Resveratrol has an antioxidant effect and stimulates DNA damage responses (DDRs). Alpha-lipoic acid neutralizes free radicals via the recycling of ascorbic acid and alpha-tocopherol. OBJECTIVE This study is a pilot investigation of the mitigation of enteritis using resveratrol and alpha-lipoic acid following histopathological study. METHODS 60 male mice were randomly assigned to six groups; control, resveratrol treatment, alpha-lipoic acid treatment, whole-body irradiation, irradiation plus resveratrol, and irradiation plus alpha-lipoic acid. The mice were irradiated with a single dose of 7 Gy from a cobalt-60 gamma-ray source. Treatment with resveratrol or alpha-lipoic acid started 24 h after irradiation and continued for 4 weeks. All mice were sacrificed after 30 days for histopathological evaluation of radiation-induced toxicities in the duodenum, jejunum and colon. RESULTS AND DISCUSSION Exposure to radiation caused mild to severe damages to vessels, goblet cells and villous. It also led to significant infiltration of macrophages and leukocytes, especially in the colon. Both resveratrol and alpha-lipoic acid were able to mitigate morphological changes. However, they could not mitigate vascular injury. CONCLUSION Resveratrol and alpha-lipoic acid could mitigate radiation-induced injuries in the small and large intestine. A comparison between these agents showed that resveratrol may be a more effective mitigator compared to alpha-lipoic acid.
Collapse
Affiliation(s)
- Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Gholamreza Hassanzadeh
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Peyman Amini
- Department of Radiology, Faculty of Paramedical, Tehran University of Medical Sciences, Tehran, Iran
| | - Dheyauldeen Shabeeb
- Department of Physiology, College of Medicine, University of Misan, Misan, Iraq
| | - Ahmed Eleojo Musa
- Department of Medical Physics, Tehran University of Medical Sciences, Tehran, Iran
| | - Ehsan Khodamoradi
- Department of Radiology and Nuclear Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mehran Mohseni
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Akbar Aliasgharzadeh
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Habiballah Moradi
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Masoud Najafi
- Department of Radiology and Nuclear Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
15
|
Lu L, Li W, Chen L, Su Q, Wang Y, Guo Z, Lu Y, Liu B, Qin S. Radiation-induced intestinal damage: latest molecular and clinical developments. Future Oncol 2019; 15:4105-4118. [PMID: 31746639 DOI: 10.2217/fon-2019-0416] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aim: To systematically review the prophylactic and therapeutic interventions for reducing the incidence or severity of intestinal symptoms among cancer patients receiving radiotherapy. Materials & methods: A literature search was conducted in the PubMed database using various search terms, including 'radiation enteritis', 'radiation enteropathy', 'radiation-induced intestinal disease', 'radiation-induced intestinal damage' and 'radiation mucositis'. The search was limited to in vivo studies, clinical trials and meta-analyses published in English with no limitation on publication date. Other relevant literature was identified based on the reference lists of selected studies. Results: The pathogenesis of acute and chronic radiation-induced intestinal damage as well as the prevention and treatment approaches were reviewed. Conclusion: There is inadequate evidence to strongly support the use of a particular strategy to reduce radiation-induced intestinal damage. More high-quality randomized controlled trials are required for interventions with limited evidence suggestive of potential benefits.
Collapse
Affiliation(s)
- Lina Lu
- School of Nuclear Science & Technology, Lanzhou University, Lanzhou 730000, Gansu, PR China.,School of Chemical Engineering, Northwest Minzu University, Lanzhou 730000, Gansu, PR China
| | - Wenjun Li
- Key Laboratory of Biology & Bioresource Utilization, Yantai Institute of Costal Zone Research, Chinese Academy of Sciences, Yantai 264003, PR China.,Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao 266071, PR China
| | - Lihua Chen
- School of Chemical Engineering, Northwest Minzu University, Lanzhou 730000, Gansu, PR China
| | - Qiong Su
- School of Chemical Engineering, Northwest Minzu University, Lanzhou 730000, Gansu, PR China
| | - Yanbin Wang
- School of Chemical Engineering, Northwest Minzu University, Lanzhou 730000, Gansu, PR China
| | - Zhong Guo
- Medical College of Northwest Minzu University, Lanzhou 730000, Gansu, PR China
| | - Yongjuan Lu
- School of Chemical Engineering, Northwest Minzu University, Lanzhou 730000, Gansu, PR China
| | - Bin Liu
- School of Nuclear Science & Technology, Lanzhou University, Lanzhou 730000, Gansu, PR China.,School of Stomatology, Lanzhou University, Lanzhou 730000, Gansu, PR China
| | - Song Qin
- Key Laboratory of Biology & Bioresource Utilization, Yantai Institute of Costal Zone Research, Chinese Academy of Sciences, Yantai 264003, PR China.,Center for Ocean Mega-Science, Chinese Academy of Sciences, Qingdao 266071, PR China
| |
Collapse
|
16
|
Jung HW, Choi JH, Jo T, Shin H, Suh JM. Systemic and Local Phenotypes of Barium Chloride Induced Skeletal Muscle Injury in Mice. Ann Geriatr Med Res 2019; 23:83-89. [PMID: 32743293 PMCID: PMC7387593 DOI: 10.4235/agmr.19.0012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/12/2019] [Accepted: 05/25/2019] [Indexed: 01/10/2023] Open
Abstract
Skeletal muscle regeneration in mice has traditionally been studied using local freeze burn or snake venom injection models. More recently, a barium chloride (BaCl2)-induced muscle injury model has been established and is gaining popularity due to the relatively simple procedure and accessibility to required reagents. Here we sought to characterize the local and systemic effects of BaCl2-induced muscle injury. For this study, a 1.2% BaCl2 solution was locally administered to the tibialis anterior (TA) muscle and local and systemic phenotypes were analyzed at different timepoints. When 50 μL of the solution was injected unilaterally in the TA muscle, no mortality was observed. However, when 100 μL of the solution was injected, 50% of the mice died within 24 h. Serum analysis of the mice injected with 50 μL of BaCl2 solution at days 1 and 7 revealed changes resembling rhabdomyolysis. At day 1 post-injection of 50 μL of the BaCl2 solution, acute suppurative inflammation was observed in gross examination of the TA muscle, while extensive hemorrhagic necrosis was revealed on histological examination. At day 7, regenerated myofibers with centralized nuclei appeared with the resolution of acute inflammatory infiltration and the muscle tissue displayed molecular signatures consistent with myofiber differentiation. The overall muscle injury and regeneration phenotypes in the BaCl2-induced muscle injury model were similar to those of the well-established freeze burn or snake venom injection models. Taken together, the BaCl2-induced muscle injury model is comparable to conventional muscle injury and regeneration models, with considerations for possible systemic effects.
Collapse
Affiliation(s)
- Hee-Won Jung
- Graduate School of Medical Science and Engineering, Korea Advanced Institute for Science and Technology, Daejeon, Korea
| | - Jin-Hyuk Choi
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute for Science and Technology, Daejeon, Korea
| | - Taehee Jo
- Graduate School of Medical Science and Engineering, Korea Advanced Institute for Science and Technology, Daejeon, Korea
| | - Hyemi Shin
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute for Science and Technology, Daejeon, Korea
| | - Jae Myoung Suh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute for Science and Technology, Daejeon, Korea
- Biomedical Science and Engineering Interdisciplinary Program, Korea Advanced Institute for Science and Technology, Daejeon, Korea
| |
Collapse
|
17
|
Tae JY, Ko Y, Park JB. Evaluation of fibroblast growth factor-2 on the proliferation of osteogenic potential and protein expression of stem cell spheroids composed of stem cells derived from bone marrow. Exp Ther Med 2019; 18:326-331. [PMID: 31258669 PMCID: PMC6566042 DOI: 10.3892/etm.2019.7543] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 04/18/2019] [Indexed: 12/11/2022] Open
Abstract
Fibroblast growth factor-2 (FGF-2) is reported to have various functions and is considered a key human mesenchymal stem cell mitogen, often supplemented to increase human mesenchymal stem cell growth rates. The purpose of this study was to evaluate the effects of FGF-2 on cellular viability and osteogenic differentiation using three-dimensional cell spheroids of stem cells. Three-dimensional cell spheroids were fabricated using concave silicon elastomer-based microwells in the presence of FGF-2 at concentrations of 0, 30, 60 and 90 ng/ml. Qualitative cellular viability was determined with a confocal microscope, and quantitative cellular viability was evaluated using a Cell Counting Kit-8 assay. Alkaline phosphatase activity and Alizarin Red S staining were used to assess osteogenic differentiation. Spheroids were well formed in silicon elastomer-based concave microwells on Day 1. The average spheroid diameters at Day 1 for FGF-2 at 0, 30, 60 and 90 ng/ml were 202.2±3.0, 206.6±22.6, 208.8±6.8 and 196.6±26.7 µm, respectively (P>0.05). The majority of the cells in the cell spheroids emitted green fluorescence. The relative Cell Counting Kit-8 assay values for FGF-2 at 0, 30, 60 and 90 ng/ml at Day 1 were 100.0±5.5, 101.8±8.8, 99.2±4.8 and 103.4±9.6% (P>0.05). The addition of FGF-2 at 60 ng/ml concentration produced the highest value for alkaline phosphatase activity. Mineralized extracellular deposits were evenly observed in each group, and the highest value was identified for FGF-2 groups at 60 ng/ml concentration for Alizarin Red S staining. Based on these findings, it was concluded that FGF-2 may increase alkaline phosphatase activity or Alizarin Red S staining, and further studies are needed to fully elucidate the mechanisms of FGF-2.
Collapse
Affiliation(s)
- Jae-Yong Tae
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Youngkyung Ko
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jun-Beom Park
- Department of Periodontics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|