1
|
Xu Y, Peng LS, Xiao CQ, Zhou Y, Wang QS, Fu H. Bisdemethoxycurcumin mitigates traumatic brain injury in rats by modulating autophagy and oxidative stress via heat shock protein 90 alpha family class A member 1-mediated nuclear translocation of transcription factor EB. Brain Res Bull 2025:111221. [PMID: 39855313 DOI: 10.1016/j.brainresbull.2025.111221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/31/2024] [Accepted: 01/19/2025] [Indexed: 01/27/2025]
Abstract
BACKGROUND Bisdemethoxycurcumin (BDMC), the primary active compound found in turmeric, exhibits diverse pharmacological properties. The study aimed to investigate the mechanisms underlying the protective effects of BDMC in traumatic brain injury (TBI). METHODS A rat TBI model was established using the Feeney's freefall epidural impact method, followed by BDMC treatment. Rat cortical neuron cells were exposed to hydrogen peroxide (H2O2) to induce oxidative stress and then treated with BDMC. The cells were also pretreated with autophagy inhibitor 3-MA and heat shock protein 90 alpha family class A member 1 (HSP90AA1) inhibitor 17-AAG. Additionally, the experiments also involved treating H2O2-exposed cortical neurons with 17-AAG and silencing HSP90AA1 expression. Co-immunoprecipitation was utilized to verify interactions between HSP90AA1 and transcription factor EB (TFEB), TFEB and nuclear factor erythroid 2 related factor 2 (Nrf2), and the localization of these complexes in the cytoplasm and nucleus. RESULTS BDMC treatment significantly reduced modified neurological severity scores, brain water content, inflammatory infiltration, oxidative stress, and apoptosis in the cerebral cortex of TBI rats. Additionally, BDMC treatment elevated the expression of Beclin 1 and light chain 3 (LC3) II/LC3 I ratio while decreasing p62 expression. It also promoted TFEB nuclear translocation and increased HSP90AA1 levels in both the cytoplasm and nucleus, along with elevated nuclear Nrf2 expressions in TBI models. In vitro experiments showed decreased malondialdehyde levels, elevated glutathione peroxidase and superoxide dismutase levels upon BDMC treatment, along with repressed cortical neurons apoptosis, elevated Beclin 1 and LC3 II/LC3 I expressions, decreased p62 expressions, reduced cytoplasmic TFEB expression, increased nuclear TFEB and Nrf2 expression, and elevated HSP90AA1 expression in the cytoplasm and nucleus. Mechanistically, BDMC mediated autophagy and oxidative stress by activating HSP90AA1/TFEB/Nrf2 axis. Finally, HSP90AA1 was shown to regulate Nrf2 expression by binding to TFEB in the cellular model. CONCLUSIONS BDMC alleviated TBI in rats by regulating autophagy and oxidative stress through HSP90AA1-mediated nuclear translocation of TFEB.
Collapse
Affiliation(s)
- Yan Xu
- The First Affiliated Hospital, Department of Critical Care Medicine, Hengyang Medical School, University of South China, No. 69 Chuanshan Road, Hengyang, 421001, Hunan Province, People's Republic of China
| | - Liang-Shan Peng
- The First Affiliated Hospital, Department of Critical Care Medicine, Hengyang Medical School, University of South China, No. 69 Chuanshan Road, Hengyang, 421001, Hunan Province, People's Republic of China
| | - Chao-Qun Xiao
- The First Affiliated Hospital, Department of Critical Care Medicine, Hengyang Medical School, University of South China, No. 69 Chuanshan Road, Hengyang, 421001, Hunan Province, People's Republic of China
| | - Yi Zhou
- The First Affiliated Hospital, Department of Critical Care Medicine, Hengyang Medical School, University of South China, No. 69 Chuanshan Road, Hengyang, 421001, Hunan Province, People's Republic of China
| | - Qiao-Sheng Wang
- The First Affiliated Hospital, Department of Critical Care Medicine, Hengyang Medical School, University of South China, No. 69 Chuanshan Road, Hengyang, 421001, Hunan Province, People's Republic of China.
| | - Hui Fu
- The First Affiliated Hospital, Department of Critical Care Medicine, Hengyang Medical School, University of South China, No. 69 Chuanshan Road, Hengyang, 421001, Hunan Province, People's Republic of China.
| |
Collapse
|
2
|
Li X, Ding Z. Cognitive dysfunction induced by cranial radiotherapy: mechanisms and therapeutic methods. Brain Res Bull 2024; 218:111106. [PMID: 39447765 DOI: 10.1016/j.brainresbull.2024.111106] [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: 05/05/2024] [Revised: 10/19/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
Cranial radiotherapy can damage normal brain tissues, inducing cognitive dysfunction in patients. Radiotherapy-induced cognitive dysfunction is associated with hippocampal injury, white matter damage and microvascular injury. In this study, the mechanisms of cognitive dysfunction induced by cranial radiotherapy and combined chemoradiotherapy are reviewed, and the advances in therapeutic methods for radiotherapy-induced brain injury are summarized. The mechanisms of radiotherapy-induced brain injury include a decline of neurogenesis, impairment of neurons and glial cells, vascular injury, oxidative stress and DNA damage, cell death, and inflammatory response. Disruption of the bloodbrain barrier (BBB) increases the exposure of the brain to chemotherapeutic agents, thus exacerbating radiotherapy-induced brain damage. The current methods used to prevent radiotherapy-induced brain injury mainly include precision radiotherapy, stem cell transplantation, and treatment with neuroprotective drugs. The combined application of precision radiotherapy and neuroprotective drugs, including antioxidants, anti-inflammatory agents and other drugs, might exert better neuroprotective effects. To resolve the issues of neuroprotective drugs, such as difficulty in crossing the BBB, nanoenzymes and drug delivery nano-systems could be applied in the future.
Collapse
Affiliation(s)
- Xuejiao Li
- Department of Radiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, China
| | - Zhongxiang Ding
- Department of Radiology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, China.
| |
Collapse
|
3
|
Li L, Liu X, Han C, Tian L, Wang Y, Han B. Ferroptosis in radiation-induced brain injury: roles and clinical implications. Biomed Eng Online 2024; 23:93. [PMID: 39261942 PMCID: PMC11389269 DOI: 10.1186/s12938-024-01288-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 08/31/2024] [Indexed: 09/13/2024] Open
Abstract
Radiation-induced brain injury (RBI) presents a significant challenge for patients undergoing radiation therapy for head, neck, and intracranial tumors. This review aims to elucidate the role of ferroptosis in RBI and its therapeutic implications. Specifically, we explore how ferroptosis can enhance the sensitivity of tumor cells to radiation while also examining strategies to mitigate radiation-induced damage to normal brain tissues. By investigating the mechanisms through which radiation increases cellular reactive oxygen species (ROS) and initiates ferroptosis, we aim to develop targeted therapeutic strategies that maximize treatment efficacy and minimize neurotoxicity. The review highlights key regulatory factors in the ferroptosis pathway, including glutathione peroxidase 4 (GPX4), cystine/glutamate antiporter system Xc- (System Xc-), nuclear factor erythroid 2-related factor 2 (NRF2), Acyl-CoA synthetase long-chain family member 4 (ACSL4), and others, and their interactions in the context of RBI. Furthermore, we discuss the clinical implications of modulating ferroptosis in radiation therapy, emphasizing the potential for selective induction of ferroptosis in tumor cells and inhibition in healthy cells. The development of advanced diagnostic tools and therapeutic strategies targeting ferroptosis offers a promising avenue for enhancing the safety and efficacy of radiation therapy, underscoring the need for further research in this burgeoning field.
Collapse
Affiliation(s)
- Lifang Li
- Department of Radiotherapy, Tianjin Medical University Baodi Hospital, Tianjin, 301800, China
| | - Xia Liu
- Department of Radiotherapy, Tianjin Medical University Baodi Hospital, Tianjin, 301800, China
| | - Chunfeng Han
- Department of Pharmacy, Tianjin Medical University Baodi Hospital, Tianjin, 301800, China
| | - Licheng Tian
- Department of Radiotherapy, Tianjin Medical University Baodi Hospital, Tianjin, 301800, China
| | - Yongzhi Wang
- Department of Radiotherapy, Tianjin Medical University Baodi Hospital, Tianjin, 301800, China
| | - Baolin Han
- Department of Radiotherapy, Tianjin Medical University Baodi Hospital, Tianjin, 301800, China.
| |
Collapse
|
4
|
Lu L, Li F, Gao Y, Kang S, Li J, Guo J. Microbiome in radiotherapy: an emerging approach to enhance treatment efficacy and reduce tissue injury. Mol Med 2024; 30:105. [PMID: 39030525 PMCID: PMC11264922 DOI: 10.1186/s10020-024-00873-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 07/08/2024] [Indexed: 07/21/2024] Open
Abstract
Radiotherapy is a widely used cancer treatment that utilizes powerful radiation to destroy cancer cells and shrink tumors. While radiation can be beneficial, it can also harm the healthy tissues surrounding the tumor. Recent research indicates that the microbiota, the collection of microorganisms in our body, may play a role in influencing the effectiveness and side effects of radiation therapy. Studies have shown that specific species of bacteria living in the stomach can influence the immune system's response to radiation, potentially increasing the effectiveness of treatment. Additionally, the microbiota may contribute to adverse effects like radiation-induced diarrhea. A potential strategy to enhance radiotherapy outcomes and capitalize on the microbiome involves using probiotics. Probiotics are living microorganisms that offer health benefits when consumed in sufficient quantities. Several studies have indicated that probiotics have the potential to alter the composition of the gut microbiota, resulting in an enhanced immune response to radiation therapy and consequently improving the efficacy of the treatment. It is important to note that radiation can disrupt the natural balance of gut bacteria, resulting in increased intestinal permeability and inflammatory conditions. These disruptions can lead to adverse effects such as diarrhea and damage to the intestinal lining. The emerging field of radiotherapy microbiome research offers a promising avenue for optimizing cancer treatment outcomes. This paper aims to provide an overview of the human microbiome and its role in augmenting radiation effectiveness while minimizing damage.
Collapse
Affiliation(s)
- Lina Lu
- School of Chemical Engineering, Northwest Minzu University, No.1, Northwest New Village, Lanzhou, Gansu, 730030, China.
- Key Laboratory of Environment-Friendly Composite Materials of the State Ethnic Affairs Commission, Lanzhou, Gansu, China.
- Gansu Provincial Biomass Function Composites Engineering Research Center, Lanzhou, Gansu, China.
- Key Laboratory for Utility of Environment-Friendly Composite Materials and Biomass in, University of Gansu Province, Lanzhou, Gansu, China.
| | - Fengxiao Li
- Department of Pharmacy, the Affiliated Hospital of Qingdao University, Qingdao, China
| | | | - Shuhe Kang
- School of Chemical Engineering, Northwest Minzu University, No.1, Northwest New Village, Lanzhou, Gansu, 730030, China
- Key Laboratory of Environment-Friendly Composite Materials of the State Ethnic Affairs Commission, Lanzhou, Gansu, China
- Gansu Provincial Biomass Function Composites Engineering Research Center, Lanzhou, Gansu, China
- Key Laboratory for Utility of Environment-Friendly Composite Materials and Biomass in, University of Gansu Province, Lanzhou, Gansu, China
| | - Jia Li
- School of Chemical Engineering, Northwest Minzu University, No.1, Northwest New Village, Lanzhou, Gansu, 730030, China
- Key Laboratory of Environment-Friendly Composite Materials of the State Ethnic Affairs Commission, Lanzhou, Gansu, China
- Gansu Provincial Biomass Function Composites Engineering Research Center, Lanzhou, Gansu, China
- Key Laboratory for Utility of Environment-Friendly Composite Materials and Biomass in, University of Gansu Province, Lanzhou, Gansu, China
| | - Jinwang Guo
- School of Chemical Engineering, Northwest Minzu University, No.1, Northwest New Village, Lanzhou, Gansu, 730030, China
- Key Laboratory of Environment-Friendly Composite Materials of the State Ethnic Affairs Commission, Lanzhou, Gansu, China
- Gansu Provincial Biomass Function Composites Engineering Research Center, Lanzhou, Gansu, China
- Key Laboratory for Utility of Environment-Friendly Composite Materials and Biomass in, University of Gansu Province, Lanzhou, Gansu, China
| |
Collapse
|
5
|
Zhong K, Liu K, Song Y, Chen S, Hu X, Xue R, Ma X, Li S, Yang J, Deng Z, Zhu X, Yuan M, Huang Y, Yin W, Chen Y, Tang Y, Shi Z. A Synthetic Steroid 5α-Androst-3β, 5, 6β-triol Alleviates Radiation-Induced Brain Injury in Mice via Inhibiting GBP5/NF-κB/NLRP3 Signal Axis. Mol Neurobiol 2024; 61:4074-4089. [PMID: 38057643 DOI: 10.1007/s12035-023-03831-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/25/2023] [Indexed: 12/08/2023]
Abstract
Radiotherapy for head and neck tumors can lead to a severe complication known as radiation-induced brain injury (RIBI). However, the underlying mechanism of RIBI development remains unclear, and limited prevention and treatment options are available. Neuroactive steroids have shown potential in treating neurological disorders. 5α-Androst-3β, 5, 6β-triol (TRIOL), a synthetic neuroprotective steroid, holds promise as a treatment candidate for RIBI patients. However, the neuroprotective effects and underlying mechanism of TRIOL on RIBI treatment are yet to be elucidated. In the present study, our findings demonstrate TRIOL's potential as a neuroprotective agent against RIBI. In gamma knife irradiation mouse model, TRIOL treatment significantly reduced brain necrosis volume, microglial activation, and neuronal loss. RNA-sequencing, immunofluorescence, real-time quantitative polymerase chain reaction, siRNA transfection, and western blotting techniques revealed that TRIOL effectively decreased microglial activation, proinflammatory cytokine release, neuron loss, and guanylate-binding protein 5 (GBP5) expression, along with its downstream signaling pathways NF-κB and NLRP3 activation in vitro. In summary, TRIOL effectively alleviate RIBI by inhibiting the GBP5/NF-κB/NLRP3 signal axis, reducing microglia activation and pro-inflammation cytokines release, rescuing neuron loss. This study highlights the potential of TRIOL as a novel and promising therapy drug for RIBI treatment.
Collapse
Affiliation(s)
- Ke Zhong
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Department of Pharmacy, Sun-Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Kejia Liu
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yu Song
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Sitai Chen
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Xia Hu
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Ruiqi Xue
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Xueying Ma
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Shaojian Li
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Jingwen Yang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Zhenhong Deng
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Xiaoqiu Zhu
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Mingjun Yuan
- Guangzhou Cellprotek Pharmaceutical Co., Ltd., H Building F/1, 3 Juquan Road, Science City, Guangzhou, 510670, China
| | - Yijun Huang
- Guangzhou Cellprotek Pharmaceutical Co., Ltd., H Building F/1, 3 Juquan Road, Science City, Guangzhou, 510670, China
| | - Wei Yin
- Guangzhou Cellprotek Pharmaceutical Co., Ltd., H Building F/1, 3 Juquan Road, Science City, Guangzhou, 510670, China
| | - Yupin Chen
- Guangzhou Cellprotek Pharmaceutical Co., Ltd., H Building F/1, 3 Juquan Road, Science City, Guangzhou, 510670, China.
| | - Yamei Tang
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Brain Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
- Guangdong Province Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.
| | - Zhongshan Shi
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| |
Collapse
|
6
|
Xu K, Liu Z, Pan S, Zhang N, Wu S, Yang G, Song X, Teng Y, Tong X. BMSCs attenuate radiation-induced brain injury induced hippocampal neuronal apoptosis through a PI3K/Akt/Bax/Bcl-2 signaling pathway. Brain Res 2024; 1829:148795. [PMID: 38331376 DOI: 10.1016/j.brainres.2024.148795] [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: 10/07/2023] [Revised: 01/26/2024] [Accepted: 02/01/2024] [Indexed: 02/10/2024]
Abstract
BACKGROUND Bone marrow mesenchymal stem cell (BMSCs) -based therapies represent a promising treatment for neurological disorders. However, therapeutic effects and mechanisms of BMSCs transplantation for radiation-induced brain injury (RIBI) have not been fully disclosed. In this article, we explored the functions of BMSCs transplantation on RIBI and investigated the protective effects of BMSCS on hippocampal neurons in RIBI as well as the related molecular mechanisms. MATERIALS AND METHODS 6-8 weeks-old rats were used to build a RIBI model. Rats in BMSC group were treated with a 3 × 106 BMSCs injection through the tail vein on the 1st day and 8th day after irradiation; rats in both control and RIBI groups were injected with an equivalent volume of physiological saline for comparisons. The Morris water maze was applied to detect the variations in cognitive function after RIBI. MRS was performed to test changes in NAA/Cr, indicating neuronal apoptosis after RIBI. TUNEL was conducted to detect apoptosis of rat hippocampal neurons, and HE staining was carried out to show pathological variations in the hippocampal region of rats. Protein levels of PI3K, P-PI3K, AKT, P-AKT, Bcl-2, and Bax proteins of rats in the hippocampal area were all determined by Western blot. RESULTS Cognitive function was reduced and hippocampal neurons underwent apoptosis in the rats of the RIBI group, and cognitive abilities, histopathological alterations, and apoptosis of hippocampal neurons were significantly improved after BMSCs treatment; the expression of PI3K, P-PI3K, AKT, P-AKT, and Bcl-2 proteins, in the hippocampal region of the rat, was up-regulated, and Bax proteins were down-regulated. CONCLUSIONS BMCSs can inhibit hippocampal neuronal apoptosis in RIBI, and the mechanism may be associated with the up-regulation of Bcl-2 and down-regulation of Bax by the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Kaina Xu
- College of Medical Technology, Qiqihar Medical University, No.333 Bukui Street, Jianhua District, Qiqihar, Heilongjiang 161006, China
| | - Zhanhong Liu
- College of Medical Technology, Qiqihar Medical University, No.333 Bukui Street, Jianhua District, Qiqihar, Heilongjiang 161006, China
| | - Shichao Pan
- College of Medical Technology, Qiqihar Medical University, No.333 Bukui Street, Jianhua District, Qiqihar, Heilongjiang 161006, China
| | - Na Zhang
- College of Medical Technology, Qiqihar Medical University, No.333 Bukui Street, Jianhua District, Qiqihar, Heilongjiang 161006, China
| | - Shuang Wu
- Department of Radiotherapy, The Third Affiliated Hospital of Qiqihar Medical University, No.27, Taishun Street, Tiefeng District, Qiqihar, Heilongjiang 161006, China
| | - Guangrun Yang
- Department of Radiotherapy, The Third Affiliated Hospital of Qiqihar Medical University, No.27, Taishun Street, Tiefeng District, Qiqihar, Heilongjiang 161006, China
| | - Xue Song
- Department of Radiotherapy, The Third Affiliated Hospital of Qiqihar Medical University, No.27, Taishun Street, Tiefeng District, Qiqihar, Heilongjiang 161006, China
| | - Ye Teng
- Department of Radiotherapy, The Third Affiliated Hospital of Qiqihar Medical University, No.27, Taishun Street, Tiefeng District, Qiqihar, Heilongjiang 161006, China
| | - Xu Tong
- Department of Radiotherapy, The Third Affiliated Hospital of Qiqihar Medical University, No.27, Taishun Street, Tiefeng District, Qiqihar, Heilongjiang 161006, China.
| |
Collapse
|
7
|
Zhang Y, Ren E, Zhang C, Wang Y, Chen X, Li L. The protective role of oily fish intake against type 2 diabetes: insights from a genetic correlation and Mendelian randomization study. Front Nutr 2024; 11:1288886. [PMID: 38567249 PMCID: PMC10986736 DOI: 10.3389/fnut.2024.1288886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 03/11/2024] [Indexed: 04/04/2024] Open
Abstract
Background and aims Previous research has underscored the association between oily fish intake and type 2 diabetes (T2DM), yet the causality remains elusive. Methods A bidirectional univariable Mendelian Randomization (MR) analysis was employed to evaluate the causal effects of oily fish and non-oily fish intake on T2DM. Replication analysis and meta-analysis were conducted to ensure robust results. Multivariable MR analysis was utilized to assess confounders, and further mediation MR analysis discerned mediating effects. Linkage Disequilibrium Score (LDSC) analysis was undertaken to compute genetic correlations. Inverse variance weighted (IVW) was the primary method, complemented by a series of sensitivity analyses. Results The LDSC analysis unveiled a significant genetic correlation between oily fish intake and T2DM (Genetic correlation: -0.102, p = 4.43 × 10-4). For each standard deviation (SD) increase in genetically predicted oily fish intake, the risk of T2DM was reduced by 38.6% (OR = 0.614, 95% CI 0.504 ~ 0.748, p = 1.24 × 10-6, False Discovery Rate (FDR) = 3.72 × 10-6). The meta-analysis across three data sources highlighted a persistent causal association (OR = 0.728, 95% CI 0.593 ~ 0.895, p = 0.003). No other causal effects were identified (all p > 0.5, FDR > 0.5). The main outcomes remained consistent in most sensitivity analyses. Both MVMR and mediation MR analyses emphasized the mediating roles of triglycerides (TG), body mass index (BMI), and 25-hydroxyvitamin D (25OHD) levels. Conclusion To encapsulate, there's an inverse association between oily fish intake and T2DM risk, suggesting potential benefits of oily fish intake in T2DM prevention.
Collapse
Affiliation(s)
- Youqian Zhang
- Department of Endocrinology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
- Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Entong Ren
- Health Science Center, Yangtze University, Jingzhou, Hubei, China
- Southern Theater General Hospital, Guangzhou, Guangdong, China
| | - Chunlong Zhang
- Health Science Center, Yangtze University, Jingzhou, Hubei, China
- Department of Nursing, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yang Wang
- Department of Neurology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaohe Chen
- Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Lin Li
- Department of Endocrinology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
| |
Collapse
|
8
|
Yin G, Wang Q, Lv T, Liu Y, Peng X, Zeng X, Huang J. The Radioprotective Effect of LBP on Neurogenesis and Cognition after Acute Radiation Exposure. Curr Radiopharm 2024; 17:257-265. [PMID: 38204264 PMCID: PMC11327742 DOI: 10.2174/0118744710274008231220055033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 11/06/2023] [Accepted: 11/14/2023] [Indexed: 01/12/2024]
Abstract
BACKGROUND Radiation exposure has been linked to the development of brain damage and cognitive impairment, but the protective effect and mechanism of Lycium barbarum pills (LBP) on radiation-induced neurological damage remains to be clarified. METHODS Behavioral tests and immunohistochemical studies were conducted to evaluate the protective effects of LBP extract (10 g/kg orally daily for 4 weeks) against radiation-induced damage on neurogenesis and cognitive function in Balb/c mice exposed to 5.5 Gy X-ray acute radiation. RESULTS The results showed that the LBP extract significantly improved body weight loss, locomotor activity and spatial learning and memory. Immunohistochemical tests revealed that the LBP extract prevented the loss of proliferating cells, newly generated neurons and interneurons, especially in the subgranular area of the dentate gyrus. CONCLUSION The findings suggest that LBP is a potential neuroprotective drug for mitigating radiation-induced neuropsychological disorders.
Collapse
Affiliation(s)
- Gang Yin
- Department of Neurology, Jingzhou Hospital Affiliated to Yangtze University, Jingzhou, Hubei, China
| | - Qinqi Wang
- Department of Internal Medicine, Wuhan No.1 Hospital, Wuhan, Hubei, China
| | - Tongtong Lv
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Yifan Liu
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Xiaochun Peng
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Xianqin Zeng
- Department of Gynaecology and Obstetrics, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jiangrong Huang
- Department of Integrative Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei, China
| |
Collapse
|
9
|
Long H, Zhao L, Xiao ZS, Li SX, Huang QL, Xiao S, Wu LL. Impact of bariatric surgery on glucose and lipid metabolism and liver and kidney function in food-induced obese diabetic rats. World J Diabetes 2023; 14:1249-1258. [PMID: 37664479 PMCID: PMC10473948 DOI: 10.4239/wjd.v14.i8.1249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/30/2023] [Accepted: 07/17/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND Obesity usually causes diabetes mellitus (DM) and is a serious danger to human health. Type 2 DM (T2DM) mostly occurs along with obesity. Foodborne obesity-induced DM is caused by an excessive long-term diet and surplus energy. Bariatric surgery can improve the symptoms of T2DM in some obese patients. But different types of bariatric surgery may have different effects. AIM To investigate the effect of bariatric surgery on glucose and lipid metabolism and liver and kidney function in rats. METHODS Male Sprague-Dawley rats aged 6-8 wk underwent Roux-en-Y gastric bypass surgery (RYGB), sleeve gastrectomy (SG), or gastric banding (GB). Glucose and insulin tolerance tests, analyses of biochemical parameters, histological examination, western blot, and quantitative real-time polymerase chain reaction were conducted. RESULTS In comparison to the sham operation group, the RYGB, SG, and GB groups had decreased body weight and food intake, reduced glucose intolerance and insulin insensitivity, downregulated biochemical parameters, alleviated morphological changes in the liver and kidneys, and decreased levels of protein kinase C β/ P66shc. The effect in the RYGB group was better than that in the SG and GB groups. CONCLUSION These results suggest that RYGB, SG and GB may be helpful for the treatment of foodborne obesity-induced DM.
Collapse
Affiliation(s)
- Hong Long
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan Province, China
| | - Lei Zhao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan Province, China
| | - Zhong-Sheng Xiao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan Province, China
| | - Shu-Xiang Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan Province, China
| | - Qiu-Lin Huang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan Province, China
| | - Shuai Xiao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan Province, China
| | - Liang-Liang Wu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, Hunan Province, China
| |
Collapse
|