Background: In Parkinson's disease (PD) brains, microglia are activated to release inflammatory factors to induce the production of reactive oxygen species (ROS) in neuron, and vice versa. Moreover, neuroinflammation and its synergistic interaction with oxidative stress contribute to the pathogenesis of PD.
Methods: In this study, we investigated whether in-house synthetic coumarin-chalcone derivatives protect human microglia HMC3 and neuroblastoma BE(2)-M17 cells against 1-methyl-4-phenyl pyridinium (MPP+)-induced neuroinflammation and associated neuronal damage.
Results: Treatment with MPP+ decreased cell viability as well as increased the release of inflammatory mediators including cytokines and nitric oxide in culture medium, and enhanced expression of microglial activation markers CD68 and MHCII in HMC3 cells. The protein levels of NLRP3, CASP1, iNOS, IL-1β, IL-6, and TNF-α were also increased in MPP+-stimulated HMC3 cells. Among the four tested compounds, LM-016, LM-021, and LM-036 at 10 μM counteracted the inflammatory action of MPP+ in HMC3 cells. In addition, LM-021 and LM-036 increased cell viability, reduced lactate dehydrogenase release, ameliorated cellular ROS production, decreased caspase-1, caspase-3 and caspase-6 activities, and promoted neurite outgrowth in MPP+-treated BE(2)-M17 cells. These protective effects were mediated by down-regulating inflammatory NLRP1, IL-1β, IL-6, and TNF-α, as well as up-regulating antioxidative NRF2, NQO1, GCLC, and PGC-1α, and neuroprotective CREB, BDNF, and BCL2.
Conclusion: The study results strengthen the involvement of neuroinflammation and oxidative stress in PD pathogenic mechanisms, and indicate the potential use of LM-021 and LM-036 as dual inflammasome inhibitors in treating both NLRP1- and NLRP3-associated PD.
{"title":"Coumarin-chalcone derivatives as dual NLRP1 and NLRP3 inflammasome inhibitors targeting oxidative stress and inflammation in neurotoxin-induced HMC3 and BE(2)-M17 cell models of Parkinson's disease.","authors":"Te-Hsien Lin, Ya-Jen Chiu, Chih-Hsin Lin, Yi-Ru Chen, Wenwei Lin, Yih-Ru Wu, Kuo-Hsuan Chang, Chiung-Mei Chen, Guey-Jen Lee-Chen","doi":"10.3389/fnagi.2024.1437138","DOIUrl":"https://doi.org/10.3389/fnagi.2024.1437138","url":null,"abstract":"<p><strong>Background: </strong>In Parkinson's disease (PD) brains, microglia are activated to release inflammatory factors to induce the production of reactive oxygen species (ROS) in neuron, and vice versa. Moreover, neuroinflammation and its synergistic interaction with oxidative stress contribute to the pathogenesis of PD.</p><p><strong>Methods: </strong>In this study, we investigated whether in-house synthetic coumarin-chalcone derivatives protect human microglia HMC3 and neuroblastoma BE(2)-M17 cells against 1-methyl-4-phenyl pyridinium (MPP<sup>+</sup>)-induced neuroinflammation and associated neuronal damage.</p><p><strong>Results: </strong>Treatment with MPP<sup>+</sup> decreased cell viability as well as increased the release of inflammatory mediators including cytokines and nitric oxide in culture medium, and enhanced expression of microglial activation markers CD68 and MHCII in HMC3 cells. The protein levels of NLRP3, CASP1, iNOS, IL-1β, IL-6, and TNF-α were also increased in MPP<sup>+</sup>-stimulated HMC3 cells. Among the four tested compounds, LM-016, LM-021, and LM-036 at 10 μM counteracted the inflammatory action of MPP<sup>+</sup> in HMC3 cells. In addition, LM-021 and LM-036 increased cell viability, reduced lactate dehydrogenase release, ameliorated cellular ROS production, decreased caspase-1, caspase-3 and caspase-6 activities, and promoted neurite outgrowth in MPP<sup>+</sup>-treated BE(2)-M17 cells. These protective effects were mediated by down-regulating inflammatory NLRP1, IL-1β, IL-6, and TNF-α, as well as up-regulating antioxidative NRF2, NQO1, GCLC, and PGC-1α, and neuroprotective CREB, BDNF, and BCL2.</p><p><strong>Conclusion: </strong>The study results strengthen the involvement of neuroinflammation and oxidative stress in PD pathogenic mechanisms, and indicate the potential use of LM-021 and LM-036 as dual inflammasome inhibitors in treating both NLRP1- and NLRP3-associated PD.</p>","PeriodicalId":12450,"journal":{"name":"Frontiers in Aging Neuroscience","volume":"16 ","pages":"1437138"},"PeriodicalIF":4.1,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11473416/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142462037","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-30eCollection Date: 2024-01-01DOI: 10.3389/fnagi.2024.1480502
Tian Wang, Guangwei Sun, Bingdong Tao
Postoperative cognitive dysfunction (POCD) poses a significant threat to patients undergoing anesthesia and surgery, particularly elderly patients. It is characterized by diminished cognitive functions post surgery, such as impaired memory and decreased concentration. The potential risk factors for POCD include age, surgical trauma, anesthetic type, and overall health condition; however, the precise mechanisms underlying POCD remain elusive. Recent studies suggest that neuroinflammation might be a primary pathogenic factor. NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasomes are implicated in exacerbating POCD by promoting the release of inflammatory factors and proteins that initiate pyroptosis, further influencing the disease process. The regulation of NLRP3 inflammasome activity, including its activation and degradation, is tightly controlled through multiple pathways and mechanisms. In addition, autophagy, a protective mechanism, regulates the NLRP3 inflammasome to control the progression of POCD. This review reviews recent findings on the role of the NLRP3 inflammasome in POCD pathogenesis and discusses therapeutic strategies aimed at reducing NLRP3 sources, inhibiting cellular pyroptosis, and enhancing autophagy.
{"title":"Updated insights into the NLRP3 inflammasome in postoperative cognitive dysfunction: emerging mechanisms and treatments.","authors":"Tian Wang, Guangwei Sun, Bingdong Tao","doi":"10.3389/fnagi.2024.1480502","DOIUrl":"https://doi.org/10.3389/fnagi.2024.1480502","url":null,"abstract":"<p><p>Postoperative cognitive dysfunction (POCD) poses a significant threat to patients undergoing anesthesia and surgery, particularly elderly patients. It is characterized by diminished cognitive functions post surgery, such as impaired memory and decreased concentration. The potential risk factors for POCD include age, surgical trauma, anesthetic type, and overall health condition; however, the precise mechanisms underlying POCD remain elusive. Recent studies suggest that neuroinflammation might be a primary pathogenic factor. NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3) inflammasomes are implicated in exacerbating POCD by promoting the release of inflammatory factors and proteins that initiate pyroptosis, further influencing the disease process. The regulation of NLRP3 inflammasome activity, including its activation and degradation, is tightly controlled through multiple pathways and mechanisms. In addition, autophagy, a protective mechanism, regulates the NLRP3 inflammasome to control the progression of POCD. This review reviews recent findings on the role of the NLRP3 inflammasome in POCD pathogenesis and discusses therapeutic strategies aimed at reducing NLRP3 sources, inhibiting cellular pyroptosis, and enhancing autophagy.</p>","PeriodicalId":12450,"journal":{"name":"Frontiers in Aging Neuroscience","volume":"16 ","pages":"1480502"},"PeriodicalIF":4.1,"publicationDate":"2024-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11474915/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142462054","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Alzheimer's disease (AD) is a debilitating progressively neurodegenerative disease. The best-characterized hallmark of AD, which is marked by behavioral alterations and cognitive deficits, is the aggregation of deposition of amyloid-beta (Aβ) and hyper-phosphorylated microtubule-associated protein Tau. Despite decades of experimental progress, the control rate of AD remains poor, and more precise deciphering is needed for potential therapeutic targets and signaling pathways involved. In recent years, phosphoinositide 3-kinase (PI3K) and Akt have been recognized for their role in the neuroprotective effect of various agents, and glycogen synthase kinase 3 (GSK3), a downstream enzyme, is also crucial in the tau phosphorylation and Aβ deposition. An overview of the function of PI3K/Akt pathway in the pathophysiology of AD is provided in this review, along with a discussion of recent developments in the pharmaceuticals and herbal remedies that target the PI3K/Akt signaling pathway. In conclusion, despite the challenges and hurdles, cumulative findings of novel targets and agents in the PI3K/Akt signaling axis are expected to hold promise for advancing AD prevention and treatment.
阿尔茨海默病(AD)是一种使人衰弱的进行性神经退行性疾病。淀粉样蛋白-β(Aβ)和高磷酸化微管相关蛋白 Tau 的聚集沉积是阿兹海默病最显著的特征,表现为行为改变和认知障碍。尽管经过数十年的实验进展,AD 的控制率仍然很低,需要对潜在的治疗靶点和相关信号通路进行更精确的解读。近年来,磷酸肌酸 3- 激酶(PI3K)和 Akt 因其在各种药物的神经保护作用中的作用而得到认可,而糖原合酶激酶 3(GSK3)作为一种下游酶,在 tau 磷酸化和 Aβ 沉积中也起着至关重要的作用。本综述概述了PI3K/Akt通路在AD病理生理学中的功能,并讨论了针对PI3K/Akt信号通路的药物和草药疗法的最新进展。总之,尽管存在挑战和障碍,PI3K/Akt 信号转导轴上的新靶点和药物的累积研究结果有望推动艾滋病的预防和治疗。
{"title":"The role of PI3K signaling pathway in Alzheimer's disease.","authors":"Jingying Pan, Qi Yao, Yankai Wang, Suyan Chang, Chenlong Li, Yongjiang Wu, Jianhong Shen, Riyun Yang","doi":"10.3389/fnagi.2024.1459025","DOIUrl":"https://doi.org/10.3389/fnagi.2024.1459025","url":null,"abstract":"<p><p>Alzheimer's disease (AD) is a debilitating progressively neurodegenerative disease. The best-characterized hallmark of AD, which is marked by behavioral alterations and cognitive deficits, is the aggregation of deposition of amyloid-beta (Aβ) and hyper-phosphorylated microtubule-associated protein Tau. Despite decades of experimental progress, the control rate of AD remains poor, and more precise deciphering is needed for potential therapeutic targets and signaling pathways involved. In recent years, phosphoinositide 3-kinase (PI3K) and Akt have been recognized for their role in the neuroprotective effect of various agents, and glycogen synthase kinase 3 (GSK3), a downstream enzyme, is also crucial in the tau phosphorylation and Aβ deposition. An overview of the function of PI3K/Akt pathway in the pathophysiology of AD is provided in this review, along with a discussion of recent developments in the pharmaceuticals and herbal remedies that target the PI3K/Akt signaling pathway. In conclusion, despite the challenges and hurdles, cumulative findings of novel targets and agents in the PI3K/Akt signaling axis are expected to hold promise for advancing AD prevention and treatment.</p>","PeriodicalId":12450,"journal":{"name":"Frontiers in Aging Neuroscience","volume":"16 ","pages":"1459025"},"PeriodicalIF":4.1,"publicationDate":"2024-09-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11466886/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142462053","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background and aims: This study aimed to investigate the effect of iron overload on acetylcholinesterase activity in the frontal lobe tissue of rats with minimal hepatic encephalopathy (MHE) and its relation to cognitive ability. By elucidating the potential mechanisms of cognitive impairment, this study may offer insights into novel therapeutic targets for MHE.
Materials and methods: Twelve Sprague-Dawley rats were purchased and randomly assigned to either the experimental or control group with six rats in each group. Following the induction of MHE, the Morris Water Maze (MWM) was utilized to assess spatial orientation and memory capacity. Subsequently, Magnetic Resonance Imaging (MRI) scans were performed to capture Quantitative Susceptibility Mapping (QSM) images of all rats' heads.
Results: Compared to the control group rats, the MHE model rats showed significantly reduced learning and memory capabilities as well as spatial orientation abilities (P < 0.05). Furthermore, the susceptibility values in the frontal lobe tissue of MHE model rats was significantly higher than that of the control group rats (P < 0.05), and the corresponding BuChE activity in the frontal lobe extract of model rats was significantly increased while BuChE activity in the peripheral blood serum was significantly decreased compared to the control group rats (P < 0.05). Meanwhile, our findings indicate a significant positive correlation between latency period and BuChE activity with susceptibility values in the MHE group.
Conclusion: The changes in BuChE activity in frontal lobe extract may be related to changes in spatial orientation and behavioral changes in MHE, and iron overload in the frontal lobe tissue may regulate changes in BuChE activity, BuChE levels appear to be iron-dependent.
{"title":"Iron overload regulates cognitive function in rats with minimal hepatic encephalopathy by inducing an increase in frontal butyrylcholinesterase activity.","authors":"Hua Lan, Xuhong Yang, Minxing Wang, Minglei Wang, Xueying Huang, Xiaodong Wang","doi":"10.3389/fnagi.2024.1447965","DOIUrl":"https://doi.org/10.3389/fnagi.2024.1447965","url":null,"abstract":"<p><strong>Background and aims: </strong>This study aimed to investigate the effect of iron overload on acetylcholinesterase activity in the frontal lobe tissue of rats with minimal hepatic encephalopathy (MHE) and its relation to cognitive ability. By elucidating the potential mechanisms of cognitive impairment, this study may offer insights into novel therapeutic targets for MHE.</p><p><strong>Materials and methods: </strong>Twelve Sprague-Dawley rats were purchased and randomly assigned to either the experimental or control group with six rats in each group. Following the induction of MHE, the Morris Water Maze (MWM) was utilized to assess spatial orientation and memory capacity. Subsequently, Magnetic Resonance Imaging (MRI) scans were performed to capture Quantitative Susceptibility Mapping (QSM) images of all rats' heads.</p><p><strong>Results: </strong>Compared to the control group rats, the MHE model rats showed significantly reduced learning and memory capabilities as well as spatial orientation abilities (<i>P</i> < 0.05). Furthermore, the susceptibility values in the frontal lobe tissue of MHE model rats was significantly higher than that of the control group rats (<i>P</i> < 0.05), and the corresponding BuChE activity in the frontal lobe extract of model rats was significantly increased while BuChE activity in the peripheral blood serum was significantly decreased compared to the control group rats (<i>P</i> < 0.05). Meanwhile, our findings indicate a significant positive correlation between latency period and BuChE activity with susceptibility values in the MHE group.</p><p><strong>Conclusion: </strong>The changes in BuChE activity in frontal lobe extract may be related to changes in spatial orientation and behavioral changes in MHE, and iron overload in the frontal lobe tissue may regulate changes in BuChE activity, BuChE levels appear to be iron-dependent.</p>","PeriodicalId":12450,"journal":{"name":"Frontiers in Aging Neuroscience","volume":"16 ","pages":"1447965"},"PeriodicalIF":4.1,"publicationDate":"2024-09-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11466795/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142462039","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Cognitive impairment (CI) is common in Parkinson's disease (PD). Multiple brain regions and their interactions are involved in PD associated CI. Magnetic resonance imaging (MRI) technology is a non-invasive method in investigating brain structure and inter-regional connections. In this study, by comparing cortical thickness, subcortical volume, and brain network topology properties in PD patients with and without CI, we aimed to understand the changes of brain structure and structural covariance network properties in PD associated CI.
Methods: A total of 18 PD patients with CI and 33 PD patients without CI were recruited. Movement Disorder Society Unified Parkinson's Disease Rating Scale, Hoehn and Yahr stage, Mini Mental State Examination Scale, Non-motor Symptom Rating Scale, Hamilton Anxiety Scale, and Hamilton Depression Scale were assessed. All participants underwent structural 3T MRI. Cortical thickness, subcortical volume, global and nodal network topology properties were measured.
Results: Compared with PD patients without CI, the volumes of white matter, thalamus and hippocampus were lower in PD patients with CI. And decreased whole-brain local efficiency is associated with CI in PD patients. While the cortical thickness and nodal network topology properties were comparable between PD patients with and without CI.
Conclusion: Our findings support the alterations of brain structure and disruption of structural covariance network are involved in PD associated CI, providing a new insight into the association between graph properties and PD associated CI.
{"title":"Changes of brain structure and structural covariance networks in Parkinson's disease associated cognitive impairment.","authors":"Rong-Pei Liu, Guo-Liang Lin, Lu-Lu Ma, Shi-Shi Huang, Cheng-Xiang Yuan, Shi-Guo Zhu, Mei-Ling Sheng, Ming Zou, Jian-Hong Zhu, Xiong Zhang, Jian-Yong Wang","doi":"10.3389/fnagi.2024.1449276","DOIUrl":"10.3389/fnagi.2024.1449276","url":null,"abstract":"<p><strong>Background: </strong>Cognitive impairment (CI) is common in Parkinson's disease (PD). Multiple brain regions and their interactions are involved in PD associated CI. Magnetic resonance imaging (MRI) technology is a non-invasive method in investigating brain structure and inter-regional connections. In this study, by comparing cortical thickness, subcortical volume, and brain network topology properties in PD patients with and without CI, we aimed to understand the changes of brain structure and structural covariance network properties in PD associated CI.</p><p><strong>Methods: </strong>A total of 18 PD patients with CI and 33 PD patients without CI were recruited. Movement Disorder Society Unified Parkinson's Disease Rating Scale, Hoehn and Yahr stage, Mini Mental State Examination Scale, Non-motor Symptom Rating Scale, Hamilton Anxiety Scale, and Hamilton Depression Scale were assessed. All participants underwent structural 3T MRI. Cortical thickness, subcortical volume, global and nodal network topology properties were measured.</p><p><strong>Results: </strong>Compared with PD patients without CI, the volumes of white matter, thalamus and hippocampus were lower in PD patients with CI. And decreased whole-brain local efficiency is associated with CI in PD patients. While the cortical thickness and nodal network topology properties were comparable between PD patients with and without CI.</p><p><strong>Conclusion: </strong>Our findings support the alterations of brain structure and disruption of structural covariance network are involved in PD associated CI, providing a new insight into the association between graph properties and PD associated CI.</p>","PeriodicalId":12450,"journal":{"name":"Frontiers in Aging Neuroscience","volume":"16 ","pages":"1449276"},"PeriodicalIF":4.1,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11464354/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142399917","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Delirium is an acute, global cognitive disorder syndrome, also known as acute brain syndrome, characterized by disturbance of attention and awareness and fluctuation of symptoms. Its incidence is high among critically ill patients. Once patients develop delirium, it increases the risk of unplanned extubation, prolongs hospital stay, increases the risk of nosocomial infection, post-intensive care syndrome-cognitive impairment, and even death. Therefore, it is of great importance to understand how delirium occurs and to reduce the incidence of delirium in critically ill patients. This paper reviews the potential pathophysiological mechanisms of delirium in critically ill patients, with the aim of better understanding its pathophysiological processes, guiding the formulation of effective prevention and treatment strategies, providing a basis for clinical medication.
{"title":"Mechanisms underlying delirium in patients with critical illness.","authors":"Ying-Ying Fan, Ruo-Yu Luo, Meng-Tian Wang, Chao-Yun Yuan, Yuan-Yuan Sun, Ji-Yong Jing","doi":"10.3389/fnagi.2024.1446523","DOIUrl":"10.3389/fnagi.2024.1446523","url":null,"abstract":"<p><p>Delirium is an acute, global cognitive disorder syndrome, also known as acute brain syndrome, characterized by disturbance of attention and awareness and fluctuation of symptoms. Its incidence is high among critically ill patients. Once patients develop delirium, it increases the risk of unplanned extubation, prolongs hospital stay, increases the risk of nosocomial infection, post-intensive care syndrome-cognitive impairment, and even death. Therefore, it is of great importance to understand how delirium occurs and to reduce the incidence of delirium in critically ill patients. This paper reviews the potential pathophysiological mechanisms of delirium in critically ill patients, with the aim of better understanding its pathophysiological processes, guiding the formulation of effective prevention and treatment strategies, providing a basis for clinical medication.</p>","PeriodicalId":12450,"journal":{"name":"Frontiers in Aging Neuroscience","volume":"16 ","pages":"1446523"},"PeriodicalIF":4.1,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11464339/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142399918","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-25eCollection Date: 2024-01-01DOI: 10.3389/fnagi.2024.1451766
Junxin Liu, Jiahui Jiang, Chuantong He, Longjian Zhou, Yi Zhang, Shuai Zhao, Zhiyou Yang
Introduction: Alzheimer's disease (AD) is the leading cause of dementia, and currently, no effective treatments are available to reverse or halt its progression in clinical practice. Although a plethora of studies have highlighted the benefits of physical exercise in combating AD, elder individuals often have limited exercise capacity. Therefore, mild physical exercise and nutritional interventions represent potential strategies for preventing and mitigating neurodegenerative diseases. Our research, along with other studies, have demonstrated that platycodin D (PD) or its metabolite, platycodigenin, derived from the medicinal plant Platycodon grandiflorus, exerts neuroprotective effects against amyloid β (Aβ)-induced neuroinflammation. However, the combined effects of PD and physical exercise on alleviating AD have yet to be explored. The current study aimed to investigate whether combined therapy could synergistically ameliorate memory deficits and AD pathology in 5 × FAD mice.
Methods: Five-month-old 5 × FAD mice were randomly assigned to four groups, and received either PD (5 mg/kg/day, p.o.), voluntary running, or a combination of both for 47 days. Nest building test, locomotion test, and Morris water maze test were used to evaluate the cognitive function. Immunohistochemical and ELISA analysis was performed to determine Aβ build-up, microglia and astrocytes hyperactivation, and survival neurons in the hippocampus and perirhinal cortex. Real-time quantitative PCR analysis was used to assess the polarization of microglia and astrocytes. HPLC analysis was performed to measure monoamine neurotransmitters in the hippocampus.
Results and discussion: The combination of PD and voluntary running synergistically restored nest-building behavior, alleviated recognition and spatial memory deficits, and showed superior effects compared to monotherapy. In addition, the PD and voluntary running combination reduced Aβ build-up, decreased hyperactivation of microglia and astrocytes in the hippocampus and perirhinal cortex, promoted the polarization of inflammatory M1 microglia and reactive astrocytes toward beneficial phenotypes, and lowered systemic circulating pro-inflammatory cytokines while increasing anti-inflammatory cytokines in 5 × FAD mice. Furthermore, combined therapy effectively protected neurons and increased levels of 5-hydroxytryptamine (5-HT) and dopamine (DA) in the hippocampus of 5 × FAD mice. In conclusion, the combination of PD and voluntary running holds great potential as a treatment for AD, offering promise for delaying onset or progression of AD.
{"title":"Platycodin D and voluntary running synergistically ameliorate memory deficits in 5 × FAD mice via mediating neuromodulation and neuroinflammation.","authors":"Junxin Liu, Jiahui Jiang, Chuantong He, Longjian Zhou, Yi Zhang, Shuai Zhao, Zhiyou Yang","doi":"10.3389/fnagi.2024.1451766","DOIUrl":"10.3389/fnagi.2024.1451766","url":null,"abstract":"<p><strong>Introduction: </strong>Alzheimer's disease (AD) is the leading cause of dementia, and currently, no effective treatments are available to reverse or halt its progression in clinical practice. Although a plethora of studies have highlighted the benefits of physical exercise in combating AD, elder individuals often have limited exercise capacity. Therefore, mild physical exercise and nutritional interventions represent potential strategies for preventing and mitigating neurodegenerative diseases. Our research, along with other studies, have demonstrated that platycodin D (PD) or its metabolite, platycodigenin, derived from the medicinal plant <i>Platycodon grandiflorus</i>, exerts neuroprotective effects against amyloid β (Aβ)-induced neuroinflammation. However, the combined effects of PD and physical exercise on alleviating AD have yet to be explored. The current study aimed to investigate whether combined therapy could synergistically ameliorate memory deficits and AD pathology in 5 × FAD mice.</p><p><strong>Methods: </strong>Five-month-old 5 × FAD mice were randomly assigned to four groups, and received either PD (5 mg/kg/day, p.o.), voluntary running, or a combination of both for 47 days. Nest building test, locomotion test, and Morris water maze test were used to evaluate the cognitive function. Immunohistochemical and ELISA analysis was performed to determine Aβ build-up, microglia and astrocytes hyperactivation, and survival neurons in the hippocampus and perirhinal cortex. Real-time quantitative PCR analysis was used to assess the polarization of microglia and astrocytes. HPLC analysis was performed to measure monoamine neurotransmitters in the hippocampus.</p><p><strong>Results and discussion: </strong>The combination of PD and voluntary running synergistically restored nest-building behavior, alleviated recognition and spatial memory deficits, and showed superior effects compared to monotherapy. In addition, the PD and voluntary running combination reduced Aβ build-up, decreased hyperactivation of microglia and astrocytes in the hippocampus and perirhinal cortex, promoted the polarization of inflammatory M1 microglia and reactive astrocytes toward beneficial phenotypes, and lowered systemic circulating pro-inflammatory cytokines while increasing anti-inflammatory cytokines in 5 × FAD mice. Furthermore, combined therapy effectively protected neurons and increased levels of 5-hydroxytryptamine (5-HT) and dopamine (DA) in the hippocampus of 5 × FAD mice. In conclusion, the combination of PD and voluntary running holds great potential as a treatment for AD, offering promise for delaying onset or progression of AD.</p>","PeriodicalId":12450,"journal":{"name":"Frontiers in Aging Neuroscience","volume":"16 ","pages":"1451766"},"PeriodicalIF":4.1,"publicationDate":"2024-09-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11461226/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142389326","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-25eCollection Date: 2024-01-01DOI: 10.3389/fnagi.2024.1462302
Saeid Taheri, Jill Prestopnik, Gary A Rosenberg
Background: Advances in in vivo MRI techniques enable cerebral barrier transfer rates (K trans ) measurement in patients with vascular cognitive impairment and dementia (VCID). However, a consensus has not been reached on the dynamic contribution and importance of cerebral barrier abnormalities to the differential diagnosis of dementia subtypes. Our goal was to investigate the dynamics of blood-brain barrier (BBB) and blood-CSF barrier (BCSFB) K trans in patients with VCID longitudinally and determine the effect of aging.
Methods: We studied subjects at two time points over two years; they were 65.5 years of age (SD = 15.94, M/F = 24/14) at the first visit. We studied 38 patients, 18 of whom had two visits. We calculated the BBB and BCSFB K trans with dynamic contrast-enhanced T1 MR, and we used 1H-MR spectroscopy to measure N-acetylaspartate (NAA) levels in the white matter as a marker of injury. In addition, we measured CSF levels of active-matrix metalloproteinase-3 (MMP3) as an inflammatory biomarker to aid in patient clustering.
Results: Longitudinal BBB measurements revealed variable dynamic behavior: after two years, the BBB K trans increased in 55% of patients and decreased in the remaining 45% unpredictably. We did not find a significant linear model of BBB K trans versus age for VCID. For healthy controls, the model was K trans = 0.0014 + 0.0002 × age, which was significant (p = 0.046). VCID patients showed a reduction in BCSFB K trans compared to healthy controls (p = 0.01). Combining NAA, CSF MMP3, and K trans in a clustering analysis separated patients into groups.
Conclusion: These results suggest that BBB K trans in VCID is dynamic and BCSFB K trans reduced by age. By combining inflammatory biomarkers with BBB K trans data, it is possible to separate VCID patients into distinct groups with different underlying pathologies.
背景:活体磁共振成像技术的进步使血管性认知障碍和痴呆(VCID)患者的脑屏障传递率(K trans)测量成为可能。然而,关于脑屏障异常对痴呆亚型鉴别诊断的动态贡献和重要性,尚未达成共识。我们的目标是纵向研究 VCID 患者血脑屏障(BBB)和血液-脑脊液屏障(BCSFB)K 转的动态变化,并确定衰老的影响:我们在两个时间点对受试者进行了为期两年的研究;受试者首次就诊时的年龄为 65.5 岁(SD = 15.94,男/女 = 24/14)。我们对 38 名患者进行了研究,其中 18 人接受了两次检查。我们用动态对比增强 T1 MR 计算了 BBB 和 BCSFB K 跨度,并用 1H-MR 光谱测量了白质中作为损伤标志物的 N-乙酰天冬氨酸(NAA)水平。此外,我们还测量了作为炎症生物标志物的活性基质金属蛋白酶-3(MMP3)的脑脊液水平,以帮助对患者进行分组:结果:纵向 BBB 测量显示出不同的动态行为:两年后,55% 的患者 BBB K 跨度增加,其余 45% 的患者 BBB K 跨度不可预测地下降。对于 VCID,我们没有发现 BBB K trans 与年龄的显著线性模型。对于健康对照组,该模型为 K trans = 0.0014 + 0.0002 × 年龄,具有显著性(p = 0.046)。与健康对照组相比,VCID 患者的 BCSFB K trans 有所减少(p = 0.01)。将 NAA、CSF MMP3 和 K trans 结合起来进行聚类分析,可将患者分为不同的组别:这些结果表明,VCID的BBB K反式是动态的,BCSFB K反式会随着年龄的增长而减少。通过将炎症生物标志物与BBB K trans数据相结合,可以将VCID患者分为具有不同潜在病理的不同组别。
{"title":"Barriers of the CNS transfer rate dynamics in patients with vascular cognitive impairment and dementia.","authors":"Saeid Taheri, Jill Prestopnik, Gary A Rosenberg","doi":"10.3389/fnagi.2024.1462302","DOIUrl":"10.3389/fnagi.2024.1462302","url":null,"abstract":"<p><strong>Background: </strong>Advances in <i>in vivo</i> MRI techniques enable cerebral barrier transfer rates (K <sub><i>trans</i></sub> ) measurement in patients with vascular cognitive impairment and dementia (VCID). However, a consensus has not been reached on the dynamic contribution and importance of cerebral barrier abnormalities to the differential diagnosis of dementia subtypes. Our goal was to investigate the dynamics of blood-brain barrier (BBB) and blood-CSF barrier (BCSFB) K <sub><i>trans</i></sub> in patients with VCID longitudinally and determine the effect of aging.</p><p><strong>Methods: </strong>We studied subjects at two time points over two years; they were 65.5 years of age (SD = 15.94, M/F = 24/14) at the first visit. We studied 38 patients, 18 of whom had two visits. We calculated the BBB and BCSFB K <sub><i>trans</i></sub> with dynamic contrast-enhanced T1 MR, and we used <sup>1</sup>H-MR spectroscopy to measure N-acetylaspartate (NAA) levels in the white matter as a marker of injury. In addition, we measured CSF levels of active-matrix metalloproteinase-3 (MMP3) as an inflammatory biomarker to aid in patient clustering.</p><p><strong>Results: </strong>Longitudinal BBB measurements revealed variable dynamic behavior: after two years, the BBB K <sub><i>trans</i></sub> increased in 55% of patients and decreased in the remaining 45% unpredictably. We did not find a significant linear model of BBB K <sub><i>trans</i></sub> versus age for VCID. For healthy controls, the model was K <sub><i>trans</i></sub> = 0.0014 + 0.0002 × age, which was significant (<i>p</i> = 0.046). VCID patients showed a reduction in BCSFB K <sub><i>trans</i></sub> compared to healthy controls (<i>p</i> = 0.01). Combining NAA, CSF MMP3, and K <sub><i>trans</i></sub> in a clustering analysis separated patients into groups.</p><p><strong>Conclusion: </strong>These results suggest that BBB K <sub><i>trans</i></sub> in VCID is dynamic and BCSFB K <sub><i>trans</i></sub> reduced by age. By combining inflammatory biomarkers with BBB K <sub><i>trans</i></sub> data, it is possible to separate VCID patients into distinct groups with different underlying pathologies.</p>","PeriodicalId":12450,"journal":{"name":"Frontiers in Aging Neuroscience","volume":"16 ","pages":"1462302"},"PeriodicalIF":4.1,"publicationDate":"2024-09-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11461252/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142389232","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Objective: This study aimed to investigate cortical activation and functional connectivity in the cortex during working memory (WM) tasks in patients with Alzheimer's disease (AD) using functional near-infrared spectroscopy (fNIRS).
Methods: A total of 17 older adults with AD and 17 cognitively normal (CN) participants were recruited. fNIRS was utilized to monitor oxygenated hemoglobin (HbO) concentrations in the frontotemporal lobe, while participants performed WM tasks to examine WM impairments in subjects with AD. Student's t-test for continuous variables and the chi-square test for categorical variables were used to compare the clinical and HbO variables between the AD and CN groups. Functional connectivity was analyzed using Pearson's correlation coefficient between the time series of each channel-to-channel pair.
Results: The changes in HbO concentrations and cortical activations during the WM task showed that the HbO concentration curve of the CN group was higher than that of the AD group during the encoding and maintenance phases of the WM task. Although in the brain region scale, there were no significant differences in average HbO concentrations between the two groups, many channels located in the frontal and temporal lobes showed significant differences (p < 0.05) in the average HbO (channels 7 and 32) and slope HbO values (channels 7, 8, 9, 23, 30, 34, and 38) during the WM task. The average functional connectivity of the AD group was significantly lower than that of the CN group (p < 0.05). The functional connectivity was stronger in the frontopolar (FP) region than in other areas in both groups.
Conclusion: This study revealed there were significant differences in HbO concentration in older adult patients with AD compared to CN during the WM task. The characteristics of HbO measured by the fNIRS technique can be valuable for distinguishing between AD and CN in older adults.
研究目的本研究旨在利用功能性近红外光谱(fNIRS)研究阿尔茨海默病患者在完成工作记忆(WM)任务时大脑皮层的激活和功能连接:利用 fNIRS 监测额颞叶的氧合血红蛋白(HbO)浓度,同时参与者执行 WM 任务,以检查 AD 患者的 WM 损伤。对连续变量采用学生 t 检验,对分类变量采用卡方检验,以比较 AD 组和 CN 组之间的临床和血红蛋白变量。功能连通性采用各通道对时间序列之间的皮尔逊相关系数进行分析:结果:WM 任务中 HbO 浓度和皮层激活的变化显示,在 WM 任务的编码和维持阶段,CN 组的 HbO 浓度曲线高于 AD 组。虽然在脑区尺度上,两组之间的平均 HbO 浓度没有显著差异,但位于额叶和颞叶的许多通道显示出显著差异(p p 结论):本研究显示,老年 AD 患者在完成 WM 任务时的 HbO 浓度与 CN 患者相比存在显著差异。通过 fNIRS 技术测量的 HbO 的特征对于区分老年 AD 和 CN 很有价值。
{"title":"Cortical activation in elderly patients with Alzheimer's disease dementia during working memory tasks: a multichannel fNIRS study.","authors":"Nairong Ruan, Xingxing Li, Ting Xu, Zheng Zhao, Xi Mei, Chengying Zheng","doi":"10.3389/fnagi.2024.1433551","DOIUrl":"10.3389/fnagi.2024.1433551","url":null,"abstract":"<p><strong>Objective: </strong>This study aimed to investigate cortical activation and functional connectivity in the cortex during working memory (WM) tasks in patients with Alzheimer's disease (AD) using functional near-infrared spectroscopy (fNIRS).</p><p><strong>Methods: </strong>A total of 17 older adults with AD and 17 cognitively normal (CN) participants were recruited. fNIRS was utilized to monitor oxygenated hemoglobin (HbO) concentrations in the frontotemporal lobe, while participants performed WM tasks to examine WM impairments in subjects with AD. Student's t-test for continuous variables and the chi-square test for categorical variables were used to compare the clinical and HbO variables between the AD and CN groups. Functional connectivity was analyzed using Pearson's correlation coefficient between the time series of each channel-to-channel pair.</p><p><strong>Results: </strong>The changes in HbO concentrations and cortical activations during the WM task showed that the HbO concentration curve of the CN group was higher than that of the AD group during the encoding and maintenance phases of the WM task. Although in the brain region scale, there were no significant differences in average HbO concentrations between the two groups, many channels located in the frontal and temporal lobes showed significant differences (<i>p</i> < 0.05) in the average HbO (channels 7 and 32) and slope HbO values (channels 7, 8, 9, 23, 30, 34, and 38) during the WM task. The average functional connectivity of the AD group was significantly lower than that of the CN group (<i>p</i> < 0.05). The functional connectivity was stronger in the frontopolar (FP) region than in other areas in both groups.</p><p><strong>Conclusion: </strong>This study revealed there were significant differences in HbO concentration in older adult patients with AD compared to CN during the WM task. The characteristics of HbO measured by the fNIRS technique can be valuable for distinguishing between AD and CN in older adults.</p>","PeriodicalId":12450,"journal":{"name":"Frontiers in Aging Neuroscience","volume":"16 ","pages":"1433551"},"PeriodicalIF":4.1,"publicationDate":"2024-09-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11461194/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142389235","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-09-25eCollection Date: 2024-01-01DOI: 10.3389/fnagi.2024.1418081
Qian-Qian Fan, Yong-Min Chen, Yong-Sen Fu, Xiao-Shan Li, Ji Zeng, Shao-Zhen Bian, Bin-Bin Li, Zhen-Hua Song
Objective: In this study, we examined the effectiveness of hyperbaric oxygen (HBO) therapy in ameliorating cognitive deficits in mice with Alzheimer's disease (AD), while also assessing its impact on the autophagic pathway within the context of AD.
Methods: 20 double-transgenic mice expressing the amyloid precursor protein and presenilin 1 (APP/PS1) were purposefully selected and randomly assigned to groups A and B. Concurrently, 20 C57BL/6 mice were chosen and randomly categorized into groups C and D, each consisting of 10 mice. Mice in groups B and D received HBO treatment. The Morris water maze assay was used to assess changes in mouse behavior. Immunohistochemistry techniques were used to quantify the expression levels of amyloid-beta 42 (Aβ42) and microtubule-associated protein 1A/1B-light chain 3 (LC3) in hippocampal tissues, while western blot analysis was used to investigate the levels of LC3-II, p62, phosphoinositide 3-kinase (PI3K), and mammalian target of rapamycin (mTOR) proteins within hippocampal tissues.
Results: Mice allocated to group B exhibited reduced escape latency and prolonged dwell time in the target quadrant compared to other groups. Histological examination revealed conspicuous plaque-like deposits of Aβ42 in the hippocampal tissues of mice in groups A and B. Group B displayed diminished Aβ42-positive reactants and augmented microtubule-associated protein 1A/1B-LC3-positive reactants compared to group A. LC3-positive reactants were also detected in the hippocampal tissues of mice in groups C and D, surpassing the levels observed in groups A and B. Furthermore, group B demonstrated significantly lower expression of mTOR protein and markedly higher expression of LC3-II protein in mouse hippocampal tissues when compared to group A (P < 0.05). Conversely, there were no significant disparities noted in PI3K and p62 protein expression between groups B and A. Notably, no discernible discrepancies were observed in the expression levels of mTOR, PI3K, LC3-II, and p62 proteins between groups C and D within mouse hippocampal tissues.
Conclusion: HBO treatment demonstrates efficacy in enhancing cognitive function in mice with AD and holds promise as a potential therapeutic intervention for AD by facilitating the activation of the mTOR pathway-mediated autophagy.
研究目的在这项研究中,我们考察了高压氧疗法(HBO)在改善阿尔茨海默病(AD)小鼠认知障碍方面的有效性,同时还评估了高压氧疗法在 AD 背景下对自噬途径的影响。方法:特意挑选了20只表达淀粉样前体蛋白和早老素1(APP/PS1)的双转基因小鼠,并将其随机分配到A组和B组;同时挑选了20只C57BL/6小鼠,并将其随机分为C组和D组,每组10只。B 组和 D 组小鼠接受 HBO 治疗。莫里斯水迷宫试验用于评估小鼠行为的变化。免疫组化技术用于量化海马组织中淀粉样β42(Aβ42)和微管相关蛋白1A/1B-轻链3(LC3)的表达水平,而Western印迹分析则用于研究海马组织中LC3-II、p62、磷酸肌醇3-激酶(PI3K)和哺乳动物雷帕霉素靶蛋白(mTOR)的水平:与其他组相比,B组小鼠的逃避潜伏期缩短,在目标象限的停留时间延长。与 A 组相比,B 组的 Aβ42 阳性反应物减少,而微管相关蛋白 1A/1B-LC3 阳性反应物增加。此外,与 A 组相比,B 组小鼠海马组织中 mTOR 蛋白的表达明显降低,LC3-II 蛋白的表达明显升高(P < 0.05)。值得注意的是,C 组和 D 组小鼠海马组织中 mTOR、PI3K、LC3-II 和 p62 蛋白的表达水平没有明显差异:HBO治疗可有效增强AD小鼠的认知功能,通过促进mTOR通路介导的自噬激活,有望成为一种潜在的AD治疗干预方法。
{"title":"Enhancement of cognitive function in mice with Alzheimer's disease through hyperbaric oxygen-induced activation of cellular autophagy.","authors":"Qian-Qian Fan, Yong-Min Chen, Yong-Sen Fu, Xiao-Shan Li, Ji Zeng, Shao-Zhen Bian, Bin-Bin Li, Zhen-Hua Song","doi":"10.3389/fnagi.2024.1418081","DOIUrl":"10.3389/fnagi.2024.1418081","url":null,"abstract":"<p><strong>Objective: </strong>In this study, we examined the effectiveness of hyperbaric oxygen (HBO) therapy in ameliorating cognitive deficits in mice with Alzheimer's disease (AD), while also assessing its impact on the autophagic pathway within the context of AD.</p><p><strong>Methods: </strong>20 double-transgenic mice expressing the amyloid precursor protein and presenilin 1 (APP/PS1) were purposefully selected and randomly assigned to groups A and B. Concurrently, 20 C57BL/6 mice were chosen and randomly categorized into groups C and D, each consisting of 10 mice. Mice in groups B and D received HBO treatment. The Morris water maze assay was used to assess changes in mouse behavior. Immunohistochemistry techniques were used to quantify the expression levels of amyloid-beta 42 (Aβ42) and microtubule-associated protein 1A/1B-light chain 3 (LC3) in hippocampal tissues, while western blot analysis was used to investigate the levels of LC3-II, p62, phosphoinositide 3-kinase (PI3K), and mammalian target of rapamycin (mTOR) proteins within hippocampal tissues.</p><p><strong>Results: </strong>Mice allocated to group B exhibited reduced escape latency and prolonged dwell time in the target quadrant compared to other groups. Histological examination revealed conspicuous plaque-like deposits of Aβ42 in the hippocampal tissues of mice in groups A and B. Group B displayed diminished Aβ42-positive reactants and augmented microtubule-associated protein 1A/1B-LC3-positive reactants compared to group A. LC3-positive reactants were also detected in the hippocampal tissues of mice in groups C and D, surpassing the levels observed in groups A and B. Furthermore, group B demonstrated significantly lower expression of mTOR protein and markedly higher expression of LC3-II protein in mouse hippocampal tissues when compared to group A (<i>P</i> < 0.05). Conversely, there were no significant disparities noted in PI3K and p62 protein expression between groups B and A. Notably, no discernible discrepancies were observed in the expression levels of mTOR, PI3K, LC3-II, and p62 proteins between groups C and D within mouse hippocampal tissues.</p><p><strong>Conclusion: </strong>HBO treatment demonstrates efficacy in enhancing cognitive function in mice with AD and holds promise as a potential therapeutic intervention for AD by facilitating the activation of the mTOR pathway-mediated autophagy.</p>","PeriodicalId":12450,"journal":{"name":"Frontiers in Aging Neuroscience","volume":"16 ","pages":"1418081"},"PeriodicalIF":4.1,"publicationDate":"2024-09-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11461206/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142389236","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}