首页 > 最新文献

Neurochemical Research最新文献

英文 中文
Electroacupuncture Attenuates Nerve Injury in Cerebral Small Vessel Disease by Protecting the Neurovascular Units
IF 3.7 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-04-22 DOI: 10.1007/s11064-025-04395-x
Shujie Li, Rui Zhao, Yongsheng Han

Studies have found that electroacupuncture (EA) can improve the neurocognitive function of cerebral small vessel disease (CSVD), restore cerebral blood flow, and protect neurovascular units. The occurrence and development of cerebral microvascular disease is highly related to neurovascular unit injury. However, it is not clear whether EA plays a therapeutic role by restoring neurovascular unit injury. To explore the possible therapeutic mechanism of EA by analyzing its effect on CSVD neurovascular units in rats with CSVD. Adult male Sprague–Dawley rats (n = 36) were used for the experiment. The rat model of bilateral carotid artery occlusion (BCAO) was established by bilateral common carotid artery ligation. The treatment group was treated with 2/100 Hz and 2–4 V continuous wave EA every day for 7 days. The water maze test and new object recognition test were used to evaluate the memory and cognition of rats. Golgi staining was performed to evaluate the synaptic plasticity. Western blotting was used to evaluate the expression of synaptic-associated proteins PSD95 and synaptophysin and neurovascular unit-associated proteins VEGF, NeuN, GFAP, and claudin5. The expression of neurovascular unit associated proteins VEGF, NeuN and GFAP was further evaluated by immunofluorescence staining. EA intervention significantly reduced cognitive memory damage, restored neuronal synaptic plasticity, and reduced neurovascular unit damage. EA significantly shortened the latency in the water maze test (p < 0.01), increased the number of platform crossings (p < 0.01) and the mean speed (p < 0.01), and increased new object recognition index (p < 0.01). EA significantly increased the total length of neuronal dendrites (p < 0.01) and the dendrite spinous density (p < 0.01). EA increased the levels of PSD95, Synaptophysin, VEGF, NeuN, GFAP and Claudin5 in the EA + BCAO group, compared with the BCAO group (p < 0.01). EA could improve the neurological function in a rat model of cerebral small vessel disease, and its mechanism may be related to the protective effect of electroacupuncture on neurovascular units.

{"title":"Electroacupuncture Attenuates Nerve Injury in Cerebral Small Vessel Disease by Protecting the Neurovascular Units","authors":"Shujie Li,&nbsp;Rui Zhao,&nbsp;Yongsheng Han","doi":"10.1007/s11064-025-04395-x","DOIUrl":"10.1007/s11064-025-04395-x","url":null,"abstract":"<div><p>Studies have found that electroacupuncture (EA) can improve the neurocognitive function of cerebral small vessel disease (CSVD), restore cerebral blood flow, and protect neurovascular units. The occurrence and development of cerebral microvascular disease is highly related to neurovascular unit injury. However, it is not clear whether EA plays a therapeutic role by restoring neurovascular unit injury. To explore the possible therapeutic mechanism of EA by analyzing its effect on CSVD neurovascular units in rats with CSVD. Adult male Sprague–Dawley rats (<i>n</i> = 36) were used for the experiment. The rat model of bilateral carotid artery occlusion (BCAO) was established by bilateral common carotid artery ligation. The treatment group was treated with 2/100 Hz and 2–4 V continuous wave EA every day for 7 days. The water maze test and new object recognition test were used to evaluate the memory and cognition of rats. Golgi staining was performed to evaluate the synaptic plasticity. Western blotting was used to evaluate the expression of synaptic-associated proteins PSD95 and synaptophysin and neurovascular unit-associated proteins VEGF, NeuN, GFAP, and claudin5. The expression of neurovascular unit associated proteins VEGF, NeuN and GFAP was further evaluated by immunofluorescence staining. EA intervention significantly reduced cognitive memory damage, restored neuronal synaptic plasticity, and reduced neurovascular unit damage. EA significantly shortened the latency in the water maze test (<i>p</i> &lt; 0.01), increased the number of platform crossings (<i>p</i> &lt; 0.01) and the mean speed (<i>p</i> &lt; 0.01), and increased new object recognition index (<i>p</i> &lt; 0.01). EA significantly increased the total length of neuronal dendrites (<i>p</i> &lt; 0.01) and the dendrite spinous density (<i>p</i> &lt; 0.01). EA increased the levels of PSD95, Synaptophysin, VEGF, NeuN, GFAP and Claudin5 in the EA + BCAO group, compared with the BCAO group (<i>p</i> &lt; 0.01). EA could improve the neurological function in a rat model of cerebral small vessel disease, and its mechanism may be related to the protective effect of electroacupuncture on neurovascular units.</p></div>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":"50 3","pages":""},"PeriodicalIF":3.7,"publicationDate":"2025-04-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://link.springer.com/content/pdf/10.1007/s11064-025-04395-x.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143856410","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Electroacupuncture Ameliorates Chronic Inflammatory Pain and Depression Comorbidity by Inhibiting Nrf2-Mediated Ferroptosis in Hippocampal Neurons
IF 3.7 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-04-21 DOI: 10.1007/s11064-025-04401-2
Guanghua Liu, Dandan Liu, Dongliang Shi, Zihua Wang, Wen Fu

Chronic inflammatory pain and depression are highly comorbid, with nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated ferroptosis in hippocampal neurons strongly associated with the onset and progression of the comorbidity. Electroacupuncture (EA), widely used to treat pain and mood disorders, may ameliorate chronic inflammatory pain and depression comorbidity (CIPDC) by inhibiting Nrf2-mediated ferroptosis in hippocampal neurons, though its mechanism of action remains partially understood. In this study, we established the CIPDC model by administering a subcutaneous injection of complete Freund’s adjuvant (CFA) into the left hind paw. Evaluations of EA’s effects on pain thresholds and depressive behaviors in CIPDC rats included paw withdrawal mechanical threshold, paw withdrawal thermal latency, sucrose preference test, open field test, and forced swim test assessments. HE staining was performed to assess the pathological and morphological alterations in hippocampal neurons. FJB staining was utilized to evaluate neuronal degeneration, while transmission electron microscopy (TEM) was employed to examine ultrastructural changes in hippocampal neuronal mitochondria. Prussian blue staining was conducted to visualize ferrous ion deposition in the hippocampus, and the contents of ferrous ion (Fe2+), malondialdehyde (MDA), and glutathione (GSH) were measured using colorimetric assay kits. Western blotting (WB) was performed to determine the relative protein expression of Nrf2, FTH1, FTL, xCT, GPX4, ACSL4, LPCAT3, and LOX in the hippocampus. Additionally, the relative mRNA expression of FTH1, FTL, xCT, GPX4, ACSL4, LPCAT3, and LOX was analyzed by PCR. Flow cytometry was used to quantify ROS levels in the hippocampus, and immunofluorescence staining was applied to detect nuclear expression of Nrf2 as well as co-localization of GPX4 with the neuronal marker NeuN. Our results demonstrate that EA upregulates nuclear Nrf2 expression in hippocampal tissue, thereby alleviating iron metabolism dysregulation, enhancing antioxidant system activity, and reducing lipid peroxidation. This process inhibits ferroptosis in hippocampal neurons, promoting their repair and remodeling, and effectively treating CIPDC.

{"title":"Electroacupuncture Ameliorates Chronic Inflammatory Pain and Depression Comorbidity by Inhibiting Nrf2-Mediated Ferroptosis in Hippocampal Neurons","authors":"Guanghua Liu,&nbsp;Dandan Liu,&nbsp;Dongliang Shi,&nbsp;Zihua Wang,&nbsp;Wen Fu","doi":"10.1007/s11064-025-04401-2","DOIUrl":"10.1007/s11064-025-04401-2","url":null,"abstract":"<div><p>Chronic inflammatory pain and depression are highly comorbid, with nuclear factor erythroid 2-related factor 2 (Nrf2)-mediated ferroptosis in hippocampal neurons strongly associated with the onset and progression of the comorbidity. Electroacupuncture (EA), widely used to treat pain and mood disorders, may ameliorate chronic inflammatory pain and depression comorbidity (CIPDC) by inhibiting Nrf2-mediated ferroptosis in hippocampal neurons, though its mechanism of action remains partially understood. In this study, we established the CIPDC model by administering a subcutaneous injection of complete Freund’s adjuvant (CFA) into the left hind paw. Evaluations of EA’s effects on pain thresholds and depressive behaviors in CIPDC rats included paw withdrawal mechanical threshold, paw withdrawal thermal latency, sucrose preference test, open field test, and forced swim test assessments. HE staining was performed to assess the pathological and morphological alterations in hippocampal neurons. FJB staining was utilized to evaluate neuronal degeneration, while transmission electron microscopy (TEM) was employed to examine ultrastructural changes in hippocampal neuronal mitochondria. Prussian blue staining was conducted to visualize ferrous ion deposition in the hippocampus, and the contents of ferrous ion (Fe<sup>2+</sup>), malondialdehyde (MDA), and glutathione (GSH) were measured using colorimetric assay kits. Western blotting (WB) was performed to determine the relative protein expression of Nrf2, FTH1, FTL, xCT, GPX4, ACSL4, LPCAT3, and LOX in the hippocampus. Additionally, the relative mRNA expression of FTH1, FTL, xCT, GPX4, ACSL4, LPCAT3, and LOX was analyzed by PCR. Flow cytometry was used to quantify ROS levels in the hippocampus, and immunofluorescence staining was applied to detect nuclear expression of Nrf2 as well as co-localization of GPX4 with the neuronal marker NeuN. Our results demonstrate that EA upregulates nuclear Nrf2 expression in hippocampal tissue, thereby alleviating iron metabolism dysregulation, enhancing antioxidant system activity, and reducing lipid peroxidation. This process inhibits ferroptosis in hippocampal neurons, promoting their repair and remodeling, and effectively treating CIPDC.</p></div>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":"50 3","pages":""},"PeriodicalIF":3.7,"publicationDate":"2025-04-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143852511","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Oxytocin Anti-Apoptotic Potential Mediates Neuroprotection Against 3-Nitropropionic Acid-Induced Huntington’s Disease-Like Pathophysiology in Rats: Involvement of Calpain-2/p25 Cdk5/MEF-2 Signaling Pathway
IF 3.7 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-04-19 DOI: 10.1007/s11064-025-04397-9
Marwa Rabie, Dalia M. El-Tanbouly, Esraa A. Kandil, Helmy M. Sayed

The increasing interest in the pro-apoptotic function of calpain-2 in the course of Huntington’s disease (HD) is attributed to the involvement of its substrate, cyclin-dependent kinase 5 (Cdk5), in neuronal death during neurodegeneration. Oxytocin has been demonstrated to suppress apoptosis in many neurodegenerative disorders. This research aimed to investigate the effect of oxytocin on several calpain 2-induced apoptogenic factors in 3-nitropropionic acid (3-NP) animal model of HD in rats. For 14 days, rats received 3-NP (10 mg/kg, i.p.), and oxytocin (160 µg/kg, i.p.) 1 h before 3-NP administration. Oxytocin reversed the detrimental effects of 3-NP on the striatum, which was evidenced by improvement of motor behavior, as well as histological picture and neurochemical balance. Oxytocin markedly reduced striatal calpain-2 and p25 Cdk5 protein expressions and increased the endogenous calpain inhibitor, calpastatin expression along with the pro-survival factor, myocyte-enhancer factor 2 (MEF-2) contents. Moreover, it suppressed striatal content of the pro-apoptotic biomarkers (BCl-2-associated X protein (Bax), tumor suppressor protein (p53), and caspase-3) and elevated striatal anti-apoptotic B-cell lymphoma/leukemia 2 (BCl-2) content. It repressed the release of mitochondrial cytochrome c and apoptosis-inducing factor (AIF) to hinder caspase-dependent and caspase-independent apoptotic neuronal death. Oxytocin could be a promising candidate for HD management by hampering both mitochondrial and non-mitochondrial apoptosis through inhibition of calpain-2/p25 Cdk5/MEF-2 pathway.

{"title":"Oxytocin Anti-Apoptotic Potential Mediates Neuroprotection Against 3-Nitropropionic Acid-Induced Huntington’s Disease-Like Pathophysiology in Rats: Involvement of Calpain-2/p25 Cdk5/MEF-2 Signaling Pathway","authors":"Marwa Rabie,&nbsp;Dalia M. El-Tanbouly,&nbsp;Esraa A. Kandil,&nbsp;Helmy M. Sayed","doi":"10.1007/s11064-025-04397-9","DOIUrl":"10.1007/s11064-025-04397-9","url":null,"abstract":"<div><p>The increasing interest in the pro-apoptotic function of calpain-2 in the course of Huntington’s disease (HD) is attributed to the involvement of its substrate, cyclin-dependent kinase 5 (Cdk5), in neuronal death during neurodegeneration. Oxytocin has been demonstrated to suppress apoptosis in many neurodegenerative disorders. This research aimed to investigate the effect of oxytocin on several calpain 2-induced apoptogenic factors in 3-nitropropionic acid (3-NP) animal model of HD in rats. For 14 days, rats received 3-NP (10 mg/kg, i.p.), and oxytocin (160 µg/kg, i.p.) 1 h before 3-NP administration. Oxytocin reversed the detrimental effects of 3-NP on the striatum, which was evidenced by improvement of motor behavior, as well as histological picture and neurochemical balance. Oxytocin markedly reduced striatal calpain-2 and p25 Cdk5 protein expressions and increased the endogenous calpain inhibitor, calpastatin expression along with the pro-survival factor, myocyte-enhancer factor 2 (MEF-2) contents. Moreover, it suppressed striatal content of the pro-apoptotic biomarkers (BCl-2-associated X protein (Bax), tumor suppressor protein (p53), and caspase-3) and elevated striatal anti-apoptotic B-cell lymphoma/leukemia 2 (BCl-2) content. It repressed the release of mitochondrial cytochrome c and apoptosis-inducing factor (AIF) to hinder caspase-dependent and caspase-independent apoptotic neuronal death. Oxytocin could be a promising candidate for HD management by hampering both mitochondrial and non-mitochondrial apoptosis through inhibition of calpain-2/p25 Cdk5/MEF-2 pathway.</p></div>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":"50 3","pages":""},"PeriodicalIF":3.7,"publicationDate":"2025-04-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143849014","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
(+)-Borneol Enhances the Antiseizure Effects of Retigabine by both Pharmacokinetic and Pharmacodynamic Interaction
IF 3.7 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-04-19 DOI: 10.1007/s11064-025-04396-w
Haiyan Xu, Zhidan Wu, Pei Wang, Jili Gong, Li Qiu, Yueling Gu, Li Zhan, Fuyun Tian, Zhaobing Gao

Epilepsy is a chronic neurological disorder characterized by recurrent seizures, approximately one-third of whom are resistant to current anti-seizure drugs (ASDs). Retigabine (RTG) is a potential treatment for treating drug-resistant epilepsy and KCNQ2-related developmental and epileptic encephalopathy (KCNQ2-DEE). However, its use is limited by side effects from high doses and long-term use. This study aims to evaluate the anticonvulsant efficacy of RTG in combination with (+)-borneol in mouse models of maximal electroshock seizure (MES) and 6-Hz (44-mA) seizure. The individual anti-seizure efficacy of RTG and (+)-borneol was evaluated in the MES and 6-Hz seizure models, then isobolographic analysis was conducted to assess their interactions. The plasma and brain concentrations of RTG were measured with and without (+)-borneol. Electrophysiological experiments using the patch-clamp technique investigated the interactions of (+)-borneol and RTG at the α1β3γ2L-GABAAR and KCNQ2 channels. Both RTG and (+)-borneol exhibited anticonvulsant activity in MES and 6-Hz seizure models. In the isobolographic analysis, the co-administration of RTG and (+)-borneol proved to be significantly more effective than predicted based on additive effects. The ED50mix was reduced by approximately 20 to 100-fold and 2 to 6-fold compared to the ED50add in the MES and 6-Hz models, respectively. The plasma and brain levels of RTG increased following co-administration with higher doses of (+)-borneol. Patch-clamp studies indicated that both RTG and (+)-borneol positively modulated α1β3γ2L-GABAAR currents and showed additive effects. However, (+)-borneol inhibited the KCNQ2 current at 100 µM and did not enhance RTG activation on KCNQ2 channels at this concentration. These results demonstrate that (+)-borneol enhances the antiseizure effects of RTG by both pharmacokinetic and pharmacodynamic interaction and this approach may be clinically effective for patients with intractable seizures or KCNQ2-DEE.

{"title":"(+)-Borneol Enhances the Antiseizure Effects of Retigabine by both Pharmacokinetic and Pharmacodynamic Interaction","authors":"Haiyan Xu,&nbsp;Zhidan Wu,&nbsp;Pei Wang,&nbsp;Jili Gong,&nbsp;Li Qiu,&nbsp;Yueling Gu,&nbsp;Li Zhan,&nbsp;Fuyun Tian,&nbsp;Zhaobing Gao","doi":"10.1007/s11064-025-04396-w","DOIUrl":"10.1007/s11064-025-04396-w","url":null,"abstract":"<div><p>Epilepsy is a chronic neurological disorder characterized by recurrent seizures, approximately one-third of whom are resistant to current anti-seizure drugs (ASDs). Retigabine (RTG) is a potential treatment for treating drug-resistant epilepsy and KCNQ2-related developmental and epileptic encephalopathy (KCNQ2-DEE). However, its use is limited by side effects from high doses and long-term use. This study aims to evaluate the anticonvulsant efficacy of RTG in combination with (+)-borneol in mouse models of maximal electroshock seizure (MES) and 6-Hz (44-mA) seizure. The individual anti-seizure efficacy of RTG and (+)-borneol was evaluated in the MES and 6-Hz seizure models, then isobolographic analysis was conducted to assess their interactions. The plasma and brain concentrations of RTG were measured with and without (+)-borneol. Electrophysiological experiments using the patch-clamp technique investigated the interactions of (+)-borneol and RTG at the α1β3γ2L-GABAAR and KCNQ2 channels. Both RTG and (+)-borneol exhibited anticonvulsant activity in MES and 6-Hz seizure models. In the isobolographic analysis, the co-administration of RTG and (+)-borneol proved to be significantly more effective than predicted based on additive effects. The ED50mix was reduced by approximately 20 to 100-fold and 2 to 6-fold compared to the ED50add in the MES and 6-Hz models, respectively. The plasma and brain levels of RTG increased following co-administration with higher doses of (+)-borneol. Patch-clamp studies indicated that both RTG and (+)-borneol positively modulated α1β3γ2L-GABAAR currents and showed additive effects. However, (+)-borneol inhibited the KCNQ2 current at 100 µM and did not enhance RTG activation on KCNQ2 channels at this concentration. These results demonstrate that (+)-borneol enhances the antiseizure effects of RTG by both pharmacokinetic and pharmacodynamic interaction and this approach may be clinically effective for patients with intractable seizures or KCNQ2-DEE.</p></div>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":"50 3","pages":""},"PeriodicalIF":3.7,"publicationDate":"2025-04-19","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143849015","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Prenatal Exposure To Valproic Acid Induces Increased Autism-Like Behaviors and Impairment of Learning and Memory Functions in Rat Offspring by Upregulating ADAM10 Expression
IF 3.7 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-04-17 DOI: 10.1007/s11064-025-04398-8
Jingyuan Yang, Xiaoli Li, Jing Tan, Ping Zhou, Lingjun Hu, Jie Chen, Tingyu Li, Yonggang Liu, Li Chen

Autism spectrum disorder (ASD) involves a complex neurodevelopmental pathogenesis. A disintegrin and metalloproteinase 10 (ADAM10) plays a crucial role in embryonic brain development and neural network stability. This study aimed to investigate the influence of ADAM10 on excitation/inhibition (E/I) balance, autism-like behaviors, and learning and memory dysfunction in rats prenatally exposed to valproic acid (VPA) and determine potential intervention strategies. The VPA-exposed group exhibited increased levels of ADAM10 and secreted amyloid precursor protein-α (sAPPα). Moreover, overexpression of glutamate decarboxylase 1 and N-methyl-D-aspartate receptors was observed. High-performance liquid chromatography-mass spectrometry revealed elevated levels of glutamate, glutamine, and γ-aminobutyric acid, as well as an E/I imbalance in the VPA group. Additionally, narrower synaptic clefts as well as increased postsynaptic density and synaptic vesicles were observed. Remarkably, intraperitoneal administration of ADAM10 inhibitor during the critical period of synaptic development significantly improved ASD-like behavior and learning and memory function in VPA-exposed rats. This intervention effectively reduced abnormally high sAPPα levels in the prefrontal cortex and corrected abnormal E/I balance. Thus, inhibiting ADAM10 overexpression may improve the E/I imbalance, alleviate core symptoms of ASD, and improve learning and memory dysfunction. The use of ADAM10 inhibitor represents a potential therapeutic strategy for treating ASD patients with intellectual disabilities.

{"title":"Prenatal Exposure To Valproic Acid Induces Increased Autism-Like Behaviors and Impairment of Learning and Memory Functions in Rat Offspring by Upregulating ADAM10 Expression","authors":"Jingyuan Yang,&nbsp;Xiaoli Li,&nbsp;Jing Tan,&nbsp;Ping Zhou,&nbsp;Lingjun Hu,&nbsp;Jie Chen,&nbsp;Tingyu Li,&nbsp;Yonggang Liu,&nbsp;Li Chen","doi":"10.1007/s11064-025-04398-8","DOIUrl":"10.1007/s11064-025-04398-8","url":null,"abstract":"<div><p>Autism spectrum disorder (ASD) involves a complex neurodevelopmental pathogenesis. A disintegrin and metalloproteinase 10 (ADAM10) plays a crucial role in embryonic brain development and neural network stability. This study aimed to investigate the influence of ADAM10 on excitation/inhibition (E/I) balance, autism-like behaviors, and learning and memory dysfunction in rats prenatally exposed to valproic acid (VPA) and determine potential intervention strategies. The VPA-exposed group exhibited increased levels of ADAM10 and secreted amyloid precursor protein-α (sAPPα). Moreover, overexpression of glutamate decarboxylase 1 and N-methyl-D-aspartate receptors was observed. High-performance liquid chromatography-mass spectrometry revealed elevated levels of glutamate, glutamine, and γ-aminobutyric acid, as well as an E/I imbalance in the VPA group. Additionally, narrower synaptic clefts as well as increased postsynaptic density and synaptic vesicles were observed. Remarkably, intraperitoneal administration of ADAM10 inhibitor during the critical period of synaptic development significantly improved ASD-like behavior and learning and memory function in VPA-exposed rats. This intervention effectively reduced abnormally high sAPPα levels in the prefrontal cortex and corrected abnormal E/I balance. Thus, inhibiting ADAM10 overexpression may improve the E/I imbalance, alleviate core symptoms of ASD, and improve learning and memory dysfunction. The use of ADAM10 inhibitor represents a potential therapeutic strategy for treating ASD patients with intellectual disabilities.</p></div>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":"50 3","pages":""},"PeriodicalIF":3.7,"publicationDate":"2025-04-17","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://link.springer.com/content/pdf/10.1007/s11064-025-04398-8.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143840296","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Unravelling the Proteinopathic Engagement of α-Synuclein, Tau, and Amyloid Beta in Parkinson’s Disease: Mitochondrial Collapse as a Pivotal Driver of Neurodegeneration
IF 3.7 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-04-16 DOI: 10.1007/s11064-025-04399-7
Tarini Ashish Sahoo, Jagdish Chand, Amarjith Thiyyar Kandy, Shanish Antony, Gomathy Subramanian

Parkinson's disease is a complex neurological ailment manifested by dopaminergic neurodegeneration in the substantia nigra of the brain. This study investigates the molecular tripartite interaction between Lewy bodies, amyloid beta, and tau protein in the pathogenesis of Parkinson's disease. Lewy bodies which have been found as the important pathological hallmark in the degenerative neurons of Parkinson’s patients, are mainly composed of α-synuclein. The accumulation of α-synuclein has been directly and indirectly linked to the severity and degree of progression of the disease. In addition, approximately 50% of Parkinson's disease cases are also described by hyperphosphorylation of tau protein indicating its significant involvement in the disease. The study further explains how α-synuclein, tau and amyloid beta can spread via cross-seeding mechanisms and accelerate each other's aggregation leading to neuronal death. Both GSK-3β and CDK5 are involved in phosphorylation which among other effects contributes to the misfolding of both α-synuclein and tau proteins that lead to neurodegeneration in Alzheimer’s disease. Several mediators, that contribute to mitochondrial damage through elevated oxidative stress pathology are clearly described. Because of the increase in the incidence of Parkinson's disease, as predicted to be 17 million when the study was being conducted, studying these pathological mechanisms is very important in trying to establish treatments. This work contributes a path to finding a multi-target treatment regimen to alleviate the burden of this devastating disease.

{"title":"Unravelling the Proteinopathic Engagement of α-Synuclein, Tau, and Amyloid Beta in Parkinson’s Disease: Mitochondrial Collapse as a Pivotal Driver of Neurodegeneration","authors":"Tarini Ashish Sahoo,&nbsp;Jagdish Chand,&nbsp;Amarjith Thiyyar Kandy,&nbsp;Shanish Antony,&nbsp;Gomathy Subramanian","doi":"10.1007/s11064-025-04399-7","DOIUrl":"10.1007/s11064-025-04399-7","url":null,"abstract":"<div><p>Parkinson's disease is a complex neurological ailment manifested by dopaminergic neurodegeneration in the substantia nigra of the brain. This study investigates the molecular tripartite interaction between Lewy bodies, amyloid beta, and tau protein in the pathogenesis of Parkinson's disease. Lewy bodies which have been found as the important pathological hallmark in the degenerative neurons of Parkinson’s patients, are mainly composed of α-synuclein. The accumulation of α-synuclein has been directly and indirectly linked to the severity and degree of progression of the disease. In addition, approximately 50% of Parkinson's disease cases are also described by hyperphosphorylation of tau protein indicating its significant involvement in the disease. The study further explains how α-synuclein, tau and amyloid beta can spread via cross-seeding mechanisms and accelerate each other's aggregation leading to neuronal death. Both GSK-3β and CDK5 are involved in phosphorylation which among other effects contributes to the misfolding of both α-synuclein and tau proteins that lead to neurodegeneration in Alzheimer’s disease. Several mediators, that contribute to mitochondrial damage through elevated oxidative stress pathology are clearly described. Because of the increase in the incidence of Parkinson's disease, as predicted to be 17 million when the study was being conducted, studying these pathological mechanisms is very important in trying to establish treatments. This work contributes a path to finding a multi-target treatment regimen to alleviate the burden of this devastating disease.</p></div>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":"50 3","pages":""},"PeriodicalIF":3.7,"publicationDate":"2025-04-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143840484","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Alpha-Pinene Ameliorates Memory Deficits in 3-Nitropropionic Acid-Induced Rat Model of Huntington’s Disease
IF 3.7 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-04-16 DOI: 10.1007/s11064-025-04393-z
Paria Hashemi, Mohammad Raman Moloudi, Helia Rahmani, Katayoun Hassanzadeh, Zakaria Vahabzadeh, Esmael Izadpanah

Memory impairment is one of the cognitive symptoms in Huntington’s disease (HD) which appears before motor dysfunction in patients. Various molecular mechanisms, including disruptions in neurotrophins levels, are involved in the occurrence of memory problems in HD. Alpha-pinene (APN), a member of the monoterpene family, exhibited beneficial effects in animal models of neurodegenerative disorders. As a result, this study assessed the impact of APN on memory in the 3-nitropropionic acid (3-NP) induced model of HD in rats. Male Wistar rats received saline, 3-NP to model HD, or APN (1, 5, or 10 mg/kg) plus 3-NP for 21 days to assess APN’s effects. Working and spatial memory were examined by the Y-maze and Morris-water-maze (MWM) tests. The mRNA levels of neurotrophins and their receptors in the brain cortex and hippocampus of the rats were quantitatively assessed through Reverse Transcription Quantitative Polymerase Chain Reaction (RT-qPCR) analysis. The results showed that APN, at all three doses, significantly prevented the disease phenotype induced by 3-NP administration. In addition, APN treatment elevated the gene expression levels of BDNF, TrkA, TrkB, and CREB, while significantly decreasing P75 NTR showing a dose-dependent effect in the brain cortex and hippocampus, compared to the 3-NP group. These findings suggest that APN alleviates 3-NP-induced memory deficits by enhancing neurotrophins and their receptor levels in an animal model of HD.

记忆障碍是亨廷顿氏病(Huntington's disease,HD)患者的认知症状之一,出现在运动功能障碍之前。包括神经营养素水平紊乱在内的各种分子机制都与 HD 记忆障碍的发生有关。α-蒎烯(APN)是单萜烯家族的一员,在神经退行性疾病的动物模型中表现出有益的作用。因此,本研究评估了α-蒎烯在 3-硝基丙酸(3-NP)诱导的 HD 模型中对大鼠记忆力的影响。雄性 Wistar 大鼠接受生理盐水、3-NP 或 APN(1、5 或 10 毫克/千克)加 3-NP 治疗 21 天,以模拟 HD,从而评估 APN 的影响。通过Y-迷宫和莫里斯水迷宫(MWM)测试检验大鼠的工作记忆和空间记忆。通过逆转录定量聚合酶链反应(RT-qPCR)分析,对大鼠大脑皮层和海马中神经营养素及其受体的 mRNA 水平进行了定量评估。结果表明,三种剂量的 APN 均能显著预防 3-NP 给药诱导的疾病表型。此外,与 3-NP 组相比,APN 治疗可提高 BDNF、TrkA、TrkB 和 CREB 的基因表达水平,同时显著降低 P75 NTR,在大脑皮层和海马中呈现剂量依赖性效应。这些研究结果表明,在 HD 动物模型中,APN 可通过提高神经营养素及其受体水平来缓解 3-NP 引起的记忆缺陷。
{"title":"Alpha-Pinene Ameliorates Memory Deficits in 3-Nitropropionic Acid-Induced Rat Model of Huntington’s Disease","authors":"Paria Hashemi,&nbsp;Mohammad Raman Moloudi,&nbsp;Helia Rahmani,&nbsp;Katayoun Hassanzadeh,&nbsp;Zakaria Vahabzadeh,&nbsp;Esmael Izadpanah","doi":"10.1007/s11064-025-04393-z","DOIUrl":"10.1007/s11064-025-04393-z","url":null,"abstract":"<div><p>Memory impairment is one of the cognitive symptoms in Huntington’s disease (HD) which appears before motor dysfunction in patients. Various molecular mechanisms, including disruptions in neurotrophins levels, are involved in the occurrence of memory problems in HD. Alpha-pinene (APN), a member of the monoterpene family, exhibited beneficial effects in animal models of neurodegenerative disorders. As a result, this study assessed the impact of APN on memory in the 3-nitropropionic acid (3-NP) induced model of HD in rats. Male Wistar rats received saline, 3-NP to model HD, or APN (1, 5, or 10 mg/kg) plus 3-NP for 21 days to assess APN’s effects. Working and spatial memory were examined by the Y-maze and Morris-water-maze (MWM) tests. The mRNA levels of neurotrophins and their receptors in the brain cortex and hippocampus of the rats were quantitatively assessed through Reverse Transcription Quantitative Polymerase Chain Reaction (RT-qPCR) analysis. The results showed that APN, at all three doses, significantly prevented the disease phenotype induced by 3-NP administration. In addition, APN treatment elevated the gene expression levels of BDNF, TrkA, TrkB, and CREB, while significantly decreasing P75 NTR showing a dose-dependent effect in the brain cortex and hippocampus, compared to the 3-NP group. These findings suggest that APN alleviates 3-NP-induced memory deficits by enhancing neurotrophins and their receptor levels in an animal model of HD.</p></div>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":"50 3","pages":""},"PeriodicalIF":3.7,"publicationDate":"2025-04-16","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143835707","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Bupivacaine Reduces the Viability of SH-SY5Y Cells and Promotes Apoptosis by the Inhibition of Akt Signaling Pathway
IF 3.7 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-04-12 DOI: 10.1007/s11064-025-04386-y
Heng Yu, Xiufeng Liu, Juan Liu, Dong Tang

Bupivacaine (BUP) is a commonly used local anesthetic, while SH-SY5Y cells are a human neuroblastoma cell line frequently employed in research on neurotoxicity and neuroprotective mechanisms. To assess the neurotoxic effects of BUP on SH-SY5Y cells and the role of threonine-serine protein kinase B (Akt) signaling in BUP-induced nerve injury. SH-SY5Y cells were divided into three groups: the control group (Control), BUP group, and BUP + SC79 group. Cell viability was assessed using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, the level of reactive oxygen species (ROS) in cells was detected using the dihydroethidium fluorescence probe method, and changes in mitochondrial membrane potential were detected by flow cytometry, while BUP-induced apoptosis was evaluated by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining. The effects of BUP on Bax, Bcl-2, Caspase-3, Caspase-9, Akt and phosphorylated Akt (p-Akt) were analyzed by Western blot (WB). Compared with the control group, the BUP group and the BUP + SC79 group showed significantly reduced cell viability, significantly increased apoptosis, significantly elevated ROS levels, significantly decreased JC-1 polymer/monomer ratio, significantly increased protein levels of Bax, caspase-3, caspase-9, Akt, and p-Akt, and significantly decreased Bcl-2 protein levels (P < 0.05). However, compared with the BUP group, the BUP + SC79 group exhibited significantly increased cell viability (P = 0.022), significantly reduced apoptosis rate (P = 0.017), significantly decreased ROS levels (P = 0.015), significantly increased JC-1 polymer/monomer ratio (P = 0.024), significantly reduced protein levels of Bax, caspase-3, caspase-9, Akt, and p-Akt (P = 0.033, 0.028, 0.030, 0.035, and 0.005, respectively), and significantly increased Bcl-2 protein levels (P = 0.024). BUP can reduce the viability of SH-SY5Y cells and promote apoptosis, which may be related to its inhibitory effect on Akt protein activity.

布比卡因(BUP)是一种常用的局部麻醉剂,而SH-SY5Y细胞是一种人类神经母细胞瘤细胞系,经常用于研究神经毒性和神经保护机制。为了评估 BUP 对 SH-SY5Y 细胞的神经毒性作用以及苏氨酸丝氨酸蛋白激酶 B(Akt)信号在 BUP 诱导的神经损伤中的作用。将 SH-SY5Y 细胞分为三组:对照组(Control)、BUP 组和 BUP + SC79 组。细胞活力用 3-(4,5-二甲基噻唑-2-基)-2,5-二苯基溴化四氮唑(MTT)检测法评估,细胞中活性氧(ROS)水平用二氢乙硫荧光探针法检测、而 BUP 诱导的细胞凋亡则通过末端脱氧核苷酸转移酶 dUTP 缺口标记(TUNEL)染色法进行评估。Western blot(WB)分析了BUP对Bax、Bcl-2、Caspase-3、Caspase-9、Akt和磷酸化Akt(p-Akt)的影响。与对照组相比,BUP 组和 BUP + SC79 组的细胞活力显著降低,细胞凋亡显著增加,ROS 水平显著升高,JC-1 聚合体/单体比显著降低,Bax、Caspase-3、Caspase-9、Akt 和 p-Akt 蛋白水平显著升高,Bcl-2 蛋白水平显著降低(P < 0.05)。然而,与 BUP 组相比,BUP + SC79 组表现出细胞活力明显提高(P = 0.022)、凋亡率明显降低(P = 0.017)、ROS 水平明显降低(P = 0.015),JC-1 聚合体/单体比值明显增加(P = 0.024),Bax、caspase-3、caspase-9、Akt 和 p-Akt 蛋白水平明显降低(分别为 P = 0.033、0.028、0.030、0.035 和 0.005),Bcl-2 蛋白水平明显增加(P = 0.024)。BUP能降低SH-SY5Y细胞的活力,促进细胞凋亡,这可能与其抑制Akt蛋白活性的作用有关。
{"title":"Bupivacaine Reduces the Viability of SH-SY5Y Cells and Promotes Apoptosis by the Inhibition of Akt Signaling Pathway","authors":"Heng Yu,&nbsp;Xiufeng Liu,&nbsp;Juan Liu,&nbsp;Dong Tang","doi":"10.1007/s11064-025-04386-y","DOIUrl":"10.1007/s11064-025-04386-y","url":null,"abstract":"<div><p>Bupivacaine (BUP) is a commonly used local anesthetic, while SH-SY5Y cells are a human neuroblastoma cell line frequently employed in research on neurotoxicity and neuroprotective mechanisms. To assess the neurotoxic effects of BUP on SH-SY5Y cells and the role of threonine-serine protein kinase B (Akt) signaling in BUP-induced nerve injury. SH-SY5Y cells were divided into three groups: the control group (Control), BUP group, and BUP + SC79 group. Cell viability was assessed using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, the level of reactive oxygen species (ROS) in cells was detected using the dihydroethidium fluorescence probe method, and changes in mitochondrial membrane potential were detected by flow cytometry, while BUP-induced apoptosis was evaluated by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining. The effects of BUP on Bax, Bcl-2, Caspase-3, Caspase-9, Akt and phosphorylated Akt (p-Akt) were analyzed by Western blot (WB). Compared with the control group, the BUP group and the BUP + SC79 group showed significantly reduced cell viability, significantly increased apoptosis, significantly elevated ROS levels, significantly decreased JC-1 polymer/monomer ratio, significantly increased protein levels of Bax, caspase-3, caspase-9, Akt, and p-Akt, and significantly decreased Bcl-2 protein levels (<i>P</i> &lt; 0.05). However, compared with the BUP group, the BUP + SC79 group exhibited significantly increased cell viability (<i>P</i> = 0.022), significantly reduced apoptosis rate (<i>P</i> = 0.017), significantly decreased ROS levels (<i>P</i> = 0.015), significantly increased JC-1 polymer/monomer ratio (<i>P</i> = 0.024), significantly reduced protein levels of Bax, caspase-3, caspase-9, Akt, and p-Akt (<i>P</i> = 0.033, 0.028, 0.030, 0.035, and 0.005, respectively), and significantly increased Bcl-2 protein levels (<i>P</i> = 0.024). BUP can reduce the viability of SH-SY5Y cells and promote apoptosis, which may be related to its inhibitory effect on Akt protein activity.</p></div>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":"50 2","pages":""},"PeriodicalIF":3.7,"publicationDate":"2025-04-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143821948","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Neuroinflammation Involving Endothelin-1 and Platelet-Activating Factor Receptors Contributes To Self-Injurious Behaviors Induced by Bay k-8644 in Adolescent Mice
IF 3.7 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-04-12 DOI: 10.1007/s11064-025-04387-x
Ngoc Kim Cuong Tran, Ji Hoon Jeong, Naveen Sharma, Yen Nhi Doan Nguyen, Jung Hoon Park, Khanh Ngan Thi Nguyen, Hoang-Yen Phi Tran, Duy-Khanh Dang, Hyoung-Chun Kim, Eun-Joo Shin

Bay k-8644, an activator of L-type voltage-gated calcium channels, induces self-injurious behaviors in mice. Although previous studies using animal models have suggested the possible implications of neuroinflammation in self-injurious behaviors, this has not yet been elucidated in the context of Bay k-8644-induced self-injurious behaviors. In this study, Bay k-8644 (50 µg, i.c.v.)-induced self-injurious behaviors were accompanied by increased expression of endothelin (ET)-1, platelet-activating factor (PAF) receptors, and Iba-1 in the striatum. Pretreatment with the ET receptor antagonist bosentan (10 mg/kg, i.p.), the PAF receptor antagonist ginkgolide B (10 mg/kg, i.p.), or the microglial activation inhibitor minocycline (40 mg/kg/day for 5 days, i.p.) significantly inhibited Bay k-8644-induced self-injurious behaviors and microglial activation in the striatum. Interestingly, bosentan also suppressed Bay k-8644-induced PAF receptor expression, indicating that ET-1 may act as an upstream modulator of the PAF signaling under these experimental conditions. Bay k-8644-induced ET-1 expression and consequent pro-inflammatory changes were reversed by the protein kinase C (PKC) inhibitor NPC-15,437 and the Ca2+/calmodulin-dependent kinase II (CaMKII) inhibitor KN-93. Moreover, Bay k-8644-induced self-injurious behaviors and microglial activation were significantly potentiated by exogenous ET-1 administration (10 pmol, i.c.v.) or by weak neuroinflammation in the striatum induced by systemic injection of low-dose lipopolysaccharide (LPS; 1 mg/kg, i.p.). Our results suggest that neuroinflammatory changes associated with ET-1/PAF signaling in the striatum contribute to Bay k-8644-induced self-injurious behaviors.

{"title":"Neuroinflammation Involving Endothelin-1 and Platelet-Activating Factor Receptors Contributes To Self-Injurious Behaviors Induced by Bay k-8644 in Adolescent Mice","authors":"Ngoc Kim Cuong Tran,&nbsp;Ji Hoon Jeong,&nbsp;Naveen Sharma,&nbsp;Yen Nhi Doan Nguyen,&nbsp;Jung Hoon Park,&nbsp;Khanh Ngan Thi Nguyen,&nbsp;Hoang-Yen Phi Tran,&nbsp;Duy-Khanh Dang,&nbsp;Hyoung-Chun Kim,&nbsp;Eun-Joo Shin","doi":"10.1007/s11064-025-04387-x","DOIUrl":"10.1007/s11064-025-04387-x","url":null,"abstract":"<div><p>Bay k-8644, an activator of L-type voltage-gated calcium channels, induces self-injurious behaviors in mice. Although previous studies using animal models have suggested the possible implications of neuroinflammation in self-injurious behaviors, this has not yet been elucidated in the context of Bay k-8644-induced self-injurious behaviors. In this study, Bay k-8644 (50 µg, i.c.v.)-induced self-injurious behaviors were accompanied by increased expression of endothelin (ET)-1, platelet-activating factor (PAF) receptors, and Iba-1 in the striatum. Pretreatment with the ET receptor antagonist bosentan (10 mg/kg, i.p.), the PAF receptor antagonist ginkgolide B (10 mg/kg, i.p.), or the microglial activation inhibitor minocycline (40 mg/kg/day for 5 days, i.p.) significantly inhibited Bay k-8644-induced self-injurious behaviors and microglial activation in the striatum. Interestingly, bosentan also suppressed Bay k-8644-induced PAF receptor expression, indicating that ET-1 may act as an upstream modulator of the PAF <i>signaling</i> under these experimental conditions. Bay k-8644-induced ET-1 expression and consequent pro-inflammatory changes were reversed by the protein kinase C (PKC) inhibitor NPC-15,437 and the Ca<sup>2+</sup>/calmodulin-dependent kinase II (CaMKII) inhibitor KN-93. Moreover, Bay k-8644-induced self-injurious behaviors and microglial activation were significantly potentiated by exogenous ET-1 administration (10 pmol, i.c.v.) or by weak neuroinflammation in the striatum induced by systemic injection of low-dose lipopolysaccharide (LPS; 1 mg/kg, i.p.). Our results suggest that neuroinflammatory changes associated with ET-1/PAF signaling in the striatum contribute to Bay k-8644-induced self-injurious behaviors.</p></div>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":"50 2","pages":""},"PeriodicalIF":3.7,"publicationDate":"2025-04-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143821963","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
OAB-14 Attenuated Glymphatic System Disorder, Neuroinflammation and Dyskinesia in Parkinson’s Disease Model Mice Induced by Rotenone
IF 3.7 3区 医学 Q2 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-04-12 DOI: 10.1007/s11064-025-04388-w
Xinyu Zhao, Ruolin Cao, Xiaoyi Tian, Peng Liu, Danyang Liu, Xin Yu, Zhonghui Zheng, Guo-liang Chen, Libo Zou

Parkinson’s Disease (PD) is a neurodegenerative disorder characterized by the pathological accumulation of alpha-synuclein (α-syn) in the neuronal cell bodies of the substantia nigra. The glymphatic system within the Central Nervous System (CNS) is responsible for clearing metabolic waste and abnormal proteins and its dysfunction may significantly contribute to the pathogenesis of PD. Our previous study showed that OAB-14, the novel small molecular compound, showed a great potential effect in APP/PS1 transgenic mice. Given the similarities in the pathogenesis of PD and Alzheimer’s disease (AD), it is pertinent to explore the therapeutic potential of OAB-14 in the context of PD. This study utilized a rotenone-induced PD mice model to evaluate the effects of oral administration of OAB-14, and its underlying mechanisms. Here we confirmed the neuroprotective effect and motor improvement of OAB-14 in rotenone-induced PD model mice. Our research has shown that OAB-14 is capable of enhancing the glymphatic system function by promoting the influx and efflux of the CSF tracers to the brain and deep cervical lymph nodes, respectively, to promote the clearance of α-syn. In addition, OAB-14 could down-regulate MyD88, NF-kB (Ser 536) phosphorylation, and TLR4 to reduce glial cell activation; and down-regulate cleaved-caspase1, NLRP3, ASC, IL-1β, IL-6, IL-18, TNF-α, and IL-10 to reduce the expression of inflammatory vesicles and pro-inflammatory factors, and to reduce neuronal oxidative stress. In summary, OAB-14 may promote the clearance of brain α-syn through the glial lymphatic system, inhibit the α-syn/TLR4/NF-κB/NLRP3 inflammatory pathway, and improve movement disorders.

{"title":"OAB-14 Attenuated Glymphatic System Disorder, Neuroinflammation and Dyskinesia in Parkinson’s Disease Model Mice Induced by Rotenone","authors":"Xinyu Zhao,&nbsp;Ruolin Cao,&nbsp;Xiaoyi Tian,&nbsp;Peng Liu,&nbsp;Danyang Liu,&nbsp;Xin Yu,&nbsp;Zhonghui Zheng,&nbsp;Guo-liang Chen,&nbsp;Libo Zou","doi":"10.1007/s11064-025-04388-w","DOIUrl":"10.1007/s11064-025-04388-w","url":null,"abstract":"<div><p>Parkinson’s Disease (PD) is a neurodegenerative disorder characterized by the pathological accumulation of alpha-synuclein (α-syn) in the neuronal cell bodies of the substantia nigra. The glymphatic system within the Central Nervous System (CNS) is responsible for clearing metabolic waste and abnormal proteins and its dysfunction may significantly contribute to the pathogenesis of PD. Our previous study showed that OAB-14, the novel small molecular compound, showed a great potential effect in APP/PS1 transgenic mice. Given the similarities in the pathogenesis of PD and Alzheimer’s disease (AD), it is pertinent to explore the therapeutic potential of OAB-14 in the context of PD. This study utilized a rotenone-induced PD mice model to evaluate the effects of oral administration of OAB-14, and its underlying mechanisms. Here we confirmed the neuroprotective effect and motor improvement of OAB-14 in rotenone-induced PD model mice. Our research has shown that OAB-14 is capable of enhancing the glymphatic system function by promoting the influx and efflux of the CSF tracers to the brain and deep cervical lymph nodes, respectively, to promote the clearance of α-syn. In addition, OAB-14 could down-regulate MyD88, NF-kB (Ser 536) phosphorylation, and TLR4 to reduce glial cell activation; and down-regulate cleaved-caspase1, NLRP3, ASC, IL-1β, IL-6, IL-18, TNF-α, and IL-10 to reduce the expression of inflammatory vesicles and pro-inflammatory factors, and to reduce neuronal oxidative stress. In summary, OAB-14 may promote the clearance of brain α-syn through the glial lymphatic system, inhibit the α-syn/TLR4/NF-κB/NLRP3 inflammatory pathway, and improve movement disorders.</p></div>","PeriodicalId":719,"journal":{"name":"Neurochemical Research","volume":"50 2","pages":""},"PeriodicalIF":3.7,"publicationDate":"2025-04-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143821949","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Neurochemical Research
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1