Molecular Neurobiology最新文献

筛选
英文 中文
Parkinson's Disease and MicroRNAs: A Duel Between Inhibition and Stimulation of Apoptosis in Neuronal Cells. 帕金森病与 MicroRNAs:抑制和刺激神经元细胞凋亡的对决。
IF 4.6 2区 医学
Molecular Neurobiology Pub Date : 2024-11-01 Epub Date: 2024-03-23 DOI: 10.1007/s12035-024-04111-w
Mohamed J Saadh, Ahmed Faisal, Mohaned Adil, Rahman S Zabibah, Abdurakhmon Mamatkulovich Mamadaliev, Mahmood Jasem Jawad, Fahad Alsaikhan, Bagher Farhood
{"title":"Parkinson's Disease and MicroRNAs: A Duel Between Inhibition and Stimulation of Apoptosis in Neuronal Cells.","authors":"Mohamed J Saadh, Ahmed Faisal, Mohaned Adil, Rahman S Zabibah, Abdurakhmon Mamatkulovich Mamadaliev, Mahmood Jasem Jawad, Fahad Alsaikhan, Bagher Farhood","doi":"10.1007/s12035-024-04111-w","DOIUrl":"10.1007/s12035-024-04111-w","url":null,"abstract":"<p><p>Parkinson's disease (PD) is one of the most prevalent diseases of central nervous system that is caused by degeneration of the substantia nigra's dopamine-producing neurons through apoptosis. Apoptosis is regulated by initiators' and executioners' caspases both in intrinsic and extrinsic pathways, further resulting in neuronal damage. In that context, targeting apoptosis appears as a promising therapeutic approach for treating neurodegenerative diseases. Non-coding RNAs-more especially, microRNAs, or miRNAs-are a promising target for the therapy of neurodegenerative diseases because they are essential for a number of cellular processes, including signaling, apoptosis, cell proliferation, and gene regulation. It is estimated that a substantial portion of coding genes (more than 60%) are regulated by miRNAs. These small regulatory molecules can have wide-reaching consequences on cellular processes like apoptosis, both in terms of intrinsic and extrinsic pathways. Furthermore, it was recommended that a disruption in miRNA expression levels could also result in perturbation of typical apoptosis pathways, which may be a factor in certain diseases like PD. The latest research on miRNAs and their impact on neural cell injury in PD models by regulating the apoptosis pathway is summarized in this review article. Furthermore, the importance of lncRNA/circRNA-miRNA-mRNA network for regulating apoptosis pathways in PD models and treatment is explored. These results can be utilized for developing new strategies in PD treatment.</p>","PeriodicalId":18762,"journal":{"name":"Molecular Neurobiology","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140194182","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Thioredoxin-1 Protects Neurons Through Inhibiting NLRP1-Mediated Neuronal Pyroptosis in Models of Alzheimer's Disease. 硫氧还蛋白-1在阿尔茨海默病模型中通过抑制NLRP1介导的神经元猝死保护神经元
IF 4.6 2区 医学
Molecular Neurobiology Pub Date : 2024-11-01 Epub Date: 2024-07-08 DOI: 10.1007/s12035-024-04341-y
Jinjing Jia, Hongjun Liu, Liyan Sun, Yunfeng Xu, Xiansi Zeng
{"title":"Thioredoxin-1 Protects Neurons Through Inhibiting NLRP1-Mediated Neuronal Pyroptosis in Models of Alzheimer's Disease.","authors":"Jinjing Jia, Hongjun Liu, Liyan Sun, Yunfeng Xu, Xiansi Zeng","doi":"10.1007/s12035-024-04341-y","DOIUrl":"10.1007/s12035-024-04341-y","url":null,"abstract":"<p><p>Alzheimer's disease (AD) is the most common neurodegenerative disease all over the world. In the last decade, accumulating proofs have evidenced that neuroinflammation is intimately implicated in the pathogenesis of AD and activation of NOD-like receptor family pyrin domain-containing 1 (NLRP1) inflammasome can induce neuronal pyroptosis and in turn lead to neuronal loss in AD. Thioredoxin-1 (Trx-1), a multifunctional molecule with anti-inflammation in human tissues, displays crucial neuroprotective roles in AD. Our previous research preliminarily found that Trx-1 inhibition enhanced the expression of NLRP1, caspase-1, and gasdermin D (GSDMD) in Aβ<sub>25-35</sub>-treated PC12 cells. However, it is largely unknown if Trx-1 can inhibit NLRP1-mediated neuronal pyroptosis in AD neurons. In this study, it was verified that the protein levels of NLRP1, caspase-1, and GSDMD were significantly increased in Aβ<sub>25-35</sub>-treated mouse HT22 and primary hippocampal neurons. Suppression of Trx-1 with PX-12, a selective inhibitor of Trx-1, or Trx-1 knockdown further activated NLRP1-mediated neuronal pyroptosis. On the contrary, lentivirus infection-mediated Trx-1 overexpression in differentiated PC12 cells dramatically reversed expression of NLRP1, caspase-1, and GSDMD. Furthermore, Trx-1 overexpression mediated by adeno-associated virus in the hippocampal tissues of APP/PS1 mice likewise attenuated the activation of NLRP1-mediated neuronal pyroptosis, as well as reduced the hippocampal deposition of Aβ and ameliorated the cognitive function of APP/PS1 mice. In conclusion, this article predicates a novel molecular mechanism by which Trx-1 exploits neuroprotection through attenuating NLRP1-mediated neuronal pyroptosis in AD models, suggesting that Trx-1 may be a promising therapeutic target for AD.</p>","PeriodicalId":18762,"journal":{"name":"Molecular Neurobiology","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141555228","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Integrated Proteomics and Protein Co-expression Network Analysis Identifies Novel Epileptogenic Mechanism in Mesial Temporal Lobe Epilepsy. 综合蛋白质组学和蛋白质共表达网络分析发现中颞叶癫痫的新型致痫机制
IF 4.6 2区 医学
Molecular Neurobiology Pub Date : 2024-11-01 Epub Date: 2024-04-30 DOI: 10.1007/s12035-024-04186-5
Arpna Srivastava, Priya Rajput, Manjari Tripathi, Poodipedi Sarat Chandra, Ramesh Doddamani, Mehar Chand Sharma, Sanjeev Lalwani, Jyotirmoy Banerjee, Aparna Banerjee Dixit
{"title":"Integrated Proteomics and Protein Co-expression Network Analysis Identifies Novel Epileptogenic Mechanism in Mesial Temporal Lobe Epilepsy.","authors":"Arpna Srivastava, Priya Rajput, Manjari Tripathi, Poodipedi Sarat Chandra, Ramesh Doddamani, Mehar Chand Sharma, Sanjeev Lalwani, Jyotirmoy Banerjee, Aparna Banerjee Dixit","doi":"10.1007/s12035-024-04186-5","DOIUrl":"10.1007/s12035-024-04186-5","url":null,"abstract":"<p><p>Over 50 million people worldwide are affected by epilepsy, a common neurological disorder that has a high rate of drug resistance and diverse comorbidities such as progressive cognitive and behavioural disorders, and increased mortality from direct or indirect effects of seizures and therapies. Despite extensive research with animal models and human studies, limited insights have been gained into the mechanisms underlying seizures and epileptogenesis, which has not translated into significant reductions in drug resistance, morbidities, or mortality. To better understand the molecular signaling networks associated with seizures in MTLE patients, we analyzed the proteome of brain samples from MTLE and control cases using an integrated approach that combines mass spectrometry-based quantitative proteomics, differential expression analysis, and co-expression network analysis. Our analyses of 20 human brain tissues from MTLE patients and 20 controls showed the organization of the brain proteome into a network of 9 biologically meaningful modules of co-expressed proteins. Of these, 6 modules are positively or negatively correlated to MTLE phenotypes with hub proteins that are altered in MTLE patients. Our study is the first to employ an integrated approach of proteomics and protein co-expression network analysis to study patients with MTLE. Our findings reveal a molecular blueprint of altered protein networks in MTLE brain and highlight dysregulated pathways and processes including altered cargo transport, neurotransmitter release from synaptic vesicles, synaptic plasticity, proteostasis, RNA homeostasis, ion transport and transmembrane transport, cytoskeleton disorganization, metabolic and mitochondrial dysfunction, blood micro-particle function, extracellular matrix organization, immune response, neuroinflammation, and cell signaling. These insights into MTLE pathogenesis suggest potential new candidates for future diagnostic and therapeutic development.</p>","PeriodicalId":18762,"journal":{"name":"Molecular Neurobiology","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140868249","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Advancements in Single-Cell RNA Sequencing Research for Neurological Diseases. 神经系统疾病的单细胞 RNA 测序研究进展。
IF 4.6 2区 医学
Molecular Neurobiology Pub Date : 2024-11-01 Epub Date: 2024-04-02 DOI: 10.1007/s12035-024-04126-3
Bingjie Yang, Shuqi Hu, Yiru Jiang, Lei Xu, Song Shu, Hao Zhang
{"title":"Advancements in Single-Cell RNA Sequencing Research for Neurological Diseases.","authors":"Bingjie Yang, Shuqi Hu, Yiru Jiang, Lei Xu, Song Shu, Hao Zhang","doi":"10.1007/s12035-024-04126-3","DOIUrl":"10.1007/s12035-024-04126-3","url":null,"abstract":"<p><p>Neurological diseases are a major cause of the global burden of disease. Although the mechanisms of the occurrence and development of neurological diseases are not fully clear, most of them are associated with cells mediating neuroinflammation. Yet medications and other therapeutic options to improve treatment are still very limited. Single-cell RNA sequencing (scRNA-seq), as a delightfully potent breakthrough technology, not only identifies various cell types and response states but also uncovers cell-specific gene expression changes, gene regulatory networks, intercellular communication, and cellular movement trajectories, among others, in different cell types. In this review, we describe the technology of scRNA-seq in detail and discuss and summarize the application of scRNA-seq in exploring neurological diseases, elaborating the corresponding specific mechanisms of the diseases as well as providing a reliable basis for new therapeutic approaches. Finally, we affirm that scRNA-seq promotes the development of the neuroscience field and enables us to have a deeper cellular understanding of neurological diseases in the future, which provides strong support for the treatment of neurological diseases and the improvement of patients' prognosis.</p>","PeriodicalId":18762,"journal":{"name":"Molecular Neurobiology","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140336234","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
NUS1 Variants Cause Lennox-Gastaut Syndrome Related to Unfolded Protein Reaction Activation. NUS1 变异导致与折叠蛋白反应激活有关的伦诺克斯-加斯豪特综合征。
IF 4.6 2区 医学
Molecular Neurobiology Pub Date : 2024-11-01 Epub Date: 2024-03-23 DOI: 10.1007/s12035-024-04123-6
Nan-Xiang Shen, Xiao-Chong Qu, Jing Yu, Cui-Xia Fan, Fu-Li Min, Ling-Ying Li, Ming-Rui Zhang, Bing-Mei Li, Jie Wang, Na He, Wei-Ping Liao, Yi-Wu Shi, Wen-Bin Li
{"title":"NUS1 Variants Cause Lennox-Gastaut Syndrome Related to Unfolded Protein Reaction Activation.","authors":"Nan-Xiang Shen, Xiao-Chong Qu, Jing Yu, Cui-Xia Fan, Fu-Li Min, Ling-Ying Li, Ming-Rui Zhang, Bing-Mei Li, Jie Wang, Na He, Wei-Ping Liao, Yi-Wu Shi, Wen-Bin Li","doi":"10.1007/s12035-024-04123-6","DOIUrl":"10.1007/s12035-024-04123-6","url":null,"abstract":"<p><p>NUS1 encodes the Nogo-B receptor, a critical regulator for unfolded protein reaction (UPR) signaling. Although several loss-of-function variants of NUS1 have been identified in patients with developmental and epileptic encephalopathy (DEE), the role of the NUS1 variant in Lennox-Gastaut syndrome (LGS), a severe child-onset DEE, remains unknown. In this study, we identified two de novo variants of NUS1, a missense variant (c.868 C > T/p.R290C) and a splice site variant (c.792-2 A > G), in two unrelated LGS patients using trio-based whole-exome sequencing performed in a cohort of 165 LGS patients. Both variants were absent in the gnomAD population and showed a significantly higher observed number of variants than expected genome-wide. The R290C variant was predicted to damage NUS1 and decrease its protein stability. The c.792-2 A > G variant caused premature termination of the protein. Knockdown of NUS1 activated the UPR pathway, resulting in apoptosis of HEK293T cells. Supplementing cells with expression of wild-type NUS1, but not the mutant (R290C), rescued UPR activation and apoptosis in NUS1 knockdown cells. Compared to wild-type Drosophila, seizure-like behaviors and excitability in projection neurons were significantly increased in Tango14 (homolog of human NUS1) knockdown and Tango14<sup>R290C/+</sup> knock-in Drosophila. Additionally, abnormal development and a small body size were observed in both mutants. Activated UPR signaling was also detected in both mutants. Thus, NUS1 is a causative gene for LGS with dominant inheritance. The pathogenicity of these variants is related to the UPR signaling activation, which may be a common pathogenic mechanism of DEE.</p>","PeriodicalId":18762,"journal":{"name":"Molecular Neurobiology","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140194181","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
ACE2 Rescues Sepsis-Associated Encephalopathy by Reducing Inflammation, Oxidative Stress, and Neuronal Apoptosis via the Nrf2/Sestrin2 Signaling Pathway. ACE2 通过 Nrf2/Sestrin2 信号通路减轻炎症、氧化应激和神经元凋亡,从而挽救败血症相关脑病。
IF 4.6 2区 医学
Molecular Neurobiology Pub Date : 2024-11-01 Epub Date: 2024-03-27 DOI: 10.1007/s12035-024-04063-1
Ya Li, Tian-Tian Wan, Jia-Xin Li, Xue Xiao, Lei Liu, Hui-Hua Li, Shu-Bin Guo
{"title":"ACE2 Rescues Sepsis-Associated Encephalopathy by Reducing Inflammation, Oxidative Stress, and Neuronal Apoptosis via the Nrf2/Sestrin2 Signaling Pathway.","authors":"Ya Li, Tian-Tian Wan, Jia-Xin Li, Xue Xiao, Lei Liu, Hui-Hua Li, Shu-Bin Guo","doi":"10.1007/s12035-024-04063-1","DOIUrl":"10.1007/s12035-024-04063-1","url":null,"abstract":"<p><p>Neuroinflammation and oxidative stress contribute to the progression of sepsis-associated encephalopathy (SAE). Angiotensin-converting enzyme 2 (ACE2) is considered to be a neuroprotective factor due to its anti-inflammatory and antioxidant properties. However, the role of ACE2 on myeloid cells in regulating SAE and the underlying mechanism warrants further exploration. SAE was induced in ACE2 transgenic (TG), knockout (KO), and bone marrow (BM) chimeric mice by cecal ligation and puncture (CLP). The expression levels of apoptosis-, oxidation- and neuroinflammation-associated mediators and morphological changes were monitored by quantitative real-time PCR analyses and histological examinations in the cortex of septic mice. The contents of angiotensin (Ang) II and Ang-(1-7) along with the activity of ACE2 were examined with commercial kits. The expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and Sestrin2 was detected by immunoblotting analysis. Our results indicated that the expression of cortical ACE2 was significantly reduced in the early phase of CLP-induced sepsis. Moreover, ACE2 overexpression in TG mice conferred neuroprotection against sepsis, as evidenced by alleviated neuronal apoptosis, oxidative stress, and proinflammatory M1-like microglial polarization, accompanied by upregulation of the Ang-(1-7), Nrf2, and Sestrin2 protein levels. Conversely, ACE2 deficiency in KO mice exacerbated SAE. The neuroprotective effects of ACE2 were further confirmed in wild-type mice transplanted with ACE2-TG and KO BM cells. Therefore, our data suggest that myeloid ACE2 exerts a protective role in the pathogenesis of SAE, potentially by activating Ang-(1-7)-Nrf2/sestrin2 signaling pathway, and highlight that upregulating ACE2 expression and activity may represent a promising approach for the treatment of SAE in patients with sepsis.</p>","PeriodicalId":18762,"journal":{"name":"Molecular Neurobiology","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140293987","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Serotonin Transporter (SLC6A4) and FK506-Binding Protein 5 (FKBP5) Genotype and Methylation Relationships with Response to Meditation in Veterans with PTSD. 创伤后应激障碍退伍军人的羟色胺转运体 (SLC6A4) 和 FK506 结合蛋白 5 (FKBP5) 基因型和甲基化与冥想反应的关系。
IF 4.6 2区 医学
Molecular Neurobiology Pub Date : 2024-11-01 Epub Date: 2024-04-27 DOI: 10.1007/s12035-024-04096-6
Adam Lee, Paul Thuras, Joshua Baller, Chuan Jiao, Bin Guo, Christopher R Erbes, Melissa A Polusny, Chunyu Liu, Baolin Wu, Kelvin O Lim, Jeffrey R Bishop
{"title":"Serotonin Transporter (SLC6A4) and FK506-Binding Protein 5 (FKBP5) Genotype and Methylation Relationships with Response to Meditation in Veterans with PTSD.","authors":"Adam Lee, Paul Thuras, Joshua Baller, Chuan Jiao, Bin Guo, Christopher R Erbes, Melissa A Polusny, Chunyu Liu, Baolin Wu, Kelvin O Lim, Jeffrey R Bishop","doi":"10.1007/s12035-024-04096-6","DOIUrl":"10.1007/s12035-024-04096-6","url":null,"abstract":"<p><p>Meditation-based interventions are novel and effective non-pharmacologic treatments for veterans with PTSD. We examined relationships between treatment response, early life trauma exposure, DNA polymorphisms, and methylation in the serotonin transporter (SLC6A4) and FK506-binding protein 5 (FKBP5) genes. DNA samples and clinical outcomes were examined in 72 veterans with PTSD who received meditation-based therapy in two separate studies of mindfulness-based stress reduction (MBSR) and Transcendental Meditation (TM). The PTSD Checklist was administered to assess symptoms at baseline and after 9 weeks of meditation intervention. We examined the SLC6A4 promoter (5HTTLPR_L/S insertion/deletion + rs25531_A/G) polymorphisms according to previously defined gene expression groups, and the FKBP5 variant rs1360780 previously associated with PTSD disease risk. Methylation for CpG sites of SLC6A4 (28 sites) and FKBP5 (45 sites) genes was quantified in DNA samples collected before and after treatment. The 5HTTLPR L<sub>A</sub>L<sub>A</sub> high expression genotype was associated with greater symptom improvement in participants exposed to early life trauma (p = 0.015). Separately, pre to post-treatment change of DNA methylation in a group of nine FKBP5 CpG sites was associated with greater symptom improvement (OR = 2.8, 95% CI 1.1-7.1, p = 0.027). These findings build on a wealth of existing knowledge regarding epigenetic and genetic relationships with PTSD disease risk to highlight the potential importance of SLC6A4 and FKBP5 for treatment mechanisms and as biomarkers of symptom improvement.</p>","PeriodicalId":18762,"journal":{"name":"Molecular Neurobiology","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140870688","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Gene Expression Analysis in T2DM and Its Associated Microvascular Diabetic Complications: Focus on Risk Factor and RAAS Pathway. T2DM 及其相关微血管糖尿病并发症的基因表达分析:关注风险因素和 RAAS 通路。
IF 4.6 2区 医学
Molecular Neurobiology Pub Date : 2024-11-01 Epub Date: 2024-03-27 DOI: 10.1007/s12035-024-04127-2
Laxmipriya Jena, Prabhsimran Kaur, Tashvinder Singh, Kangan Sharma, Sushil Kotru, Anjana Munshi
{"title":"Gene Expression Analysis in T2DM and Its Associated Microvascular Diabetic Complications: Focus on Risk Factor and RAAS Pathway.","authors":"Laxmipriya Jena, Prabhsimran Kaur, Tashvinder Singh, Kangan Sharma, Sushil Kotru, Anjana Munshi","doi":"10.1007/s12035-024-04127-2","DOIUrl":"10.1007/s12035-024-04127-2","url":null,"abstract":"<p><p>Prolonged hyperglycemic conditions in type 2 diabetes mellitus (T2DM) cause pathological and functional damage to many organs and tissues, including the kidneys, retina, skin, and neuronal tissues, resulting in the development of microvascular diabetic complications. The altered renin angiotensin aldosterone system (RAAS) pathway has been reported to play an important role in the development of insulin resistance in T2DM and associated complications. The current study was carried out to evaluate the association of risk factors and altered expression of RAAS genes in T2DM patients without complications and T2DM patients with complications (retinopathy, nephropathy, and neuropathy). Four hundred and twenty subjects including 140 healthy controls, 140 T2DM patients with diabetic complications, and 140 T2DM patients without diabetic complications were included in the study. Risk factors associated with the development of T2DM and diabetic complications were evaluated. Further, expression analysis of RAAS genes (AGT, ACE, ACE2, and AGT1R) was carried out using qRTPCR in healthy controls, T2DM patients with complications, and T2DM patients without complications. Various risk factors like urban background, higher BMI, alcoholism, smoking, and family history of diabetes among others were found to be associated with the development of T2DM as well as diabetic complications. The expression level of AGT, ACE, and AGT1R was found to be upregulated whereas ACE2 was found to be downregulated in T2DM patients with complications and T2DM patients without complications as compared to controls. Altered expression of the studied genes of RAAS pathway is associated with the development of microvascular diabetic complications.</p>","PeriodicalId":18762,"journal":{"name":"Molecular Neurobiology","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140293988","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Bifenthrin Caused Parkinson's-Like Symptoms Via Mitochondrial Autophagy and Ferroptosis Pathway Stereoselectively in Parkin-/- Mice and C57BL/6 Mice. 联苯菊酯在Parkin-/-小鼠和C57BL/6小鼠中通过线粒体自噬和铁蛋白沉积途径立体选择性地引起帕金森病样症状
IF 4.6 2区 医学
Molecular Neurobiology Pub Date : 2024-11-01 Epub Date: 2024-05-01 DOI: 10.1007/s12035-024-04140-5
Ying Zhang, Baorong Zhang
{"title":"Bifenthrin Caused Parkinson's-Like Symptoms Via Mitochondrial Autophagy and Ferroptosis Pathway Stereoselectively in Parkin<sup>-/-</sup> Mice and C57BL/6 Mice.","authors":"Ying Zhang, Baorong Zhang","doi":"10.1007/s12035-024-04140-5","DOIUrl":"10.1007/s12035-024-04140-5","url":null,"abstract":"<p><p>It has been proposed that pyrethroid exposure contributes to the increasing prevalence of neurodegenerative diseases. However, the potential mechanisms remain unclear. The current study aimed to investigate the effects of the widely used pyrethroid bifenthrin on Parkinson's disease (PD) risk. Bifenthrin (1S-cis-bifenthrin, 1R-cis-bifenthrin, raceme) was administered to male Parkin<sup>-/-</sup> mice and C57BL/6 mice by oral gavage at a dose of 10 mg/kg bw/day for 28 days. Bifenthrin exposure significantly increased the time of pole climbing and decreased the period of rotarod running, indicating that bifenthrin decreased motor coordination in Parkin<sup>-/-</sup> mice, which was more evident by 1S-cis-bifenthrin. Furthermore, administration of bifenthrin induced obvious decreases in tyrosine hydroxylase (TH)<sup>+</sup> cell count and the protein expression of TH. Increased protein of mitochondrial autophagy LC3B and p62 was observed after exposure to bifenthrin. Increased iron deposition and protein expression of iron transport transferrin (Tf) and transferrin receptor 2 (TfR2) was detected. 1S-cis-bifenthrin bound with Tf, TfR2, and GPX4 with lower binding energies than 1R-cis-bifenthrin, resulting in stronger interactions with these proteins. These results show structure-dependent PD-like effects of bifenthrin on motor activity and coordination associated with the disturbed mitochondrial autophagy and ferroptosis-related pathway. These data demonstrate that pyrethroid exposure increases the potential of Parkinson's-like symptoms via the ferroptosis pathway in Parkin<sup>-/-</sup> mice that is more pronounced than in C57BL/6 mice, providing a prospective enantioselective toxic effect of environmental neurotoxins on PD risk.</p>","PeriodicalId":18762,"journal":{"name":"Molecular Neurobiology","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140865245","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
Roles of KCNA2 in Neurological Diseases: from Physiology to Pathology. KCNA2 在神经系统疾病中的作用:从生理学到病理学。
IF 4.6 2区 医学
Molecular Neurobiology Pub Date : 2024-11-01 Epub Date: 2024-03-22 DOI: 10.1007/s12035-024-04120-9
Changning Xie, Miriam Kessi, Fei Yin, Jing Peng
{"title":"Roles of KCNA2 in Neurological Diseases: from Physiology to Pathology.","authors":"Changning Xie, Miriam Kessi, Fei Yin, Jing Peng","doi":"10.1007/s12035-024-04120-9","DOIUrl":"10.1007/s12035-024-04120-9","url":null,"abstract":"<p><p>Potassium voltage-gated channel subfamily a member 2 (Kv1.2, encoded by KCNA2) is highly expressed in the central and peripheral nervous systems. Based on the patch clamp studies, gain-of function (GOF), loss-of-function (LOF), and a mixed type (GOF/LOF) variants can cause different conditions/disorders. KCNA2-related neurological diseases include epilepsy, intellectual disability (ID), attention deficit/hyperactive disorder (ADHD), autism spectrum disorder (ASD), pain as well as autoimmune and movement disorders. Currently, the molecular mechanisms for the reported variants in causing diverse disorders are unknown. Consequently, this review brings up to date the related information regarding the structure and function of Kv1.2 channel, expression patterns, neuronal localizations, and tetramerization as well as important cell and animal models. In addition, it provides updates on human genetic variants, genotype-phenotype correlations especially highlighting the deep insight into clinical prognosis of KCNA2-related developmental and epileptic encephalopathy, mechanisms, and the potential treatment targets for all KCNA2-related neurological disorders.</p>","PeriodicalId":18762,"journal":{"name":"Molecular Neurobiology","volume":null,"pages":null},"PeriodicalIF":4.6,"publicationDate":"2024-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140189952","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"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
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
确定
请完成安全验证×
相关产品
×
本文献相关产品
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信