Intrinsically disordered regions (IDRs) in proteins can undergo liquid-liquid phase separation (LLPS) for functional assembly, but this increases the chance of forming disease-associated amyloid fibrils. Not all amyloid fibrils form through LLPS however, and the importance of LLPS relative to other pathways in fibril formation remains unclear. We investigated this question in TDP-43, a motor neuron disease and dementia-causing protein that undergoes LLPS, using thioflavin T (ThT) fluorescence, NMR, transmission electron microscopy (TEM), and wide-angle X-ray scattering (WAXS) experiments. Using a fluorescence probe modified from ThT strategically designed for targeting protein assembly rather than β-sheets and supported by TEM images, we propose that the biphasic ThT signals observed under LLPS-favoring conditions are due to the presence of amorphous aggregates. These aggregates represent an intermediate state that diverges from the direct pathway to β-sheet-dominant fibrils. Under non-LLPS conditions in contrast (at low pH or at physiological conditions in a construct with key LLPS residues removed), the protein forms a hydrogel. Real-time WAXS data, ThT signals, and TEM images collectively demonstrate that the gelation process circumvents LLPS and yet still results in the formation of fibril-like structural networks. We suggest that the IDR of TDP-43 forms disease-causing amyloid fibrils regardless of the formation pathway. Our findings shed light on why both LLPS-promoting and LLPS-inhibiting mutants are found in TDP-43-related diseases.
蛋白质中的内在无序区(IDR)可以通过液-液相分离(LLPS)进行功能组装,但这会增加形成与疾病相关的淀粉样纤维的几率。然而,并非所有淀粉样纤维都是通过液-液相分离形成的,而且液-液相分离相对于其他途径在纤维形成中的重要性仍不清楚。我们利用硫黄素 T(ThT)荧光、核磁共振、透射电子显微镜(TEM)和广角 X 射线散射(WAXS)实验对 TDP-43 这一运动神经元疾病和痴呆症致病蛋白进行了研究。我们利用一种由 ThT 改良的荧光探针,以蛋白质组装而非β-片层为目标进行战略性设计,并辅以 TEM 图像,提出在有利于 LLPS 的条件下观察到的双相 ThT 信号是由于无定形聚集体的存在。这些聚集体代表了一种中间状态,与β片状纤维的直接路径不同。与此相反,在非 LLPS 条件下(低 pH 值或生理条件下去除关键 LLPS 残基的构建体),蛋白质会形成水凝胶。实时 WAXS 数据、ThT 信号和 TEM 图像共同证明,凝胶化过程绕过了 LLPS,但仍能形成纤维状结构网络。我们认为,TDP-43的IDR会形成致病的淀粉样纤维,而与形成途径无关。我们的发现揭示了为什么在TDP-43相关疾病中同时发现了促进LLPS和抑制LLPS的突变体。
{"title":"TDP-43 Amyloid Fibril Formation via Phase Separation-Related and -Unrelated Pathways.","authors":"Pin-Han Lin, Guan-Wei Wu, Yu-Hao Lin, Jing-Rou Huang, U-Ser Jeng, Wei-Min Liu, Jie-Rong Huang","doi":"10.1021/acschemneuro.4c00503","DOIUrl":"10.1021/acschemneuro.4c00503","url":null,"abstract":"<p><p>Intrinsically disordered regions (IDRs) in proteins can undergo liquid-liquid phase separation (LLPS) for functional assembly, but this increases the chance of forming disease-associated amyloid fibrils. Not all amyloid fibrils form through LLPS however, and the importance of LLPS relative to other pathways in fibril formation remains unclear. We investigated this question in TDP-43, a motor neuron disease and dementia-causing protein that undergoes LLPS, using thioflavin T (ThT) fluorescence, NMR, transmission electron microscopy (TEM), and wide-angle X-ray scattering (WAXS) experiments. Using a fluorescence probe modified from ThT strategically designed for targeting protein assembly rather than β-sheets and supported by TEM images, we propose that the biphasic ThT signals observed under LLPS-favoring conditions are due to the presence of amorphous aggregates. These aggregates represent an intermediate state that diverges from the direct pathway to β-sheet-dominant fibrils. Under non-LLPS conditions in contrast (at low pH or at physiological conditions in a construct with key LLPS residues removed), the protein forms a hydrogel. Real-time WAXS data, ThT signals, and TEM images collectively demonstrate that the gelation process circumvents LLPS and yet still results in the formation of fibril-like structural networks. We suggest that the IDR of TDP-43 forms disease-causing amyloid fibrils regardless of the formation pathway. Our findings shed light on why both LLPS-promoting and LLPS-inhibiting mutants are found in TDP-43-related diseases.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2024-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11488477/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142363399","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}
Pub Date : 2024-10-02Epub Date: 2024-09-20DOI: 10.1021/acschemneuro.4c00453
Victor L B França, Eveline M Bezerra, Roner F da Costa, Hernandes F Carvalho, Valder N Freire, Geanne Matos
Seven treatments are approved for Alzheimer's disease, but five of them only relieve symptoms and do not alter the course of the disease. Aducanumab (Adu) and lecanemab are novel disease-modifying antiamyloid-β (Aβ) human monoclonal antibodies that specifically target the pathophysiology of Alzheimer's disease (AD) and were recently approved for its treatment. However, their administration is associated with serious side effects, and their use is limited to early stages of the disease. Therefore, drug discovery remains of great importance in AD research. To gain new insights into the development of novel drugs for Alzheimer's disease, a combination of techniques was employed, including mutation screening, molecular dynamics, and quantum biochemistry. These were used to outline the interfacial interactions of the Aducanumab::Aβ2-7 complex. Our analysis identified critical stabilizing contacts, revealing up to 40% variation in the affinity of the Adu chains for Aβ2-7 depending on the conformation outlined. Remarkably, two complementarity determining regions (CDRs) of the Adu heavy chain (HCDR3 and HCDR2) and one CDR of the Adu light chain (LCDR3) accounted for approximately 77% of the affinity of Adu for Aβ2-7, confirming their critical role in epitope recognition. A single mutation, originally reported to have the potential to increase the affinity of Adu for Aβ2-7, was shown to decrease its structural stability without increasing the overall binding affinity. Mimetic peptides that have the potential to inhibit Aβ aggregation were designed by using computational outcomes. Our results support the use of these peptides as promising drugs with great potential as inhibitors of Aβ aggregation.
{"title":"Alzheimer's Disease Immunotherapy and Mimetic Peptide Design for Drug Development: Mutation Screening, Molecular Dynamics, and a Quantum Biochemistry Approach Focusing on Aducanumab::Aβ2-7 Binding Affinity.","authors":"Victor L B França, Eveline M Bezerra, Roner F da Costa, Hernandes F Carvalho, Valder N Freire, Geanne Matos","doi":"10.1021/acschemneuro.4c00453","DOIUrl":"10.1021/acschemneuro.4c00453","url":null,"abstract":"<p><p>Seven treatments are approved for Alzheimer's disease, but five of them only relieve symptoms and do not alter the course of the disease. Aducanumab (Adu) and lecanemab are novel disease-modifying antiamyloid-β (Aβ) human monoclonal antibodies that specifically target the pathophysiology of Alzheimer's disease (AD) and were recently approved for its treatment. However, their administration is associated with serious side effects, and their use is limited to early stages of the disease. Therefore, drug discovery remains of great importance in AD research. To gain new insights into the development of novel drugs for Alzheimer's disease, a combination of techniques was employed, including mutation screening, molecular dynamics, and quantum biochemistry. These were used to outline the interfacial interactions of the Aducanumab::Aβ<sub>2-7</sub> complex. Our analysis identified critical stabilizing contacts, revealing up to 40% variation in the affinity of the Adu chains for Aβ<sub>2-7</sub> depending on the conformation outlined. Remarkably, two complementarity determining regions (CDRs) of the Adu heavy chain (HCDR3 and HCDR2) and one CDR of the Adu light chain (LCDR3) accounted for approximately 77% of the affinity of Adu for Aβ<sub>2-7</sub>, confirming their critical role in epitope recognition. A single mutation, originally reported to have the potential to increase the affinity of Adu for Aβ<sub>2-7</sub>, was shown to decrease its structural stability without increasing the overall binding affinity. Mimetic peptides that have the potential to inhibit Aβ aggregation were designed by using computational outcomes. Our results support the use of these peptides as promising drugs with great potential as inhibitors of Aβ aggregation.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":" ","pages":"3543-3562"},"PeriodicalIF":4.1,"publicationDate":"2024-10-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11450751/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142277140","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}
Pub Date : 2024-10-01DOI: 10.1021/acschemneuro.4c00530
Kristof Pota, Shrikant Nilewar, Christina Mantsorov, Lindsay Zumwalt, Nam Nguyen, Cameron J Bowers, David M Freire, Robert B Benafield, Giridhar R Akkaraju, Kayla N Green
Numerous small molecules have been studied for their ability to counteract oxidative stress, a key contributor to neurodegenerative diseases such as Alzheimer's. Despite these efforts, the pharmacological properties and structure-activity relationships of these compounds remain insufficiently understood, yet they are critical in evaluating a drug molecule's therapeutic potential. A modified tetra-aza macrocycle has demonstrated strong antioxidant activity through various mechanisms; however, its limited permeability presents challenges for advanced formulation studies. To enhance permeability while preserving the beneficial reactivity of the parent molecule, two synthetic modifications involving indole functionality were explored and compared to modifications using methyl groups alone. New synthetic strategies were developed to produce the indole-containing molecules, which were characterized by 1D/2D NMR techniques. Isoelectric points, metal binding, and radical scavenging activity were determined to validate that the reactivity of the parent molecules was retained. The permeability of all molecules explored was improved. Protection against oxidative stress through activation of the Nrf2 pathway was demonstrated for molecules containing indoles in cellular models by measuring ROS levels upon treatment and mRNA levels of HO-1 and Nrf2. In contrast, no protection or Nrf2 activation was observed with the methylation of the O- or N atom. These results suggest that while alkylation improves permeability overall, concomitant antioxidant protection and positive permeability are achieved with the indole congeners alone.
{"title":"Impact of Indole Inclusion in the Design of Multi-Tactical Metal-Binding Tetra-Aza Macrocycles that Target the Molecular Features of Neurodegeneration.","authors":"Kristof Pota, Shrikant Nilewar, Christina Mantsorov, Lindsay Zumwalt, Nam Nguyen, Cameron J Bowers, David M Freire, Robert B Benafield, Giridhar R Akkaraju, Kayla N Green","doi":"10.1021/acschemneuro.4c00530","DOIUrl":"https://doi.org/10.1021/acschemneuro.4c00530","url":null,"abstract":"<p><p>Numerous small molecules have been studied for their ability to counteract oxidative stress, a key contributor to neurodegenerative diseases such as Alzheimer's. Despite these efforts, the pharmacological properties and structure-activity relationships of these compounds remain insufficiently understood, yet they are critical in evaluating a drug molecule's therapeutic potential. A modified tetra-aza macrocycle has demonstrated strong antioxidant activity through various mechanisms; however, its limited permeability presents challenges for advanced formulation studies. To enhance permeability while preserving the beneficial reactivity of the parent molecule, two synthetic modifications involving indole functionality were explored and compared to modifications using methyl groups alone. New synthetic strategies were developed to produce the indole-containing molecules, which were characterized by 1D/2D NMR techniques. Isoelectric points, metal binding, and radical scavenging activity were determined to validate that the reactivity of the parent molecules was retained. The permeability of all molecules explored was improved. Protection against oxidative stress through activation of the Nrf2 pathway was demonstrated for molecules containing indoles in cellular models by measuring ROS levels upon treatment and mRNA levels of HO-1 and Nrf2. In contrast, no protection or Nrf2 activation was observed with the methylation of the O- or N atom. These results suggest that while alkylation improves permeability overall, concomitant antioxidant protection and positive permeability are achieved with the indole congeners alone.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2024-10-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142337194","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}
Pub Date : 2024-09-30DOI: 10.1021/acschemneuro.4c00313
Nevine Fathy, Merna A Labib, Reham M Essam, Noha A El-Boghdady
Major depressive disorder (MDD) is considered a major cause of suicide worldwide. As previous studies revealed that neuroinflammation is a significant factor in the etiology of MDD, this study proposed to unravel the possible antidepressant effect of Empagliflozin (EMPA) through targeting miRNA-134 (miR-134)/brain-derived neurotrophic factor (BDNF) and liver kinase B1 (LKB1)/adenosine 5'-monophosphate-activated protein kinase (AMPK)/silent information regulator 1 (SIRT1) axes in ovariectomized (OVX) female rats. Rats were assigned randomly to four groups: Sham operation (SO), OVX, OVX + EMPA (10 mg/kg/day, p.o.), and OVX + EMPA + Dorsomorphin (DORSO) (25 μg/day/rat, i.v.). Drugs were administered for 28 days after 2 weeks of surgery. EMPA debilitated OVX-induced depressive-like behavior by mitigating the immobility time in the tail suspension test and forced swimming test. Moreover, EMPA curtailed OVX-induced alterations of serum estradiol, hippocampal serotonin, miR-134 expression, as well as BDNF. EMPA also dwindled OVX-induced changes of hippocampal p-LKB1/LKB1, p-AMPK/AMPK, SIRT1, and inflammatory markers (nuclear factor-kappa-B, interleukin-1 beta, interleukin-6, and tumor necrosis factor alpha). Additionally, the EMPA-treated group exhibited marked improvement in different brain regions' histopathology. However, DORSO coadministration reversed most of EMPA's beneficial effects. The current study displayed the modulatory role of EMPA on miR-134/BDNF and LKB1/AMPK/SIRT1 axes, thus offering a partial explanation of its antidepressant efficacy and proposing EMPA as a novel therapeutic avenue for MDD.
{"title":"The Interplay between MiR-134/BDNF and LKB1/AMPK/SIRT1 Accentuates the Antidepressant Efficacy of Empagliflozin in Ovariectomized Rats.","authors":"Nevine Fathy, Merna A Labib, Reham M Essam, Noha A El-Boghdady","doi":"10.1021/acschemneuro.4c00313","DOIUrl":"https://doi.org/10.1021/acschemneuro.4c00313","url":null,"abstract":"<p><p>Major depressive disorder (MDD) is considered a major cause of suicide worldwide. As previous studies revealed that neuroinflammation is a significant factor in the etiology of MDD, this study proposed to unravel the possible antidepressant effect of Empagliflozin (EMPA) through targeting miRNA-134 (miR-134)/brain-derived neurotrophic factor (BDNF) and liver kinase B1 (LKB1)/adenosine 5'-monophosphate-activated protein kinase (AMPK)/silent information regulator 1 (SIRT1) axes in ovariectomized (OVX) female rats. Rats were assigned randomly to four groups: Sham operation (SO), OVX, OVX + EMPA (10 mg/kg/day, p.o.), and OVX + EMPA + Dorsomorphin (DORSO) (25 μg/day/rat, i.v.). Drugs were administered for 28 days after 2 weeks of surgery. EMPA debilitated OVX-induced depressive-like behavior by mitigating the immobility time in the tail suspension test and forced swimming test. Moreover, EMPA curtailed OVX-induced alterations of serum estradiol, hippocampal serotonin, miR-134 expression, as well as BDNF. EMPA also dwindled OVX-induced changes of hippocampal p-LKB1/LKB1, p-AMPK/AMPK, SIRT1, and inflammatory markers (nuclear factor-kappa-B, interleukin-1 beta, interleukin-6, and tumor necrosis factor alpha). Additionally, the EMPA-treated group exhibited marked improvement in different brain regions' histopathology. However, DORSO coadministration reversed most of EMPA's beneficial effects. The current study displayed the modulatory role of EMPA on miR-134/BDNF and LKB1/AMPK/SIRT1 axes, thus offering a partial explanation of its antidepressant efficacy and proposing EMPA as a novel therapeutic avenue for MDD.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2024-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142337195","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}
Caffeoylquinic acids (CQAs) and feruloylquinic acids (FQAs), as cinnamoylquinic acids, have neurogenesis promotion effects. We studied for the first time the neurogenesis-enhancing effect of 3,4,5-tri-feruloylquinic acid (TFQA) compared to 3,4,5-tri-caffeoylquinic acid (TCQA), which has a similar structure, and explored their different cellular and molecular mechanisms in neural stem cells (NSCs) of mice brains. After 2 weeks of incubation, we first assessed the number and size of NSCs in TCQA and TFQA treatments. In NSCs treated for TCQA and TFQA, the NSC proliferation gene expression as well as neuronal and glial cell differentiation gene expressions improved. In the microarray assay, the erythroblastic oncogene B (ErbB) signaling pathway, as the common signaling of TCQA and TFQA treatments, was focused on and discussed. In our study, TCQA and TFQA treatments in NSCs showed a significant performance on improving synapse growth and neurogenesis compared with no treatment of NSCs. The two treatments in NSCs also had a significant activation of the ErbB signaling pathway, protein kinase B (AKT), and mitogen-activated protein kinase (MAPK) kinases. In particular, the TCQA-expressed proliferation gene myelocytomatosis oncogene (Myc) had the greatest connections significantly. TFQA treatment remarkably regulated the differentiation gene jun proto-oncogene (Jun), which was the gene with greatest direct relations, while Myc was also induced in TFQA treatment. In the overall quantitative real-time polymerase chain reaction (PCR) assay, TFQA had more outstanding neural proliferation and differentiation capabilities than TCQA in NSCs. Our study suggests that TFQA has greater therapeutic potential in neurogenesis promotion and neurodegenerative diseases compared with TCQA.
{"title":"Comparison of Neurogenesis Promotion Effects between Cinnamoylquinic Acids in Neural Stem Cells from Adult Mice Brains.","authors":"Hongyu Lin, Kazunori Sasaki, Farhana Ferdousi, Hiroko Isoda","doi":"10.1021/acschemneuro.4c00329","DOIUrl":"10.1021/acschemneuro.4c00329","url":null,"abstract":"<p><p>Caffeoylquinic acids (CQAs) and feruloylquinic acids (FQAs), as cinnamoylquinic acids, have neurogenesis promotion effects. We studied for the first time the neurogenesis-enhancing effect of 3,4,5-tri-feruloylquinic acid (TFQA) compared to 3,4,5-tri-caffeoylquinic acid (TCQA), which has a similar structure, and explored their different cellular and molecular mechanisms in neural stem cells (NSCs) of mice brains. After 2 weeks of incubation, we first assessed the number and size of NSCs in TCQA and TFQA treatments. In NSCs treated for TCQA and TFQA, the NSC proliferation gene expression as well as neuronal and glial cell differentiation gene expressions improved. In the microarray assay, the erythroblastic oncogene B (ErbB) signaling pathway, as the common signaling of TCQA and TFQA treatments, was focused on and discussed. In our study, TCQA and TFQA treatments in NSCs showed a significant performance on improving synapse growth and neurogenesis compared with no treatment of NSCs. The two treatments in NSCs also had a significant activation of the ErbB signaling pathway, protein kinase B (AKT), and mitogen-activated protein kinase (MAPK) kinases. In particular, the TCQA-expressed proliferation gene myelocytomatosis oncogene (<i>Myc</i>) had the greatest connections significantly. TFQA treatment remarkably regulated the differentiation gene jun proto-oncogene (<i>Jun</i>), which was the gene with greatest direct relations, while <i>Myc</i> was also induced in TFQA treatment. In the overall quantitative real-time polymerase chain reaction (PCR) assay, TFQA had more outstanding neural proliferation and differentiation capabilities than TCQA in NSCs. Our study suggests that TFQA has greater therapeutic potential in neurogenesis promotion and neurodegenerative diseases compared with TCQA.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2024-09-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11487612/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142337259","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}
Chemotherapy-induced neuropathic pain (CINP) presents a significant challenge in cancer treatment, necessitating novel therapeutic approaches. The intricate relationship between CINP and the gut-brain axis indicates a crucial role for the gut microbiota in pain modulation during cancer therapy. In this study, we investigated the effect of gut microbiota and their modulation on CINP in rats. Cisplatin administration (20 mg/kg, ip) disrupted the integrity of the blood-spinal cord barrier, as evidenced by reduced expression of tight junction proteins occludin and claudin-5 and increased leakage of pro-inflammatory cytokines into the spinal cord. Fecal microbiota transplantation (FMT, 0.5 mL of P.O.) from healthy rats over 21 days restored barrier integrity, as confirmed by Evan's blue assay. FMT intervention halted the progression of cisplatin-induced pain, demonstrated through a battery of pain assays assessing mechanical, thermal, and cold allodynia alongside hyperalgesia measurements. Additionally, FMT treatment reduced oxidative stress and modulated neuro-inflammatory markers, resulting in a rebalanced cytokine profile with decreased levels of neuro-inflammatory cytokines (IL-6 and TNFα) and increased expression of the anti-inflammatory cytokine IL-10. Gut microbiota-mediated IL-1β/NF-κB signaling emerged as a critical factor in leukocyte recruitment and microglial activation, highlighting the gut-brain axis as a key regulatory nexus in managing cisplatin-induced neuropathic pain. These findings underscore the therapeutic potential of targeting gut microbiota modulation as a promising strategy for alleviating CINP and improving the well-being of cancer patients undergoing chemotherapy.
化疗诱发的神经性疼痛(CINP)是癌症治疗中的一项重大挑战,需要采用新的治疗方法。CINP 与肠道-大脑轴之间错综复杂的关系表明,肠道微生物群在癌症治疗期间的疼痛调节中起着至关重要的作用。在这项研究中,我们调查了肠道微生物群及其调节对大鼠 CINP 的影响。顺铂给药(20 毫克/千克,ip)破坏了血液-脊髓屏障的完整性,表现为紧密连接蛋白occludin和claudin-5的表达减少以及促炎细胞因子渗漏到脊髓的增加。健康大鼠的粪便微生物群移植(FMT,0.5 mL P.O.)可在 21 天内恢复屏障的完整性,埃文氏蓝检测法证实了这一点。FMT 的干预阻止了顺铂诱导的疼痛的发展,这一点通过一系列疼痛试验得到了证实,这些试验评估了机械痛、热痛和冷痛以及痛觉减退。此外,FMT疗法还能降低氧化应激,调节神经炎症标志物,从而重新平衡细胞因子谱,降低神经炎症细胞因子(IL-6和TNFα)的水平,增加抗炎细胞因子IL-10的表达。肠道微生物群介导的IL-1β/NF-κB信号转导是白细胞招募和小胶质细胞活化的关键因素,这突出表明肠道-大脑轴是控制顺铂诱导的神经性疼痛的关键调控纽带。这些发现凸显了以肠道微生物群调节为目标的治疗潜力,它是减轻顺铂诱导的神经病理性疼痛和改善化疗中癌症患者健康状况的一种有前途的策略。
{"title":"Fecal Microbiota Transplantation-Mediated Rebalancing of the Gut-Brain Axis Alleviates Cisplatin-Induced Neuropathic Pain.","authors":"Mousmi Rani, Akhilesh, Deepak Chouhan, Ankit Uniyal, Vinod Tiwari","doi":"10.1021/acschemneuro.4c00267","DOIUrl":"https://doi.org/10.1021/acschemneuro.4c00267","url":null,"abstract":"<p><p>Chemotherapy-induced neuropathic pain (CINP) presents a significant challenge in cancer treatment, necessitating novel therapeutic approaches. The intricate relationship between CINP and the gut-brain axis indicates a crucial role for the gut microbiota in pain modulation during cancer therapy. In this study, we investigated the effect of gut microbiota and their modulation on CINP in rats. Cisplatin administration (20 mg/kg, ip) disrupted the integrity of the blood-spinal cord barrier, as evidenced by reduced expression of tight junction proteins occludin and claudin-5 and increased leakage of pro-inflammatory cytokines into the spinal cord. Fecal microbiota transplantation (FMT, 0.5 mL of P.O.) from healthy rats over 21 days restored barrier integrity, as confirmed by Evan's blue assay. FMT intervention halted the progression of cisplatin-induced pain, demonstrated through a battery of pain assays assessing mechanical, thermal, and cold allodynia alongside hyperalgesia measurements. Additionally, FMT treatment reduced oxidative stress and modulated neuro-inflammatory markers, resulting in a rebalanced cytokine profile with decreased levels of neuro-inflammatory cytokines (IL-6 and TNFα) and increased expression of the anti-inflammatory cytokine IL-10. Gut microbiota-mediated IL-1β/NF-κB signaling emerged as a critical factor in leukocyte recruitment and microglial activation, highlighting the gut-brain axis as a key regulatory nexus in managing cisplatin-induced neuropathic pain. These findings underscore the therapeutic potential of targeting gut microbiota modulation as a promising strategy for alleviating CINP and improving the well-being of cancer patients undergoing chemotherapy.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2024-09-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142337262","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}
Pub Date : 2024-09-27DOI: 10.1021/acschemneuro.4c00507
Dishank Patel, Ritu Soni, Jigna Shah
Parkinson's disease (PD) is the second most prevailing degenerative disease that deals with dopaminergic neuronal loss and deficiency of dopamine in SNpc and striatum. Manifestations primarily include motor symptoms like tremor, rigidity, and akinesia/dyskinesia along with some nonmotor symptoms like GI and olfactory dysfunction. α-Synuclein pathogenesis is the major cause behind progression of PD; however there are many underlying molecular mechanisms behind the pathophysiology of PD. Sirtuins are small molecular deacetylases that have an imperative role in pathology of such neurodegenerative disorders like PD. Sirtuins are majorly classified according to their location; nuclear (SIRT1,7,6), mitochondrial sirtuins (SIRT3-5), and cytosolic (SIRT2). These actively take part in pathological development and possess independent actions. In this review, the role of nuclear sirtuins is individualistically explored for better understanding of PD pathology and development of advanced therapeutics targeting sirtuins.
{"title":"Decoding the Role of Nuclear Sirtuins in Parkinson's Pathogenesis.","authors":"Dishank Patel, Ritu Soni, Jigna Shah","doi":"10.1021/acschemneuro.4c00507","DOIUrl":"https://doi.org/10.1021/acschemneuro.4c00507","url":null,"abstract":"<p><p>Parkinson's disease (PD) is the second most prevailing degenerative disease that deals with dopaminergic neuronal loss and deficiency of dopamine in SNpc and striatum. Manifestations primarily include motor symptoms like tremor, rigidity, and akinesia/dyskinesia along with some nonmotor symptoms like GI and olfactory dysfunction. α-Synuclein pathogenesis is the major cause behind progression of PD; however there are many underlying molecular mechanisms behind the pathophysiology of PD. Sirtuins are small molecular deacetylases that have an imperative role in pathology of such neurodegenerative disorders like PD. Sirtuins are majorly classified according to their location; nuclear (SIRT1,7,6), mitochondrial sirtuins (SIRT3-5), and cytosolic (SIRT2). These actively take part in pathological development and possess independent actions. In this review, the role of nuclear sirtuins is individualistically explored for better understanding of PD pathology and development of advanced therapeutics targeting sirtuins.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2024-09-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142337260","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}
Pub Date : 2024-09-26DOI: 10.1021/acschemneuro.4c00411
Xidong Pan, Zhifeng Lei, Jiang Chen, Congcong Jia, Jie Deng, Ying Liu, Xingmei Luo, Likun Wang, Dan Zi, Zhen Wang, Song Li, Jun Tan
While amyloidopathy and tauopathy have been recognized as hallmarks in Alzheimer's disease (AD) brain, recently, increasing lines of evidence have supported the pathological roles of cerebrovascular changes in the pathogenesis and progression of AD. Restoring or ameliorating the impaired cerebrovascular function during the early phase of the disease may yield benefits against the cognitive decline in AD. In the present study, we evaluated the potential therapeutic effects of nicergoline [NG, a well-known α1 adrenergic receptor (ADR) blocker and vasodilator] against AD through ameliorating vascular abnormalities. Our in vitro data revealed that NG could reverse β-amyloid1-42 (Aβ1-42)-induced PKC/ERK1/2 activation, the downstream pathway of α1-ADR activation, in α1-ADR-overexpressed N2a cells. NG also blocked Aβ1-42- or phenylephrine-induced constrictions in isolated rat arteries. All these in vitro data may suggest ADR-dependent impacts of Aβ on vascular function and the reversal effect of NG. In addition, the ameliorating impacts of NG treatment on cerebral vasoconstriction, vasoremodeling, and cognitive decline were investigated in vivo in a PSAPP transgenic AD mouse model. Consistent with in vitro findings, the chronic treatment of NG significantly ameliorated the cerebrovascular dysfunctions and Aβ plaque depositions in the brain. Moreover, an improved cognitive performance was also observed. Taken together, our findings supported the beneficial effects of NG on AD through adrenergic-related mechanisms and highlighted the therapeutic potential of α1-adrenergic vasomodulators against AD pathologies.
虽然淀粉样蛋白病和牛磺酸病已被认为是阿尔茨海默病(AD)大脑的特征,但最近越来越多的证据表明,脑血管变化在阿尔茨海默病的发病和发展过程中起着病理作用。在疾病的早期阶段恢复或改善受损的脑血管功能可能会对缓解阿尔茨海默病的认知功能衰退有益。在本研究中,我们评估了尼麦角林(NG,一种著名的α1肾上腺素能受体(ADR)阻断剂和血管扩张剂)通过改善血管异常对AD的潜在治疗效果。我们的体外研究数据显示,在α1-ADR过量表达的N2a细胞中,NG可以逆转β-淀粉样蛋白1-42(Aβ1-42)诱导的PKC/ERK1/2激活,这是α1-ADR激活的下游途径。NG 还能阻断 Aβ1-42- 或苯肾上腺素诱导的离体大鼠动脉收缩。所有这些体外数据可能表明,Aβ对血管功能的影响依赖于ADR,而NG具有逆转作用。此外,还在 PSAPP 转基因 AD 小鼠模型中研究了 NG 治疗对脑血管收缩、血管重塑和认知能力下降的改善作用。与体外研究结果一致,长期服用 NG 能明显改善脑血管功能障碍和脑内 Aβ 斑块沉积。此外,还观察到认知能力有所改善。综上所述,我们的研究结果支持伍格通过肾上腺素能相关机制对注意力缺失症产生有益影响,并强调了α1-肾上腺素能血管调节剂对注意力缺失症病理的治疗潜力。
{"title":"Blocking α<sub>1</sub> Adrenergic Receptor as a Novel Target for Treating Alzheimer's Disease.","authors":"Xidong Pan, Zhifeng Lei, Jiang Chen, Congcong Jia, Jie Deng, Ying Liu, Xingmei Luo, Likun Wang, Dan Zi, Zhen Wang, Song Li, Jun Tan","doi":"10.1021/acschemneuro.4c00411","DOIUrl":"https://doi.org/10.1021/acschemneuro.4c00411","url":null,"abstract":"<p><p>While amyloidopathy and tauopathy have been recognized as hallmarks in Alzheimer's disease (AD) brain, recently, increasing lines of evidence have supported the pathological roles of cerebrovascular changes in the pathogenesis and progression of AD. Restoring or ameliorating the impaired cerebrovascular function during the early phase of the disease may yield benefits against the cognitive decline in AD. In the present study, we evaluated the potential therapeutic effects of nicergoline [NG, a well-known α1 adrenergic receptor (ADR) blocker and vasodilator] against AD through ameliorating vascular abnormalities. Our in vitro data revealed that NG could reverse β-amyloid<sub>1-42</sub> (Aβ<sub>1-42</sub>)-induced PKC/ERK1/2 activation, the downstream pathway of α1-ADR activation, in α1-ADR-overexpressed N2a cells. NG also blocked Aβ<sub>1-42</sub>- or phenylephrine-induced constrictions in isolated rat arteries. All these in vitro data may suggest ADR-dependent impacts of Aβ on vascular function and the reversal effect of NG. In addition, the ameliorating impacts of NG treatment on cerebral vasoconstriction, vasoremodeling, and cognitive decline were investigated in vivo in a PSAPP transgenic AD mouse model. Consistent with in vitro findings, the chronic treatment of NG significantly ameliorated the cerebrovascular dysfunctions and Aβ plaque depositions in the brain. Moreover, an improved cognitive performance was also observed. Taken together, our findings supported the beneficial effects of NG on AD through adrenergic-related mechanisms and highlighted the therapeutic potential of α1-adrenergic vasomodulators against AD pathologies.</p>","PeriodicalId":13,"journal":{"name":"ACS Chemical Neuroscience","volume":" ","pages":""},"PeriodicalIF":4.1,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142337258","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}