首页 > 最新文献

Neurobiology of Disease最新文献

英文 中文
Pathological gain-of-function human variants in the GRIK2 kainate receptor gene cause wide-ranging behavioral dysfunction and seizures in mouse models. 在小鼠模型中,GRIK2盐酸盐受体基因的病理性功能获得性人类变异引起广泛的行为功能障碍和癫痫发作。
IF 5.6 2区 医学 Q1 NEUROSCIENCES Pub Date : 2025-12-12 DOI: 10.1016/j.nbd.2025.107226
Brynna T Webb, Hieu Trinh, Emily A Breach, Kendall M Foote, Erica Binelli, Geoffrey T Swanson

De novo variants in a subset of ionotropic glutamate receptor (iGluR) genes cause nonsyndromic neurodevelopmental disorders (NDDs). Two recurrent variants in the kainate receptor (KAR) gene GRIK2 result in the gain-of-function (GoF) substitutions p.Ala657Thr and p.Thr660Lys in a critical pore-forming domain of the GluK2 subunit. Disorders in individuals with these variants manifest as intellectual disability, developmental delay, motor impairments, and, in the case of p.Thr660Lys, epilepsy. To explore their pathogenicity and phenotypic consequences in vivo, we generated knock-in mouse models harboring orthologous Grik2 mutations. Behavioral analyses revealed a range of developmental, motor, cognitive, and naturalistic behavior impairments in both lines, with the mouse model of the variant of p.Thr660Lys, GluK2(T660K), exhibiting more severe phenotypes, consistent with clinical observations in humans. GluK2(T660K) mice also display interictal EEG abnormalities and handling-induced seizures. These models establish the first in vivo platforms for dissecting the underlying mechanisms of NDDs caused by a GoF mutation in the GluK2 KAR subunit and represent crucial tools for therapeutic development.

嗜离子性谷氨酸受体(iGluR)基因亚群的新生变异导致非综合征性神经发育障碍(ndd)。盐酸盐受体(KAR)基因GRIK2的两个复发变异体导致GluK2亚基关键孔形成结构域的p.Ala657Thr和p.Thr660Lys的功能获得(GoF)替换。这些变异个体的疾病表现为智力残疾、发育迟缓、运动障碍,以及p.s thr660lys患者的癫痫。为了探索它们在体内的致病性和表型后果,我们建立了含有同源Grik2突变的敲入小鼠模型。行为学分析揭示了这两种品系的一系列发育、运动、认知和自然行为障碍,p.Thr660Lys, GluK2(T660K)变异的小鼠模型表现出更严重的表型,与人类临床观察结果一致。GluK2(T660K)小鼠也表现出间期脑电图异常和处理诱发的癫痫发作。这些模型建立了第一个体内平台,用于剖析GluK2 KAR亚基中GoF突变引起的ndd的潜在机制,并代表了治疗开发的重要工具。
{"title":"Pathological gain-of-function human variants in the GRIK2 kainate receptor gene cause wide-ranging behavioral dysfunction and seizures in mouse models.","authors":"Brynna T Webb, Hieu Trinh, Emily A Breach, Kendall M Foote, Erica Binelli, Geoffrey T Swanson","doi":"10.1016/j.nbd.2025.107226","DOIUrl":"https://doi.org/10.1016/j.nbd.2025.107226","url":null,"abstract":"<p><p>De novo variants in a subset of ionotropic glutamate receptor (iGluR) genes cause nonsyndromic neurodevelopmental disorders (NDDs). Two recurrent variants in the kainate receptor (KAR) gene GRIK2 result in the gain-of-function (GoF) substitutions p.Ala657Thr and p.Thr660Lys in a critical pore-forming domain of the GluK2 subunit. Disorders in individuals with these variants manifest as intellectual disability, developmental delay, motor impairments, and, in the case of p.Thr660Lys, epilepsy. To explore their pathogenicity and phenotypic consequences in vivo, we generated knock-in mouse models harboring orthologous Grik2 mutations. Behavioral analyses revealed a range of developmental, motor, cognitive, and naturalistic behavior impairments in both lines, with the mouse model of the variant of p.Thr660Lys, GluK2(T660K), exhibiting more severe phenotypes, consistent with clinical observations in humans. GluK2(T660K) mice also display interictal EEG abnormalities and handling-induced seizures. These models establish the first in vivo platforms for dissecting the underlying mechanisms of NDDs caused by a GoF mutation in the GluK2 KAR subunit and represent crucial tools for therapeutic development.</p>","PeriodicalId":19097,"journal":{"name":"Neurobiology of Disease","volume":" ","pages":"107226"},"PeriodicalIF":5.6,"publicationDate":"2025-12-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145757153","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
Central integration mechanisms of neurovascular unit dysfunction and novel synergistic therapeutic strategies. 神经血管单位功能障碍的中枢整合机制及新的协同治疗策略。
IF 5.6 2区 医学 Q1 NEUROSCIENCES Pub Date : 2025-12-12 DOI: 10.1016/j.nbd.2025.107224
Haoran Wang, Yuanzheng Qiao, Haimin Lu, Xitong Bo, Fuxiang Chen, Niu Pu, Yilong Zhou, Qiong Cheng

The neurovascular unit (NVU) is a highly integrated multicellular complex composed of neurons, astrocytes, microglia, brain microvascular endothelial cells (BMECs), pericytes, and the extracellular matrix (ECM). It forms the structural and functional basis of the blood-brain barrier (BBB) and is pivotal for maintaining the homeostasis of the brain. Traditional neuroprotective strategies targeting individual cell types have shown limited efficacy in central nervous system (CNS) diseases, mainly due to the neglect of intricate intercellular crosstalk within the NVU. In this review, we first systematically summarize the core mechanisms by which the NVU functional unit causes NVU dysfunction in representative acute CNS injuries (ischemic/hemorrhagic stroke, traumatic brain injury), neurodegenerative diseases (Alzheimer's disease, Parkinson's disease, multiple sclerosis), and systemic diseases (diabetic encephalopathy, depression). Based on this, we innovatively summarize and clarify six major cross-disease pathological mechanisms of NVU dysfunction, including intercellular communication disorders, abnormal epigenetic modifications, microbiome-NVU interaction dysregulation, metabolic reprogramming dysfunction, neuroimmune-vascular coupling imbalance, and mechanical microenvironment imbalance. Additionally, we integrate emerging NVU models (co-culture systems, organoids, microfluidic chips, 3D bioprinting) with multi-omics technologies to establish a cross-scale dynamic research paradigm, and propose multicomponent coordinated regulatory strategies for NVU-targeted therapies. This framework aims to expand the understanding of NVU-centered pathological processes across diverse CNS diseases and provides a novel theoretical basis for precise therapeutic interventions, thereby bridging the gap between basic research and clinical translation.

神经血管单元(NVU)是一个高度整合的多细胞复合体,由神经元、星形胶质细胞、小胶质细胞、脑微血管内皮细胞(BMECs)、周细胞和细胞外基质(ECM)组成。它构成血脑屏障(BBB)的结构和功能基础,是维持大脑稳态的关键。传统的针对单个细胞类型的神经保护策略对中枢神经系统(CNS)疾病的疗效有限,主要是由于忽视了NVU内复杂的细胞间串扰。在这篇综述中,我们首先系统地总结了NVU功能单元在典型的急性中枢神经系统损伤(缺血性/出血性中风、创伤性脑损伤)、神经退行性疾病(阿尔茨海默病、帕金森病、多发性硬化症)和全身性疾病(糖尿病性脑病、抑郁症)中引起NVU功能障碍的核心机制。在此基础上,我们创新性地总结和阐明了NVU功能障碍的六大跨疾病病理机制,包括细胞间通讯障碍、表观遗传异常修饰、微生物组-NVU相互作用失调、代谢重编程障碍、神经免疫-血管偶联失衡和机械微环境失衡。此外,我们将新兴的NVU模型(共培养系统、类器官、微流控芯片、3D生物打印)与多组学技术相结合,建立跨尺度的动态研究范式,并提出针对NVU靶向治疗的多组分协调调控策略。该框架旨在扩大对以nvu为中心的多种中枢神经系统疾病病理过程的理解,并为精确的治疗干预提供新的理论基础,从而弥合基础研究与临床转化之间的差距。
{"title":"Central integration mechanisms of neurovascular unit dysfunction and novel synergistic therapeutic strategies.","authors":"Haoran Wang, Yuanzheng Qiao, Haimin Lu, Xitong Bo, Fuxiang Chen, Niu Pu, Yilong Zhou, Qiong Cheng","doi":"10.1016/j.nbd.2025.107224","DOIUrl":"https://doi.org/10.1016/j.nbd.2025.107224","url":null,"abstract":"<p><p>The neurovascular unit (NVU) is a highly integrated multicellular complex composed of neurons, astrocytes, microglia, brain microvascular endothelial cells (BMECs), pericytes, and the extracellular matrix (ECM). It forms the structural and functional basis of the blood-brain barrier (BBB) and is pivotal for maintaining the homeostasis of the brain. Traditional neuroprotective strategies targeting individual cell types have shown limited efficacy in central nervous system (CNS) diseases, mainly due to the neglect of intricate intercellular crosstalk within the NVU. In this review, we first systematically summarize the core mechanisms by which the NVU functional unit causes NVU dysfunction in representative acute CNS injuries (ischemic/hemorrhagic stroke, traumatic brain injury), neurodegenerative diseases (Alzheimer's disease, Parkinson's disease, multiple sclerosis), and systemic diseases (diabetic encephalopathy, depression). Based on this, we innovatively summarize and clarify six major cross-disease pathological mechanisms of NVU dysfunction, including intercellular communication disorders, abnormal epigenetic modifications, microbiome-NVU interaction dysregulation, metabolic reprogramming dysfunction, neuroimmune-vascular coupling imbalance, and mechanical microenvironment imbalance. Additionally, we integrate emerging NVU models (co-culture systems, organoids, microfluidic chips, 3D bioprinting) with multi-omics technologies to establish a cross-scale dynamic research paradigm, and propose multicomponent coordinated regulatory strategies for NVU-targeted therapies. This framework aims to expand the understanding of NVU-centered pathological processes across diverse CNS diseases and provides a novel theoretical basis for precise therapeutic interventions, thereby bridging the gap between basic research and clinical translation.</p>","PeriodicalId":19097,"journal":{"name":"Neurobiology of Disease","volume":" ","pages":"107224"},"PeriodicalIF":5.6,"publicationDate":"2025-12-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145757164","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
MAPK family members differentially regulate pThr175 tau-mediated pathogenicity. MAPK家族成员差异调控pThr175 tau介导的致病性。
IF 5.6 2区 医学 Q1 NEUROSCIENCES Pub Date : 2025-12-12 DOI: 10.1016/j.nbd.2025.107223
Neil Donison, Matthew A Hintermayer, Jacqueline Palik, Jessica Fisher, Kathryn Volkening, Michael J Strong

The phosphorylation of tau is a critical determinant of both its physiological function and the induction of pathological misfolding and aggregation. We have previously provided evidence that tau phosphorylation at Thr175 results in the exposure of the N-terminal phosphatase-activating domain (PAD) leading to the subsequent phosphorylation of Thr231, and formation of tau oligomers. A number of tauopathies, including chronic traumatic encephalopathy (CTE), amyotrophic lateral sclerosis with cognitive impairment (ALSci), and experimental traumatic brain injury (TBI) have been proposed to be associated with this cascade of events. However, the cellular mechanism by which Thr175 tau is phosphorylated remains unclear. In this study we identified ERK2, JNK1, and p38 as candidate kinases through molecular and histological analyses in a rodent model of TBI, where increased kinase activity and protein interaction were associated with pThr175 tau. We confirmed that both ERK2 and JNK1 are capable of phosphorylating Thr175 tau in vitro, but only ERK2-mediated phosphorylation of Thr175 tau induced the pathological cascade characterized by PAD exposure and the generation of oligomeric, truncated and neurofibrillary tau. Thr175 phosphorylation was also associated with an altered interaction between tau and the molecular chaperone protein DnaJC7, which regulates tau misfolding. Additionally, we observed that pThr175 and pThr231 tau were increased by oxidative stress, which was associated with the activation of the MAPK signaling pathways. These findings further clarify the mechanisms leading to Thr175 tau phosphorylation and its role in pathological tau formation by identifying ERK1 and JNK2 as important cellular mediators.

tau蛋白的磷酸化是其生理功能和诱导病理性错误折叠和聚集的关键决定因素。我们之前提供的证据表明,tau蛋白Thr175位点磷酸化导致n端磷酸酶激活域(PAD)暴露,导致随后Thr231位点磷酸化,并形成tau低聚物。包括慢性创伤性脑病(CTE)、肌萎缩侧索硬化症伴认知障碍(ALSci)和实验性创伤性脑损伤(TBI)在内的许多tau病都被认为与这一系列事件有关。然而,Thr175 tau蛋白磷酸化的细胞机制尚不清楚。在这项研究中,我们通过对TBI啮齿动物模型的分子和组织学分析确定了ERK2、JNK1和p38作为候选激酶,其中激酶活性和蛋白质相互作用的增加与pThr175 tau蛋白有关。我们证实ERK2和JNK1都能够在体外磷酸化Thr175 tau,但只有ERK2介导的Thr175 tau磷酸化诱导了以PAD暴露为特征的病理级联反应,并产生了低聚体、截断和神经原纤维tau。Thr175磷酸化还与tau蛋白与分子伴侣蛋白DnaJC7之间相互作用的改变有关,DnaJC7调节tau蛋白错误折叠。此外,我们观察到pThr175和pThr231 tau蛋白在氧化应激下增加,这与MAPK信号通路的激活有关。这些发现通过鉴定ERK1和JNK2是重要的细胞介质,进一步阐明了导致Thr175 tau磷酸化的机制及其在病理性tau形成中的作用。
{"title":"MAPK family members differentially regulate pThr175 tau-mediated pathogenicity.","authors":"Neil Donison, Matthew A Hintermayer, Jacqueline Palik, Jessica Fisher, Kathryn Volkening, Michael J Strong","doi":"10.1016/j.nbd.2025.107223","DOIUrl":"https://doi.org/10.1016/j.nbd.2025.107223","url":null,"abstract":"<p><p>The phosphorylation of tau is a critical determinant of both its physiological function and the induction of pathological misfolding and aggregation. We have previously provided evidence that tau phosphorylation at Thr175 results in the exposure of the N-terminal phosphatase-activating domain (PAD) leading to the subsequent phosphorylation of Thr231, and formation of tau oligomers. A number of tauopathies, including chronic traumatic encephalopathy (CTE), amyotrophic lateral sclerosis with cognitive impairment (ALSci), and experimental traumatic brain injury (TBI) have been proposed to be associated with this cascade of events. However, the cellular mechanism by which Thr175 tau is phosphorylated remains unclear. In this study we identified ERK2, JNK1, and p38 as candidate kinases through molecular and histological analyses in a rodent model of TBI, where increased kinase activity and protein interaction were associated with pThr175 tau. We confirmed that both ERK2 and JNK1 are capable of phosphorylating Thr175 tau in vitro, but only ERK2-mediated phosphorylation of Thr175 tau induced the pathological cascade characterized by PAD exposure and the generation of oligomeric, truncated and neurofibrillary tau. Thr175 phosphorylation was also associated with an altered interaction between tau and the molecular chaperone protein DnaJC7, which regulates tau misfolding. Additionally, we observed that pThr175 and pThr231 tau were increased by oxidative stress, which was associated with the activation of the MAPK signaling pathways. These findings further clarify the mechanisms leading to Thr175 tau phosphorylation and its role in pathological tau formation by identifying ERK1 and JNK2 as important cellular mediators.</p>","PeriodicalId":19097,"journal":{"name":"Neurobiology of Disease","volume":" ","pages":"107223"},"PeriodicalIF":5.6,"publicationDate":"2025-12-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145757099","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
Bioenergetic and protein processing imbalances in iPSC-dopamine neurons from individuals with idiopathic Parkinson's disease. 特发性帕金森病患者ipsc -多巴胺神经元的生物能量和蛋白质加工失衡
IF 5.6 2区 医学 Q1 NEUROSCIENCES Pub Date : 2025-12-12 DOI: 10.1016/j.nbd.2025.107222
Kelsey Bernard, Mandi J Corenblum, Paola Tonino, Lalitha Madhavan

Patient induced pluripotent stem cell (iPSC)-based models represent a powerful human system to gain insights into the etiopathology of Parkinson's disease (PD). Here, we studied several iPSC-derived dopamine neuron (iPSC-DAN) lines, from individuals with idiopathic PD, which is the most common form of PD. Specifically, using iPSC-DAN differentiated for 50-55 days, we performed an in-depth analysis of different bioenergetic pathways and cellular quality control mechanisms in the cells. Our results showed wide ranging impairments in oxidative phosphorylation (OXPHOS), glycolysis and creatine kinase pathways in the PD dopamine (DA) neurons. Specifically, the PD neurons exhibited reduced oxygen consumption rates (OCR) at baseline and after challenges with mitochondrial inhibitors, as well as decreased glycolytic reserves measured via ECAR. This translated to lower OCR:ECAR ratios signifying more reliance on glycolysis vs OXPHOS in the PD cells. Moreover, a mislocalization of creatine kinase B to mitochondria was seen in the PD cells. These energetic changes occurred alongside the enhanced expression of mitochondrial fission proteins, disrupted mitophagy and oxidative stress. Additionally, the PD neurons contained more monomeric, phosphorylated, and aggregated forms of alpha synuclein and displayed reduced viability. Ultrastructural examination through immuno-electron microscopy showed more alpha synuclein gold particles directly associated with mitochondria and packed into autophagic vesicles. In essence, these data capture a web of key changes, associated with neuronal degeneration, in human iPSC-DAN from persons with idiopathic PD.

基于患者诱导多能干细胞(iPSC)的模型代表了一个强大的人类系统,可以深入了解帕金森病(PD)的病因病理学。在这里,我们研究了来自特发性PD患者的几种ipsc衍生的多巴胺神经元(iPSC-DAN)系,这是最常见的PD形式。具体来说,使用分化50-55 天的iPSC-DAN,我们深入分析了细胞中不同的生物能量途径和细胞质量控制机制。我们的研究结果显示,PD多巴胺(DA)神经元的氧化磷酸化(OXPHOS)、糖酵解和肌酸激酶途径存在广泛的损伤。具体来说,PD神经元在基线和线粒体抑制剂刺激后表现出氧气消耗率(OCR)的降低,以及通过ECAR测量的糖酵解储备的降低。这转化为较低的OCR:ECAR比率,表明PD细胞更依赖糖酵解而不是OXPHOS。此外,在PD细胞中发现肌酸激酶B在线粒体中的错误定位。这些能量变化伴随着线粒体分裂蛋白的增强表达、线粒体自噬的破坏和氧化应激而发生。此外,PD神经元含有更多的单体,磷酸化和聚集形式的α突触核蛋白,并显示出降低的活力。免疫电镜超微结构检查显示更多的α -突触核蛋白金颗粒与线粒体直接相关,并被包裹在自噬囊泡中。从本质上讲,这些数据捕获了特发性PD患者iPSC-DAN中与神经元变性相关的关键变化网络。
{"title":"Bioenergetic and protein processing imbalances in iPSC-dopamine neurons from individuals with idiopathic Parkinson's disease.","authors":"Kelsey Bernard, Mandi J Corenblum, Paola Tonino, Lalitha Madhavan","doi":"10.1016/j.nbd.2025.107222","DOIUrl":"https://doi.org/10.1016/j.nbd.2025.107222","url":null,"abstract":"<p><p>Patient induced pluripotent stem cell (iPSC)-based models represent a powerful human system to gain insights into the etiopathology of Parkinson's disease (PD). Here, we studied several iPSC-derived dopamine neuron (iPSC-DAN) lines, from individuals with idiopathic PD, which is the most common form of PD. Specifically, using iPSC-DAN differentiated for 50-55 days, we performed an in-depth analysis of different bioenergetic pathways and cellular quality control mechanisms in the cells. Our results showed wide ranging impairments in oxidative phosphorylation (OXPHOS), glycolysis and creatine kinase pathways in the PD dopamine (DA) neurons. Specifically, the PD neurons exhibited reduced oxygen consumption rates (OCR) at baseline and after challenges with mitochondrial inhibitors, as well as decreased glycolytic reserves measured via ECAR. This translated to lower OCR:ECAR ratios signifying more reliance on glycolysis vs OXPHOS in the PD cells. Moreover, a mislocalization of creatine kinase B to mitochondria was seen in the PD cells. These energetic changes occurred alongside the enhanced expression of mitochondrial fission proteins, disrupted mitophagy and oxidative stress. Additionally, the PD neurons contained more monomeric, phosphorylated, and aggregated forms of alpha synuclein and displayed reduced viability. Ultrastructural examination through immuno-electron microscopy showed more alpha synuclein gold particles directly associated with mitochondria and packed into autophagic vesicles. In essence, these data capture a web of key changes, associated with neuronal degeneration, in human iPSC-DAN from persons with idiopathic PD.</p>","PeriodicalId":19097,"journal":{"name":"Neurobiology of Disease","volume":" ","pages":"107222"},"PeriodicalIF":5.6,"publicationDate":"2025-12-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145757079","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
Astrocytes differentiated from patient iPSCs model the rare leukodystrophy MLC and uncover disease-linked maturation defects and Kir4.1 channel dysfunction 从患者iPSCs分化的星形胶质细胞模拟罕见的白质营养不良的MLC,揭示疾病相关的成熟缺陷和Kir4.1通道功能障碍。
IF 5.6 2区 医学 Q1 NEUROSCIENCES Pub Date : 2025-12-08 DOI: 10.1016/j.nbd.2025.107218
Angela Lanciotti , Maria Stefania Brignone , Chiara De Nuccio , Sara Sposito , Elena Sofia Caprini , Marcello Belfiore , Francesco Nicita , Caterina Veroni , Chiara Meloni , Rosalba Carrozzo , Teresa Rizza , Chiara Aiello , Jacopo Sartorelli , Enrico Bertini , Sergio Visentin , Elena Ambrosini
Megalencephalic leukoencephalopathy with subcortical cysts (MLC) is a rare leukodystrophy caused by astrocyte dysfunction. Mutations in the MLC1 gene, which encodes the astrocyte-specific membrane protein MLC1 represent the main cause. MLC is characterized by myelin vacuolation, subcortical cysts, and brain edema. Clinically, patients show motor impairments such as ataxia and spasticity, and epilepsy. Currently, the function of MLC1 and the molecular mechanisms underlying MLC remain poorly understood, limiting therapeutic development. This is especially relevant since symptom reversibility has been observed in some patients.
To date, functional studies have mainly relied on mouse models, which do not fully reproduce human pathology. To develop a more relevant disease model, we generated astrocytes from induced pluripotent stem cells (iPSCs) derived from fibroblasts of three healthy donors and three MLC patients.
Using molecular, biochemical, electrophysiological, and imaging approaches, we found that MLC astrocytes show impaired volume regulation, cytoplasmic vacuolation, and altered EGF receptor expression, consistent with prior MLC models. Notably, we also revealed endosomal alterations, increased proliferation, and abnormal expression of the critical astrocyte maturation markers EAAT1, GFAP, Cx43, AQP4, and Kir4.1, the latter causing impaired potassium currents in patient-derived cells.
These results provide the first evidence that MLC1 mutations alter astrocyte maturation and potassium homeostasis, potentially contributing to disease pathogenesis.
Our patient-specific iPSC-derived model offers novel insights into the molecular basis of MLC and highlights the role of MLC1 in astrocyte development. This platform represents a valuable tool for preclinical drug screening and supports the development of personalized therapeutic strategies for this rare leukodystrophy.
巨脑白质脑病伴皮质下囊肿(MLC)是一种罕见的由星形胶质细胞功能障碍引起的脑白质营养不良。编码星形细胞特异性膜蛋白MLC1的MLC1基因突变是主要原因。MLC的特征是髓鞘空泡化、皮质下囊肿和脑水肿。临床上,患者表现为运动障碍,如共济失调和痉挛,以及癫痫。目前,MLC1的功能和MLC的分子机制仍然知之甚少,限制了治疗的发展。这一点尤其重要,因为在一些患者中观察到症状可逆性。迄今为止,功能研究主要依赖于小鼠模型,不能完全再现人类病理。为了建立更相关的疾病模型,我们从3名健康供体和3名MLC患者的成纤维细胞中提取的诱导多能干细胞(iPSCs)生成星形胶质细胞。通过分子、生化、电生理和成像方法,我们发现MLC星形胶质细胞表现出体积调节受损、细胞质空泡化和EGF受体表达改变,与先前的MLC模型一致。值得注意的是,我们还发现了内体改变、增殖增加和关键星形胶质细胞成熟标志物EAAT1、GFAP、Cx43、AQP4和Kir4.1的异常表达,后者导致患者来源细胞的钾电流受损。这些结果提供了MLC1突变改变星形细胞成熟和钾稳态的第一个证据,可能有助于疾病的发病机制。我们的患者特异性ipsc衍生模型为MLC的分子基础提供了新的见解,并强调了MLC1在星形胶质细胞发育中的作用。该平台代表了临床前药物筛选的宝贵工具,并支持针对这种罕见的白质营养不良的个性化治疗策略的发展。
{"title":"Astrocytes differentiated from patient iPSCs model the rare leukodystrophy MLC and uncover disease-linked maturation defects and Kir4.1 channel dysfunction","authors":"Angela Lanciotti ,&nbsp;Maria Stefania Brignone ,&nbsp;Chiara De Nuccio ,&nbsp;Sara Sposito ,&nbsp;Elena Sofia Caprini ,&nbsp;Marcello Belfiore ,&nbsp;Francesco Nicita ,&nbsp;Caterina Veroni ,&nbsp;Chiara Meloni ,&nbsp;Rosalba Carrozzo ,&nbsp;Teresa Rizza ,&nbsp;Chiara Aiello ,&nbsp;Jacopo Sartorelli ,&nbsp;Enrico Bertini ,&nbsp;Sergio Visentin ,&nbsp;Elena Ambrosini","doi":"10.1016/j.nbd.2025.107218","DOIUrl":"10.1016/j.nbd.2025.107218","url":null,"abstract":"<div><div>Megalencephalic leukoencephalopathy with subcortical cysts (MLC) is a rare leukodystrophy caused by astrocyte dysfunction. Mutations in the <em>MLC1</em> gene, which encodes the astrocyte-specific membrane protein MLC1 represent the main cause. MLC is characterized by myelin vacuolation, subcortical cysts, and brain edema. Clinically, patients show motor impairments such as ataxia and spasticity, and epilepsy. Currently, the function of MLC1 and the molecular mechanisms underlying MLC remain poorly understood, limiting therapeutic development. This is especially relevant since symptom reversibility has been observed in some patients.</div><div>To date, functional studies have mainly relied on mouse models, which do not fully reproduce human pathology. To develop a more relevant disease model, we generated astrocytes from induced pluripotent stem cells (iPSCs) derived from fibroblasts of three healthy donors and three MLC patients.</div><div>Using molecular, biochemical, electrophysiological, and imaging approaches, we found that MLC astrocytes show impaired volume regulation, cytoplasmic vacuolation, and altered EGF receptor expression, consistent with prior MLC models. Notably, we also revealed endosomal alterations, increased proliferation, and abnormal expression of the critical astrocyte maturation markers EAAT1, GFAP, Cx43, AQP4, and Kir4.1, the latter causing impaired potassium currents in patient-derived cells.</div><div>These results provide the first evidence that MLC1 mutations alter astrocyte maturation and potassium homeostasis, potentially contributing to disease pathogenesis.</div><div>Our patient-specific iPSC-derived model offers novel insights into the molecular basis of MLC and highlights the role of MLC1 in astrocyte development. This platform represents a valuable tool for preclinical drug screening and supports the development of personalized therapeutic strategies for this rare leukodystrophy.</div></div>","PeriodicalId":19097,"journal":{"name":"Neurobiology of Disease","volume":"218 ","pages":"Article 107218"},"PeriodicalIF":5.6,"publicationDate":"2025-12-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145724897","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
Cortical and subcortical gray matter alterations link cerebral small vessel disease burden to motor slowing: A cross-sectional and longitudinal study 皮层和皮层下灰质改变将大脑小血管疾病负担与运动减慢联系起来:一项横断面和纵向研究
IF 5.6 2区 医学 Q1 NEUROSCIENCES Pub Date : 2025-12-08 DOI: 10.1016/j.nbd.2025.107220
Pengcheng Liang , Tao Chen , Yena Che , Nan Zhang , Xinyue Zhang , Na Wang , Yuanyuan Wang , Yiwen Chen , Zhenyu Cheng , Changhu Liang , Lingfei Guo , Meng Li

Background

Cerebral small vessel disease (CSVD) causes cortical atrophy and motor decline, but the specific cortical regions involved and their mediating role remain unclear. We aimed to determine whether regional cortical thickness mediates the association between CSVD severity and motor function.

Methods and materials

We recruited 354 participants with CSVD (mean age 57.2 ± 11.4 years), of whom 55 had 16-month follow-up. Participants underwent 3.0 T MRI with 3D T1-weighted MPRAGE (1 mm3 isotropic) and motor testing (TUG and 3-m walk test). Cortical thickness was quantified using FreeSurfer v6.0 with longitudinal processing for follow-up scans. General linear models tested cross-sectional associations, linear mixed-effects models examined longitudinal effects, and mediation analysis assessed indirect effects.

Results

Significant negative associations were observed between cortical thickness and CSVD severity in the right insula, left rostral anterior cingulate cortex, and left lateral occipital cortex. Additionally, significant associations were found between cortical thickness at multiple time points in the right insula (β = 0.594, P = 0.024) and TUG test scores. The thickness of the right insular cortex mediated the relationship between CSVD severity and TUG performance (mean [SE] indirect effect, 0.085 [0.045]; 95 % CI, 0.015–0.197).

Conclusions

CSVD severity was associated with cortical thinning in specific cortical regions, and right insular cortical thickness levels were related to motor performance and partially mediated the association between CSVD severity and mobility impairment.

Ethics approval and consent to participate

All study procedures were approved by the Ethical Committee of the Institutional Review Board (IRB) of the Shandong Institute of Medical Imaging (2019–002). The study was conducted in accordance with the Declaration of Helsinki. All participants signed an informed consent form before the commencement of the study.
背景:脑血管病(CSVD)引起皮质萎缩和运动能力下降,但具体涉及的皮质区域及其介导作用尚不清楚。我们的目的是确定区域皮质厚度是否介导CSVD严重程度和运动功能之间的关联。方法和材料:我们招募了354例CSVD患者(平均年龄57.2 ± 11.4 岁),其中55例随访16个月。参与者接受3.0 T MRI和3D t1加权MPRAGE(1 mm3各向同性)和运动测试(TUG和3米步行测试)。使用FreeSurfer v6.0对皮质厚度进行量化,并对后续扫描进行纵向处理。一般线性模型检验横截面关联,线性混合效应模型检验纵向效应,中介分析评估间接效应。结果:右脑岛、左扣带前吻侧皮层和左枕外侧皮层皮层厚度与CSVD严重程度呈显著负相关。此外,右脑岛多个时间点的皮质厚度(β = 0.594,P = 0.024)与TUG测试分数之间存在显著相关性。右岛叶皮层厚度介导了CSVD严重程度与TUG表现之间的关系(平均[SE]间接效应,0.085[0.045];95 % CI, 0.015-0.197)。结论:CSVD严重程度与特定皮质区域的皮质变薄有关,右岛叶皮质厚度水平与运动表现有关,并部分介导CSVD严重程度与活动障碍之间的关联。伦理批准和参与同意:所有研究程序均经山东省医学影像研究所机构审查委员会(IRB)伦理委员会批准(2019-002)。这项研究是根据《赫尔辛基宣言》进行的。所有参与者在研究开始前都签署了一份知情同意书。
{"title":"Cortical and subcortical gray matter alterations link cerebral small vessel disease burden to motor slowing: A cross-sectional and longitudinal study","authors":"Pengcheng Liang ,&nbsp;Tao Chen ,&nbsp;Yena Che ,&nbsp;Nan Zhang ,&nbsp;Xinyue Zhang ,&nbsp;Na Wang ,&nbsp;Yuanyuan Wang ,&nbsp;Yiwen Chen ,&nbsp;Zhenyu Cheng ,&nbsp;Changhu Liang ,&nbsp;Lingfei Guo ,&nbsp;Meng Li","doi":"10.1016/j.nbd.2025.107220","DOIUrl":"10.1016/j.nbd.2025.107220","url":null,"abstract":"<div><h3>Background</h3><div>Cerebral small vessel disease (CSVD) causes cortical atrophy and motor decline, but the specific cortical regions involved and their mediating role remain unclear. We aimed to determine whether regional cortical thickness mediates the association between CSVD severity and motor function.</div></div><div><h3>Methods and materials</h3><div><em>We</em> recruited 354 participants with CSVD (mean age 57.2 ± 11.4 years), of whom 55 had 16-month follow-up. Participants underwent 3.0 T MRI with 3D T1-weighted MPRAGE (1 mm<sup>3</sup> isotropic) and motor testing (TUG and 3-m walk test). Cortical thickness was quantified using FreeSurfer v6.0 with longitudinal processing for follow-up scans. General linear models tested cross-sectional associations, linear mixed-effects models examined longitudinal effects, and mediation analysis assessed indirect effects.</div></div><div><h3>Results</h3><div>Significant negative associations were observed between cortical thickness and CSVD severity in the right insula, left rostral anterior cingulate cortex, and left lateral occipital cortex. Additionally, significant associations were found between cortical thickness at multiple time points in the right insula (β = 0.594, <em>P</em> = 0.024) and TUG test scores. The thickness of the right insular cortex mediated the relationship between CSVD severity and TUG performance (mean [SE] indirect effect, 0.085 [0.045]; 95 % CI, 0.015–0.197).</div></div><div><h3>Conclusions</h3><div>CSVD severity was associated with cortical thinning in specific cortical regions, and right insular cortical thickness levels were related to motor performance and partially mediated the association between CSVD severity and mobility impairment.</div></div><div><h3>Ethics approval and consent to participate</h3><div>All study procedures were approved by the Ethical Committee of the Institutional Review Board (IRB) of the Shandong Institute of Medical Imaging (2019–002). The study was conducted in accordance with the Declaration of Helsinki. All participants signed an informed consent form before the commencement of the study.</div></div>","PeriodicalId":19097,"journal":{"name":"Neurobiology of Disease","volume":"218 ","pages":"Article 107220"},"PeriodicalIF":5.6,"publicationDate":"2025-12-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145724924","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 peripheral metabolic and inflammatory biomarker signatures are associated with clinical deterioration in Creutzfeldt–Jakob disease 综合外周代谢和炎症生物标志物特征与克雅氏病的临床恶化有关。
IF 5.6 2区 医学 Q1 NEUROSCIENCES Pub Date : 2025-12-08 DOI: 10.1016/j.nbd.2025.107221
Zhong-Yun Chen , Jia-Hui Hou , Min Chu , Yi-Hao Wang , Rui Liu , Jing Zhang , Hong Ye , Miao Qu , Li-Yong Wu

Introduction

Systemic metabolic and inflammatory disturbances are implicated in neurodegenerative diseases, but comprehensive metabolomic profiling in Creutzfeldt–Jakob disease (CJD) remains limited, and the potential interplay between peripheral metabolism and inflammatory or tissue-remodeling processes is poorly understood.

Objectives

This study aimed to characterize the plasma metabolome and inflammatory/proteolytic profile in CJD, and to evaluate their individual and combined associations with cerebral glucose metabolism and clinical severity.

Methods

From January 2020 to July 2023, we recruited patients with probable or definite genetic CJD and age- and sex-matched healthy controls (HCs) at Xuanwu Hospital. All participants underwent plasma metabolomic profiling, cytokine testing, brain 18F-FDG PET/MRI, and clinical assessments. Orthogonal projections to latent structures-discriminant analysis (OPLS-DA) identified differentially abundant metabolites (variable importance in projection >1, p < 0.05). Linear and interaction models, adjusted for age and sex, evaluated associations with cerebral hypometabolism and clinical outcomes.

Results

We enrolled 40 CJD patients (mean age 60.8 years, 42.5 % female) and 40 HCs. OPLS-DA revealed clear separation between groups, and 42 differentially abundant metabolites were identified from the initial 163 metabolites that differed between groups. Enrichment analysis revealed dysregulation in metabolic pathways related to amino acid biosynthesis, alanine, aspartate and glutamate metabolism, ABC transporters, glycerophospholipid metabolism, and others. Levels of IL-4, IL-18, IL-22, IFN-γ, IL-1β, IL-6, MMP-1, and MMP-8 were significantly elevated in CJD patients. Multiple metabolites and these peripheral factors correlated with hypometabolism in vulnerable brain regions and with cognitive and functional decline. Interaction analyses further showed that specific metabolite–mediator combinations (e.g., Glycerophosphocholine, Glyceric acid, Asparagine, Dimethylglycine, 3-Phosphoglycerate with IL-1β, IL-4, IL-6, IL-22, MMP-8) were significantly associated with regional hypometabolism and clinical deterioration.

Conclusion

CJD involves coupled peripheral metabolic and inflammatory/proteolytic disturbances that may be associated with more severe brain degeneration and clinical decline, suggesting a possible multifaceted systemic component accompanying disease pathology.
系统性代谢和炎症紊乱与神经退行性疾病有关,但克雅氏病(CJD)的全面代谢组学分析仍然有限,外周代谢与炎症或组织重塑过程之间的潜在相互作用尚不清楚。目的:本研究旨在表征克雅氏病的血浆代谢组和炎症/蛋白水解谱,并评估它们与脑糖代谢和临床严重程度的个体和联合关联。方法:从2020年1月至2023年7月,我们在宣武医院招募了可能或确定的遗传性CJD患者和年龄和性别匹配的健康对照(hc)。所有参与者都进行了血浆代谢组学分析、细胞因子测试、脑18F-FDG PET/MRI和临床评估。正交预测到潜在结构-判别分析(OPLS-DA)鉴定出差异丰富的代谢物(预测的可变重要性bbb1, p )结果:我们招募了40名CJD患者(平均年龄60.8 岁,女性42.5 %)和40名hc患者。OPLS-DA显示各组之间存在明显的分离,从163个初始代谢物中鉴定出42个差异丰富的代谢物。富集分析显示与氨基酸生物合成、丙氨酸、天冬氨酸和谷氨酸代谢、ABC转运蛋白、甘油磷脂代谢等相关的代谢途径失调。CJD患者IL-4、IL-18、IL-22、IFN-γ、IL-1β、IL-6、MMP-1、MMP-8水平显著升高。多种代谢物和这些外围因素与大脑脆弱区域的代谢低下以及认知和功能下降相关。相互作用分析进一步表明,特定代谢物-介质组合(如甘油酰胆碱、甘油三酸、天冬酰胺、二甲基甘氨酸、3-磷酸甘油酸与IL-1β、IL-4、IL-6、IL-22、MMP-8)与局部低代谢和临床恶化显著相关。结论:克雅氏病涉及外周代谢和炎症/蛋白水解紊乱,这些紊乱可能与更严重的脑变性和临床衰退有关,提示可能存在伴随疾病病理的多方面系统性成分。
{"title":"Integrated peripheral metabolic and inflammatory biomarker signatures are associated with clinical deterioration in Creutzfeldt–Jakob disease","authors":"Zhong-Yun Chen ,&nbsp;Jia-Hui Hou ,&nbsp;Min Chu ,&nbsp;Yi-Hao Wang ,&nbsp;Rui Liu ,&nbsp;Jing Zhang ,&nbsp;Hong Ye ,&nbsp;Miao Qu ,&nbsp;Li-Yong Wu","doi":"10.1016/j.nbd.2025.107221","DOIUrl":"10.1016/j.nbd.2025.107221","url":null,"abstract":"<div><h3>Introduction</h3><div>Systemic metabolic and inflammatory disturbances are implicated in neurodegenerative diseases, but comprehensive metabolomic profiling in Creutzfeldt–Jakob disease (CJD) remains limited, and the potential interplay between peripheral metabolism and inflammatory or tissue-remodeling processes is poorly understood.</div></div><div><h3>Objectives</h3><div>This study aimed to characterize the plasma metabolome and inflammatory/proteolytic profile in CJD, and to evaluate their individual and combined associations with cerebral glucose metabolism and clinical severity.</div></div><div><h3>Methods</h3><div>From January 2020 to July 2023, we recruited patients with probable or definite genetic CJD and age- and sex-matched healthy controls (HCs) at Xuanwu Hospital. All participants underwent plasma metabolomic profiling, cytokine testing, brain <sup>18</sup>F-FDG PET/MRI, and clinical assessments. Orthogonal projections to latent structures-discriminant analysis (OPLS-DA) identified differentially abundant metabolites (variable importance in projection &gt;1, <em>p</em> &lt; 0.05). Linear and interaction models, adjusted for age and sex, evaluated associations with cerebral hypometabolism and clinical outcomes.</div></div><div><h3>Results</h3><div>We enrolled 40 CJD patients (mean age 60.8 years, 42.5 % female) and 40 HCs. OPLS-DA revealed clear separation between groups, and 42 differentially abundant metabolites were identified from the initial 163 metabolites that differed between groups. Enrichment analysis revealed dysregulation in metabolic pathways related to amino acid biosynthesis, alanine, aspartate and glutamate metabolism, ABC transporters, glycerophospholipid metabolism, and others. Levels of IL-4, IL-18, IL-22, IFN-γ, IL-1β, IL-6, MMP-1, and MMP-8 were significantly elevated in CJD patients. Multiple metabolites and these peripheral factors correlated with hypometabolism in vulnerable brain regions and with cognitive and functional decline. Interaction analyses further showed that specific metabolite–mediator combinations (e.g., Glycerophosphocholine, Glyceric acid, Asparagine, Dimethylglycine, 3-Phosphoglycerate with IL-1β, IL-4, IL-6, IL-22, MMP-8) were significantly associated with regional hypometabolism and clinical deterioration.</div></div><div><h3>Conclusion</h3><div>CJD involves coupled peripheral metabolic and inflammatory/proteolytic disturbances that may be associated with more severe brain degeneration and clinical decline, suggesting a possible multifaceted systemic component accompanying disease pathology.</div></div>","PeriodicalId":19097,"journal":{"name":"Neurobiology of Disease","volume":"218 ","pages":"Article 107221"},"PeriodicalIF":5.6,"publicationDate":"2025-12-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145724950","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
Applying biologically anchored subtypes to advance precision medicine in autism spectrum disorder 应用生物学锚定亚型推进自闭症谱系障碍的精准医学。
IF 5.6 2区 医学 Q1 NEUROSCIENCES Pub Date : 2025-12-06 DOI: 10.1016/j.nbd.2025.107219
Tae-Yong Choi , Aileen Gunawan , DaYeong Seo , Jinkyu Park , Eun Hee Ahn , Sang Won Suh , Marc V. Fuccillo , Kyuhyun Choi
Autism spectrum disorder (ASD) is heterogeneous at every level, from behavior to molecular pathways, limiting the value of subgrouping schemes built on surface phenotypes alone. We synthesize evidence that biologically anchored subtypes, defined by convergent genetics, developmental timing, and brain–body crosstalk, offer a tractable path to precision medicine. Leveraging advances in large-scale genomic resources and computational analytics, we propose a multi-axis framework: (i) genetic architecture spanning rare variants and polygenic load, (ii) developmental windows from mid-gestation to infancy divergence and regression, and (iii) brain–body interactions shaping plasticity and symptom expression. This framework enables mechanism-guided therapeutic strategies through biomarker-stratified enrollment, target-engagement readouts, and circuit-anchored outcomes. Preclinical platforms, genetically engineered mice and patient-derived induced pluripotent stem cells (iPSCs), demonstrate convergence onto limited synaptic and connectivity “neurotypes,” enabling causal links from gene to circuit to behavior and proof-of-concept rescue. We close with priorities: standardized multi-platform characterization, decision tools linking subtype labels to interventions, and stratified trials that co-report clinical and biological endpoints, with ethical guardrails to ensure early stratification expands opportunity while advancing individualized care.
自闭症谱系障碍(ASD)在从行为到分子途径的各个层面上都是异质性的,这限制了仅基于表面表型的亚群方案的价值。我们综合证据,生物锚定亚型,由趋同遗传学,发育时间和脑-体串扰定义,提供了一个易于处理的路径,以精确医学。利用大规模基因组资源和计算分析的进步,我们提出了一个多轴框架:(i)跨越罕见变异和多基因负荷的遗传结构,(ii)从妊娠中期到婴儿期的发育窗口分化和回归,以及(iii)脑-体相互作用塑造可塑性和症状表达。该框架通过生物标志物分层入组、目标参与读数和回路锚定结果实现机制指导的治疗策略。临床前平台,基因工程小鼠和患者衍生的诱导多能干细胞(iPSCs),证明了收敛到有限的突触和连接“神经类型”,实现了从基因到电路到行为的因果联系和概念验证拯救。最后,我们提出了以下优先事项:标准化的多平台表征,将亚型标签与干预措施联系起来的决策工具,以及联合报告临床和生物学终点的分层试验,以及确保早期分层在推进个体化护理的同时扩大机会的伦理保障。
{"title":"Applying biologically anchored subtypes to advance precision medicine in autism spectrum disorder","authors":"Tae-Yong Choi ,&nbsp;Aileen Gunawan ,&nbsp;DaYeong Seo ,&nbsp;Jinkyu Park ,&nbsp;Eun Hee Ahn ,&nbsp;Sang Won Suh ,&nbsp;Marc V. Fuccillo ,&nbsp;Kyuhyun Choi","doi":"10.1016/j.nbd.2025.107219","DOIUrl":"10.1016/j.nbd.2025.107219","url":null,"abstract":"<div><div>Autism spectrum disorder (ASD) is heterogeneous at every level, from behavior to molecular pathways, limiting the value of subgrouping schemes built on surface phenotypes alone. We synthesize evidence that biologically anchored subtypes, defined by convergent genetics, developmental timing, and brain–body crosstalk, offer a tractable path to precision medicine. Leveraging advances in large-scale genomic resources and computational analytics, we propose a multi-axis framework: (i) genetic architecture spanning rare variants and polygenic load, (ii) developmental windows from mid-gestation to infancy divergence and regression, and (iii) brain–body interactions shaping plasticity and symptom expression. This framework enables mechanism-guided therapeutic strategies through biomarker-stratified enrollment, target-engagement readouts, and circuit-anchored outcomes. Preclinical platforms, genetically engineered mice and patient-derived induced pluripotent stem cells (iPSCs), demonstrate convergence onto limited synaptic and connectivity “neurotypes,” enabling causal links from gene to circuit to behavior and proof-of-concept rescue. We close with priorities: standardized multi-platform characterization, decision tools linking subtype labels to interventions, and stratified trials that co-report clinical and biological endpoints, with ethical guardrails to ensure early stratification expands opportunity while advancing individualized care.</div></div>","PeriodicalId":19097,"journal":{"name":"Neurobiology of Disease","volume":"218 ","pages":"Article 107219"},"PeriodicalIF":5.6,"publicationDate":"2025-12-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145708625","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
Shank3B−/− pathophysiology: Early metformin treatment rescues behavioural deficits and normalises exacerbated mRNA translation Shank3B-/-病理生理学:早期二甲双胍治疗可挽救行为缺陷并使加剧的mRNA翻译正常化。
IF 5.6 2区 医学 Q1 NEUROSCIENCES Pub Date : 2025-12-04 DOI: 10.1016/j.nbd.2025.107217
Laura Marsal-García , Jung-Hyun Choi , Eve Peraldi , Pei You Wu , Cong Loc Dang , R. Anne McKinney , Ilse Gantois , Nahum Sonenberg
Phelan-McDermid syndrome (PMS), a rare neurodevelopmental disorder associated with autism spectrum disorder and intellectual disability, is caused by either deletions in human chromosome 22q13 or mutations in the SH3 and multiple ankyrin repeat domains 3 (SHANK3) gene. PMS is highly debilitating, and existing treatments are ineffective. SHANK3 interacts with at least 3 synaptic receptors through synaptic-associated proteins, some of which are upstream of the mammalian/mechanistic target of rapamycin complex 1 (mTORC1) and mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) signalling pathways. Metformin, an inhibitor of the mTORC1 and MAPK/ERK signalling pathways, was shown to correct core phenotypes in a fragile X mouse model, offering therapeutic potential for PMS. Male Shank3B−/−, a PMS mouse model, and wild-type mice were treated from birth with metformin (5 mg/mL) or vehicle. Shank3B−/− mice displayed increased self-grooming, decreased social interaction, reduced duration and frequency of ultrasonic vocalisations, and impaired hippocampal-dependent memory. Upregulated mTORC1 activity was observed in the hippocampus and prefrontal cortex, along with decreased synaptosomal protein expression in the striatum of GluN2B (an N-methyl-d-aspartate receptor (NMDAR) subunit), Homer1 (a synaptic-associated protein) and GluA2 (an α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) subunit), all of which interact with Shank3. Metformin treatment from birth corrected core behavioural impairments, exaggerated mRNA translation, and decreased striatal synaptic protein expression. Considering its exceptional safety profile, metformin is a promising therapeutic option for a rapid clinical translation of PMS treatment.
Phelan-McDermid综合征(PMS)是一种罕见的神经发育障碍,与自闭症谱系障碍和智力残疾相关,由人类染色体22q13缺失或SH3和多锚蛋白重复结构域3 (SHANK3)基因突变引起。经前症候群非常虚弱,现有的治疗方法无效。SHANK3通过突触相关蛋白与至少3种突触受体相互作用,其中一些位于哺乳动物/雷帕霉素复合物1 (mTORC1)的机制靶点和丝裂原活化蛋白激酶/细胞外信号调节激酶(MAPK/ERK)信号通路的上游。二甲双胍是一种mTORC1和MAPK/ERK信号通路的抑制剂,在脆性X小鼠模型中被证明可以纠正核心表型,为经前症候群提供治疗潜力。雄性经前期综合征小鼠模型Shank3B-/-和野生型小鼠从出生起就接受二甲双胍(5 mg/mL)或对照剂治疗。Shank3B-/-小鼠表现出自我梳理增加,社会互动减少,超声波发声的持续时间和频率减少,海马体依赖性记忆受损。海马和前额叶皮层中mTORC1活性上调,纹状体中GluN2B (n -甲基-d-天冬氨酸受体(NMDAR)亚基)、Homer1(突触相关蛋白)和GluA2 (α-氨基-3-羟基-5-甲基-4-异氧唑丙酸受体(AMPAR)亚基)的突触体蛋白表达下降,所有这些蛋白都与Shank3相互作用。二甲双胍治疗从出生纠正核心行为障碍,夸大mRNA翻译,降低纹状体突触蛋白表达。考虑到其特殊的安全性,二甲双胍是一种有前途的治疗选择,用于经前症候群治疗的快速临床转化。
{"title":"Shank3B−/− pathophysiology: Early metformin treatment rescues behavioural deficits and normalises exacerbated mRNA translation","authors":"Laura Marsal-García ,&nbsp;Jung-Hyun Choi ,&nbsp;Eve Peraldi ,&nbsp;Pei You Wu ,&nbsp;Cong Loc Dang ,&nbsp;R. Anne McKinney ,&nbsp;Ilse Gantois ,&nbsp;Nahum Sonenberg","doi":"10.1016/j.nbd.2025.107217","DOIUrl":"10.1016/j.nbd.2025.107217","url":null,"abstract":"<div><div>Phelan-McDermid syndrome (PMS), a rare neurodevelopmental disorder associated with autism spectrum disorder and intellectual disability, is caused by either deletions in human chromosome 22q13 or mutations in the SH3 and multiple ankyrin repeat domains 3 <em>(SHANK3)</em> gene. PMS is highly debilitating, and existing treatments are ineffective. SHANK3 interacts with at least 3 synaptic receptors through synaptic-associated proteins, some of which are upstream of the mammalian/mechanistic target of rapamycin complex 1 (mTORC1) and mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) signalling pathways. Metformin, an inhibitor of the mTORC1 and MAPK/ERK signalling pathways, was shown to correct core phenotypes in a fragile X mouse model, offering therapeutic potential for PMS. Male <em>Shank3B</em><sup><em>−/−</em></sup>, a PMS mouse model, and wild-type mice were treated from birth with metformin (5 mg/mL) or vehicle. <em>Shank3B</em><sup><em>−/−</em></sup> mice displayed increased self-grooming, decreased social interaction, reduced duration and frequency of ultrasonic vocalisations, and impaired hippocampal-dependent memory. Upregulated mTORC1 activity was observed in the hippocampus and prefrontal cortex, along with decreased synaptosomal protein expression in the striatum of GluN2B (an <em>N</em>-methyl-<span>d</span>-aspartate receptor (NMDAR) subunit), Homer1 (a synaptic-associated protein) and GluA2 (an α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor (AMPAR) subunit), all of which interact with Shank3. Metformin treatment from birth corrected core behavioural impairments, exaggerated mRNA translation, and decreased striatal synaptic protein expression. Considering its exceptional safety profile, metformin is a promising therapeutic option for a rapid clinical translation of PMS treatment.</div></div>","PeriodicalId":19097,"journal":{"name":"Neurobiology of Disease","volume":"218 ","pages":"Article 107217"},"PeriodicalIF":5.6,"publicationDate":"2025-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145695775","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
Somatosensory cortical progesterone receptor signaling regulates the episodic to chronic migraine transformation 体感皮质孕酮受体信号调节偏头痛发作性到慢性的转变。
IF 5.6 2区 医学 Q1 NEUROSCIENCES Pub Date : 2025-12-04 DOI: 10.1016/j.nbd.2025.107215
Suchitra Joshi , John Williamson , Daniel Etcheberrigaray , Howard P. Goodkin , Jaideep Kapur
Women during their reproductive years are more susceptible to migraines than men, largely due to the effects of female reproductive hormones. Progesterone receptor (PR) signaling influences episodic migraine susceptibility. Here, we investigated the role of PRs in regulating the transformation from episodic to chronic migraine.
Adult female and male C57Bl6 mice, female conditional PR knockout (PRKO) and littermate wild-type (WT) mice, and female mice expressing a Cre recombinase under the progesterone receptor (Pgr) promoter were used. Migraine transformation was induced using five systemic alternate-day nitroglycerin (NTG) injections. Periorbital mechanical thresholds were assessed using von Frey monofilaments. AAV9-mediated transduction was used to express muscarinic acetylcholine receptor-based DREADD complex hM4Di for silencing the activity of PR-expressing somatosensory cortical neurons. Somatosensory cortical neuronal activation was assessed using cFos immunohistochemistry.
Gonadally intact female mice were three times more susceptible to developing transformed migraines; they experienced faster pain sensitization after repeated NTG injections compared to males, and their recovery from pain was also slower. PR agonist segesterone promoted migraine transformation in females. In contrast, migraine transformation was slower in PRKO females than in WT females, and their recovery was quicker. PRs are expressed in the somatosensory cortex, and after five NTG injections, there was stronger cFos immunoreactivity in the somatosensory, insular, and cingulate cortices of female WT mice than in PRKO mice. When PR-expressing somatosensory cortical neurons were chemo-genetically silenced, the transition from acute to chronic migraine slowed.
PR activation on the somatosensory cortex neurons contributes to the transformation of acute to chronic migraine.
处于生育年龄的女性比男性更容易患偏头痛,这主要是由于女性生殖激素的影响。孕激素受体(PR)信号影响发作性偏头痛的易感性。在此,我们研究了pr在偏头痛发作性向慢性转变中的作用。研究对象为成年雌性和雄性C57Bl6小鼠、雌性条件PR敲除(PRKO)和野生型(WT)小鼠以及在孕激素受体(Pgr)启动子下表达Cre重组酶的雌性小鼠。采用5次全身隔天注射硝酸甘油(NTG)诱导偏头痛转化。使用von Frey单丝评估眶周力学阈值。aav9介导的转导被用于表达毒蕈碱乙酰胆碱受体为基础的DREADD复合物hM4Di,以沉默表达pr的体感觉皮质神经元的活性。采用cFos免疫组织化学评估体感觉皮层神经元的激活。性腺完整的雌性小鼠患变异性偏头痛的可能性是正常小鼠的三倍;与男性相比,在重复注射NTG后,她们的疼痛敏感化速度更快,而且她们从疼痛中恢复的速度也更慢。PR激动剂孕酮促进女性偏头痛转化。相比之下,PRKO女性的偏头痛转化速度比WT女性慢,恢复速度更快。pr在体感觉皮层表达,经5次NTG注射后,雌性WT小鼠体感觉皮层、岛状皮层和扣带皮层的cFos免疫反应性比PRKO小鼠强。当表达pr的体感觉皮质神经元被化学基因沉默时,从急性偏头痛到慢性偏头痛的转变减慢。体感觉皮层神经元的PR激活有助于急性偏头痛向慢性偏头痛的转变。
{"title":"Somatosensory cortical progesterone receptor signaling regulates the episodic to chronic migraine transformation","authors":"Suchitra Joshi ,&nbsp;John Williamson ,&nbsp;Daniel Etcheberrigaray ,&nbsp;Howard P. Goodkin ,&nbsp;Jaideep Kapur","doi":"10.1016/j.nbd.2025.107215","DOIUrl":"10.1016/j.nbd.2025.107215","url":null,"abstract":"<div><div>Women during their reproductive years are more susceptible to migraines than men, largely due to the effects of female reproductive hormones. Progesterone receptor (PR) signaling influences episodic migraine susceptibility. Here, we investigated the role of PRs in regulating the transformation from episodic to chronic migraine.</div><div>Adult female and male C57Bl6 mice, female conditional PR knockout (PRKO) and littermate wild-type (WT) mice, and female mice expressing a Cre recombinase under the progesterone receptor (<em>Pgr</em>) promoter were used. Migraine transformation was induced using five systemic alternate-day nitroglycerin (NTG) injections. Periorbital mechanical thresholds were assessed using von Frey monofilaments. AAV9-mediated transduction was used to express muscarinic acetylcholine receptor-based DREADD complex hM4Di for silencing the activity of PR-expressing somatosensory cortical neurons. Somatosensory cortical neuronal activation was assessed using cFos immunohistochemistry.</div><div>Gonadally intact female mice were three times more susceptible to developing transformed migraines; they experienced faster pain sensitization after repeated NTG injections compared to males, and their recovery from pain was also slower. PR agonist segesterone promoted migraine transformation in females. In contrast, migraine transformation was slower in PRKO females than in WT females, and their recovery was quicker. PRs are expressed in the somatosensory cortex, and after five NTG injections, there was stronger cFos immunoreactivity in the somatosensory, insular, and cingulate cortices of female WT mice than in PRKO mice. When PR-expressing somatosensory cortical neurons were chemo-genetically silenced, the transition from acute to chronic migraine slowed.</div><div>PR activation on the somatosensory cortex neurons contributes to the transformation of acute to chronic migraine.</div></div>","PeriodicalId":19097,"journal":{"name":"Neurobiology of Disease","volume":"218 ","pages":"Article 107215"},"PeriodicalIF":5.6,"publicationDate":"2025-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145695870","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
期刊
Neurobiology of Disease
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:604180095
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