Loading...

Table of Content

    15 June 2026, Volume 21 Issue 6 Previous Issue   
    For Selected: Toggle Thumbnails
    Voltage-gated sodium channels in the nervous system: Molecular physiology to therapeutic interventions
    Ni Li, Lin Yan, Anna Peng, Xuefei Fu, Huan Qin, Kai Yao
    2026, 21 (6):  2085-2106.  doi: 10.4103/NRR.NRR-D-25-00260
    Abstract ( 146 )   PDF (6258KB) ( 128 )   Save

    Voltage-gated sodium channels are essential ionic-conductance pathways in the nervous system, which play an irreplaceable role in modulating neuronal excitability and signal transduction. This review comprehensively analyzes the molecular mechanisms and pathophysiological significance of voltage-gated sodium channels, with particular emphasis on elucidating the molecular-action mechanisms of the distinct subtypes of these channels, including Nav1.1, Nav1.2, and Nav1.6, across various neurological disorders such as familial hemiplegic migraine, epilepsy, autism spectrum disorder, and retinal dysfunction. This review also provides a comprehensive overview of the pathogenic mechanisms associated with voltage-gated sodium channels, and systematically clarifies the evolutionary pathway of treatment strategies from conventional to innovative approaches. It analyzes two major categories of conventional sodium channel blockers and their applications: antiepileptic drugs (such as carbamazepine, lamotrigine, and phenytoin) and antiarrhythmic drugs (such as lidocaine, flecainide, and quinidine). However, these conventional blockers show limitations because of the lack of selectivity, driving research toward more precise therapeutic directions. Additionally, this review evaluates gabapentin, cannabidiol, and calcium channel blockers with different mechanisms of action. These drugs modulate neuronal excitability from multiple perspectives, providing diverse options for symptom relief. This review also highlights advances in gene therapy for specific diseases, such as STK-001, which promotes effective splicing of the SCN1A gene, and ETX101, which utilizes adeno-associated virus 9 vectors to deliver engineered transcription factors. These two agents provide targeted therapeutic solutions for Dravet syndrome. Furthermore, this review summarizes some innovative therapeutic agents in clinical trials, including PRAX-222 (for SCN2A gain-of-function mutation-related epilepsy), which has received Food and Drug Administration orphan drug designation, and the selective Nav1.6 inhibitor NBI-921352 (for SCN8A-related epilepsy). Collectively, this review comprehensively compares the advantages and disadvantages of conventional drugs and gene therapy and envisions future treatment strategies that integrate the strengths of both approaches, facilitating personalized precision medicine to provide more accurate and effective treatment options for patients with ion channel diseases.

    Related Articles | Metrics
    Regulatory T cells in stroke inflammation: Therapeutic perspectives
    Ziyi Sun, Hongyu Zhou, Yongjun Wang, Zixiao Li
    2026, 21 (6):  2107-2123.  doi: 10.4103/NRR.NRR-D-24-01424
    Abstract ( 141 )   PDF (10659KB) ( 14 )   Save
    Regulatory T cells are crucial immunomodulatory cells that play essential roles in both ischemic stroke and intracerebral hemorrhage. These cells are vital in post-stroke inflammation since they suppress immune responses and promote tissue repair. This review thoroughly examines the dynamic changes in the number and function of regulatory T cells and highlights their distinct roles at various stages of stroke progression. In the acute phase (within 5–7 days), regulatory T cells exert neuroprotective effects primarily by reducing inflammation. In the chronic phase (7 days post-onset), these cells support neuroregeneration and functional recovery. The review also explores the emerging role of regulatory T cells in the brain–gut axis, a key mediator of the systemic immune responses following stroke, and discusses its relevance in modulating post-stroke inflammation and repair. Various strategies aimed at enhancing regulatory T cell responses include adoptive transfer of regulatory T cells, administration of pharmacological agents, and induction of mucosal tolerance. All these approaches can potentially enhance the immunomodulatory and repair functions of regulatory T cells. Nevertheless, despite the promising preclinical results, the translation of regulatory T cell–based therapies into clinical practice is associated with challenges related to optimal timing, dosage, and long-term efficacy. Overall, targeting regulatory T cells is a novel and promising immunoregulatory approach for mitigating stroke-induced injury and promoting neural repair.
    Related Articles | Metrics
    Research progress on the Sonic Hedgehog signaling pathway in the central nervous system: Novel insights
    Nuokun Li, Shiyi Wen, Dandan Li, Yaning Shi, Zhigang Mei, Danhong Liu, Hui Yang, Yuhong Wang, Xiaoyuan Lin, Yun Xiang, Hongbo Wen, Pan Meng
    2026, 21 (6):  2124-2140.  doi: 10.4103/NRR.NRR-D-24-01239
    Abstract ( 132 )   PDF (6258KB) ( 498 )   Save
    Over the past few decades, the Sonic Hedgehog protein has become a pivotal player in many biological processes, including tumourigenesis, embryonic development, and protective mechanisms after cerebral damage. The Sonic Hedgehog signaling pathway is crucial in the central nervous system, with implications in a diverse range of diseases, including Parkinson’s disease, Alzheimer’s disease, spinal cord injury, traumatic brain injury, depression, Sonic Hedgehog medulloblastoma, and stroke. In this comprehensive review, we examined Sonic Hedgehog from the perspective of canonical and non-canonical pathways, elucidating their complex connections to the central nervous system. Subsequently, we summarize the latest advancements in drug therapies that offer novel strategies for treating neurological diseases by modulating the Sonic Hedgehog protein. Finally, we summarize and extend the technologies and tools for studying the Sonic Hedgehog signaling field, with the aim of providing new research ideas and methods.
    Related Articles | Metrics
    Implications of mitochondrial phosphatidylethanolamine in neuronal health and neurodegeneration
    Yantao Zuo, Niharika Amireddy, Qian Cai
    2026, 21 (6):  2141-2156.  doi: 10.4103/NRR.NRR-D-25-00201
    Abstract ( 76 )   PDF (1533KB) ( 80 )   Save
    Phosphatidylethanolamine is a major phospholipid class abundant in the brain, particularly in the inner leaflet of the plasma and mitochondrial membranes. Although it is primarily synthesized from phosphatidylserine via decarboxylation in mitochondria or from ethanolamine via the cytidine diphosphate-ethanolamine pathway in the endoplasmic reticulum, phosphatidylethanolamine that resides in mitochondria is preferentially produced locally and is distinct and separate from the pool of phosphatidylethanolamine made in the endoplasmic reticulum. Mitochondriaderived phosphatidylethanolamine is not only essential for mitochondrial integrity but also is exported to other organelles to fulfill diverse cellular functions. Neurons are highly enriched with phosphatidylethanolamine, and the importance of phosphatidylethanolamine metabolism in neuronal health has recently been recognized following its reported links to Alzheimer’s disease, Parkinson’s disease, and hereditary spastic paraplegia, among other neurological disorders. Indeed, disturbances in mitochondrial function and phosphatidylethanolamine metabolism and the resulting neuronal dysfunction are the common features of individuals suffering from these diseases, highlighting the great importance of maintaining proper phosphatidylethanolamine homeostasis in neurons. In this review, we summarize the current knowledge of phosphatidylethanolamine metabolism and its role in neuronal function with a special emphasis on the phosphatidylethanolamine biosynthetic pathway in mitochondria. We then review findings on how phosphatidylethanolamine biosynthesis is affected in major neurodegenerative diseases. Finally, we highlight promising future research areas that will help advance the understanding of neuronal phosphatidylethanolamine mechanisms and identify phosphatidylethanolamine-targeted therapeutic strategies for combating such brain diseases.
    Related Articles | Metrics
    Modulation of mitochondrial dysfunction: Mechanisms and strategies for the use of natural products to treat stroke
    Na Qin, Rujuan Liu, Rong Deng, Liuliu Shi, Lei Wang , Ting Zhu
    2026, 21 (6):  2157-2174.  doi: 10.4103/NRR.NRR-D-25-00016
    Abstract ( 121 )   PDF (8690KB) ( 16 )   Save
    Modulations of mitochondrial dysfunction, which involve a series of dynamic processes such as mitochondrial biogenesis, mitochondrial fusion and fission, mitochondrial transport, mitochondrial autophagy, mitochondrial apoptosis, and oxidative stress, play an important role in the onset and progression of stroke. With a better understanding of the critical role of mitochondrial dysfunction modulations in post-stroke neurological injury, these modulations have emerged as a potential target for stroke prevention and treatment. Additionally, since effective treatments for stroke are extremely limited and natural products currently offer some outstanding advantages, we focused on the findings and mechanisms of action related to the use of natural products for targeting mitochondrial dysfunction in the treatment of stroke. Natural products achieve neuroprotective through multi-target regulation of mitochondrial dysfunction encompassing the following processes: (1) Mitochondrial biogenesis: Cordyceps and hydroxysafflor yellow A activate the peroxisome proliferator–activated receptor gamma coactivator 1-alpha/nuclear respiratory factor pathway, promote mitochondrial DNA replication and respiratory chain protein synthesis, and thereby restore energy supply in the ischemic penumbra. (2) Mitochondrial dynamics balance: Ginsenoside Rb3 promotes Opa1-mediated neural stem cell migration and diffusion for recovery of damaged brain tissue. (3) Mitochondrial autophagy: Gypenoside XVII selectively eliminates damaged mitochondria via the phosphatase and tensin homolog-induced kinase 1/Parkin pathway and blocks reactive oxygen species and the NODlike receptor protein 3 inflammasome cascade, thereby alleviating blood–brain barrier damage. (4) Anti-apoptotic mechanisms: Ginkgolide K inhibits Bax mitochondrial translocation and downregulates caspase-3/9 activity, reducing neuronal programmed death induced by ischemia-reperfusion. (5) Oxidative stress regulation: Scutellarin exerts antioxidant properties and improves neurological function by modulating the extracellular signal-regulated kinase 5-Kruppel-like factor 2-endothelial nitric oxide synthase signaling pathway. (6) Intercellular mitochondrial transport: Neuroprotective effects of Chrysophanol are associated with accelerated mitochondrial transfer from astrocytes to neurons. Existing studies have confirmed that natural products exhibit neuroprotective effects through multidimensional interventions targeting mitochondrial dysfunction in both ischemic and hemorrhagic stroke models. However, their clinical translation still faces challenges, such as the difficulty in standardization due to component complexity, insufficient cross-regional clinical data, and the lack of long-term safety evaluations. Future research should aim to integrate new technologies, such as singlecell sequencing and organoid models, to deeply explore the mitochondria-targeting mechanisms of natural products and validate their efficacy through multicenter clinical trials, providing theoretical support and translational pathways for the development of novel anti-stroke drugs.
    Related Articles | Metrics
    Bacterial extracellular vesicles in the brain: Pathological effects and therapeutic possibilities
    Yaiza M. Arenas, Paula Izquierdo-Altarejos, Gaspar Pérez-Martínez, Vicente Felipo, Marta Llansola
    2026, 21 (6):  2175-2185.  doi: 10.4103/NRR.NRR-D-25-00236
    Abstract ( 82 )   PDF (2275KB) ( 106 )   Save
    The mechanisms leading to neurological and neurodegenerative diseases are not completely known, and new, more effective, therapeutic treatments are necessary for most neurological pathologies. The treatment of neurological and neurodegenerative diseases is complicated due to the blood–brain barrier, which makes it difficult for drugs to access the brain areas in which they must act to improve the pathology. A tool that can help to overcome this difficulty is the use of extracellular vesicles, which can easily cross the blood–brain barrier. The extracellular vesicles are considered a main way of communication between the brain and the rest of the body, with important implications for the physiopathology and therapy of neurological diseases. In recent years, the involvement of microbiota in many neurological pathologies, as well as its possible therapeutic role, has also become evident. A key mediator in the pathologic and beneficial effects of microbiota seems to be the bacterial extracellular vesicles. There is an important communication between the brain and the intestinal microbiota (the gut–brain axis), by which the microbiota influences brain function, impacts on mental health, and plays a role in different neurological and neurodegenerative diseases. The identification of the mechanisms involved in this gut–brain axis is essential to understanding the mechanisms of neurological pathologies and to developing more effective treatments for these diseases. Bacterial extracellular vesicles would play a relevant role in these processes. This review compiles the recent information and evidence on the role of bacterial extracellular vesicles in brain pathologies and on the therapeutic utility of bacterial extracellular vesicles in neurological and neurodegenerative diseases. One advantage of bacterial extracellular vesicles compared to extracellular vesicles derived from other cell types, such as stem cells, is that bacterial extracellular vesicles are generally easier to produce and modify. Bacterial extracellular vesicles may be easily modified to target a specific pathology and/ or to enhance its therapeutic efficacy. Although the studies are still scarce, they open a wide field of possibilities for future studies, which will lead to a deeper understanding of the role of microbiota and bacterial extracellular vesicles in neurological pathologies and the underlying mechanisms, as well as to the development of new treatments based on the use of bacterial extracellular vesicles in neurological diseases.
    Related Articles | Metrics
    Exercise training promotes nerve cell repair and regeneration after spinal cord injury
    Tianyu Zhai , Shuting Ren , Shenghao Qian, Caizhen Shi, Bingbing Wang, Can Zhang, Li Dan, Juan Shen, Feng Gao, Yanling Yang, Youlei Li , Lin Zhao
    2026, 21 (6):  2186-2207.  doi: 10.4103/NRR.NRR-D-24-01677
    Abstract ( 82 )   PDF (24357KB) ( 24 )   Save
    Spinal cord injury is a severe neurological condition characterized by the permanent loss of nerve cell function and a failure in neural circuit reconstruction—key factors contributing to disability. Therefore, exploring effective strategies to promote the repair and regeneration of nerve cells after spinal cord injury is crucial for optimizing patient prognosis. The purpose of this paper is to conduct an in-depth review of the pathological changes in nerve cells after spinal cord injury and to present the state of research on the role of exercise training in promoting the repair and regeneration of nerve cells after spinal cord injury. In terms of the intrinsic growth capacity of neurons, disruptions in the dynamic balance between growth cones and the cytoskeleton, the dysregulation of transcription factors, abnormal protein signaling transduction, and altered epigenetic modifications collectively hinder axonal regeneration. Additionally, the microenvironment of neurons undergoes a series of complex changes, initially manifesting as edema, which may be exacerbated by spinal cord ischemiareperfusion injury, further increasing the extent of nerve cell damage. The abnormal proliferation of astrocytes leads to the formation of glial scars, creating a physical barrier to nerve regeneration. The inflammatory response triggered by the excessive activation of microglia negatively impacts the process of nerve repair. Non-invasive interventions involving exercise training have shown significant potential in promoting nerve repair as part of a comprehensive treatment strategy for spinal cord injury. Specifically, exercise training can reshape the growth cone and cytoskeletal structures of neurons, regulate transcription factor activity, modulate protein signaling pathways, and influence epigenetic modifications, thereby activating the intrinsic repair mechanisms of neurons. Moreover, exercise training can regulate the activation state of astrocytes, optimize the inflammatory response and metabolic processes, promote astrocyte polarization, enhance angiogenesis, reduce glial scar formation, and modulate the expression levels of nerve growth factors. It also effectively helps regulate microglial activation, promotes axonal regeneration, and improves phagocytic function, thereby optimizing the microenvironment for nerve repair. In terms of clinical translation, we summarize the preliminary results of new drug research and development efforts, the development of innovative devices, and the use of exercise training in promoting clinical advancements in nerve repair following spinal cord injury, while considering their limitations and future application prospects. In summary, this review systematically analyzes findings relating to the pathological changes occurring in nerve cells after spinal cord injury and emphasizes the critical role of exercise training in facilitating the repair and regeneration of nerve cells. This work is expected to provide new ideas and methods for the rehabilitation of patients with spinal cord injury.
    Related Articles | Metrics
    ErbB signaling in brain injury regeneration: Pathway interactions and therapeutic potential
    Patricia Pérez-García, Nora Martínez-Gómez, Sonia Vázquez-de Górgolas, Andrea Chamorro-Francisco, Ricardo Pardillo-Díaz, Pedro Nunez-Abades, Carmen Castro, Livia Carrascal
    2026, 21 (6):  2208-2220.  doi: 10.4103/NRR.NRR-D-25-00155
    Abstract ( 66 )   PDF (7414KB) ( 6 )   Save
    The ErbB signaling network has recently emerged as a key modulator of central nervous system responses to injury. This review provides a comprehensive overview of ErbB receptors and their ligands, highlighting canonical and non-canonical signaling mechanisms relevant to brain damage. We explore how ErbB signaling is dynamically regulated following injury and how it orchestrates processes such as neuroinflammation, gliosis, and neural repair. Special attention is given to its interplay with other critical pathways, including Notch signaling, and its roles within adult neurogenic niches, where it modulates neural stem cell behavior in response to damage. Based on accumulating preclinical evidence, we propose two therapeutic strategies for targeting ErbB signaling in brain injury: (1) dampening neuroinflammation through ErbB inhibition and (2) promoting neuroprotection and neurogenesis via neuregulin-1-mediated activation. The first strategy is supported by studies, which demonstrate that inhibition of ErbB1 limits neuroinflammation and supports neural repair in preclinical models. The latter strategy is supported by emerging studies demonstrating the significant potential of novel protein kinase C activating diterpenes in modulating ErbB signaling pathways through the regulation of neuregulin-1 release. Diterpenes, by influencing the ErbB pathway, may uniquely bridge the gap between neuroprotection and regeneration. Their potential to modulate inflammation and promote pro-regenerative cellular environments positions them as promising tools in the development of targeted therapies. By dissecting these mechanisms, we aim to shed light on the translational potential of ErbB-targeted therapies and their capacity to enhance endogenous repair processes in the injured brain.
    Related Articles | Metrics
    The role of gut microbiota–mitochondria crosstalk in neurodegeneration: Underlying mechanisms and potential therapies
    Tianjuan Ju, Yaoyuan Zhang, Lipeng Liu, Xitong Zhao, Xinwei Li, Changfeng Liu, Shukai Sun, Li-an Wu
    2026, 21 (6):  2221-2242.  doi: 10.4103/NRR.NRR-D-24-01419
    Abstract ( 88 )   PDF (22589KB) ( 43 )   Save
    Emerging evidence suggests that the gut microbiota is closely associated with the pathological manifestations of multiple neurodegenerative diseases via the gut-brain axis, which refers to the crosstalk between the gut and the central nervous system. More importantly, mitochondria have been considered prominent mediators of the interplay between the gut microbiota and the brain. Intestinal microbes may modulate mitochondrial function in the central nervous system to affect the progression of neurodegenerative diseases. Mitochondria are essential for meeting the host’s substantial neuronal metabolic demands, maintaining excitability, and facilitating synaptic transmission. Dysfunctional mitochondria are considered critical hallmarks of various neurodegenerative diseases. Therefore, this review provides novel insights into the intricate roles of gut microbiota-mitochondrial crosstalk in the underlying mechanisms during the progression of neurodegeneration, as well as the existing potential therapeutic strategies for neurodegenerative disorders. These suggest intestinal microbiota-mitochondrial interaction play a crucial role in the occurrence and development of neurodegenerative diseases, and targeting this interaction may be a promising therapeutic approach to neurodegenerative diseases. However, this review found that there was relatively little research on the effect of this crosstalk on other neurodegenerative diseases, such as Huntington’s disease and Multiple sclerosis, and the potential therapeutic strategies were translated into clinical trials, which face many challenges in developing personalized treatment plans based on the unique gut microbiota of different individuals
    Related Articles | Metrics
    DNAJB6: A guardian against neurodegeneration
    Jónvá Hentze , Anna Gelman, Tomasz Brudek, Christian Hansen
    2026, 21 (6):  2243-2253.  doi: 10.4103/NRR.NRR-D-24-01504
    Abstract ( 67 )   PDF (3333KB) ( 92 )   Save
    Amyloid protein aggregation plays a major role in multiple neurodegenerative diseases and is likely the primary driving force for the progression of most of these diseases. Multiple recent studies have highlighted that the DNAJ homolog subfamily B member 6 (DNAJB6) chaperone is particularly interesting, when it comes to preventing amyloidogenic proteins from aggregating. It has been shown that DNAJB6 can prevent the aggregation of polyglutamine-expanded proteins in models of Huntington’s disease. Likewise, it can suppress aggregation of α-synuclein in models of Parkinson’s disease and other synucleinopathies. Finally, it has been shown that DNAJB6 can block aggregation of multiple additional amyloid proteins involved in Alzheimer’s disease and other tauopathies as well. We believe there is yet much to learn about the protective role of DNAJB6 in the brain, but this focused review summarizes, what we know so far of this chaperone. It describes the biological role of DNAJB6 in the brain and its interaction with Hsp70, with particular emphasis on the studies that show its ability to prevent amyloid protein aggregation in vitro and in vivo. Moreover, recent work on dysregulation of the expression of DNAJB6 in brain clinical tissue is discussed. Finally, we discuss potential therapeutic perspectives as we believe this protein is a promising druggable target.
    Related Articles | Metrics
    Nanomedicines and stroke: Advantages in chronic inflammation treatment and neural regeneration
    Chuhan Liu, Yuanyuan Ran, Changbin Hu, Mengjie Wang, Ning Li, Zhi Yang, Zitong Ding, Chenye Qiao, Jianing Xi, Wei Su, Lin Ye, Zongjian Liu
    2026, 21 (6):  2254-2274. 
    Abstract ( 54 )   PDF (9142KB) ( 10 )   Save
    Stroke can be categorized as ischemic and hemorrhagic on the basis of its origin. The pathophysiology following a stroke is complex, and is characterized by ongoing inflammation, neuronal injury, and the accumulation of reactive oxygen species in the brain, all of which reflect a dynamic process of change. This complexity hinders achievement of significant therapeutic outcomes with standard stroke treatment procedures, limiting post-stroke recovery. This review presents an innovative poststroke therapeutic approach that utilizes nanomedicines to modify the cerebral microenvironment. It highlights the primary roles of chronic inflammation and nerve repair issues in causing prolonged impairment in stroke patients. Traditional therapies show limited effectiveness in achieving neuroprotection, immunoregulation, and neural regeneration during the subacute and chronic phases of stroke. Therefore, effective stroke management requires the use of specific therapeutic strategies tailored to the pathological characteristics of each phase. Various types of nanomedicines possess distinct physicochemical properties and can be selected on the basis of the specific therapeutic needs. Surface-modification technologies have significantly enhanced the ability of nanomedicines to penetrate the blood–brain barrier and improve their targeting capabilities in drug administration. However, the stability, biocompatibility, and long-term safety of nanomedicines require further optimization for clinical application. Nanomedicines represent a novel approach to stroke treatment through targeted delivery and multifaceted regulatory mechanisms. These medicines provide distinct advantages, particularly in addressing chronic inflammation and promoting nerve regeneration. As a result, nanomedicines are expected to significantly improve rehabilitation outcomes and quality of life for stroke patients in the future, emerging as a crucial modality for stroke treatment.
    Related Articles | Metrics
    Low-intensity transcranial ultrasound neuromodulation promotes neuronal regeneration: A new hope for noninvasive treatment of neurodegenerative diseases
    Shu Xia, Chen He, Yunfei Li, Hao Li, Bo Wang, Long Xu, Xudong Zhao
    2026, 21 (6):  2275-2294.  doi: 10.4103/NRR.NRR-D-25-00113
    Abstract ( 165 )   PDF (6779KB) ( 84 )   Save
    Neurodegenerative diseases, which are characterized by progressive neuronal loss and the lack of disease-modifying therapies, are becoming a major global health challenge. The existing neuromodulation techniques, such as deep brain stimulation and transcranial magnetic stimulation, show limitations such as invasiveness, restricted cortical targeting, and irreversible tissue effects. In this context, low-intensity transcranial ultrasound has emerged as a promising noninvasive alternative that can penetrate deep into the brain and modulate neuroplasticity. This review comprehensively assesses the therapeutic mechanisms, efficacy, and translational potential of low-intensity transcranial ultrasound in treating neurodegenerative diseases, with emphasis on its role in promoting neuronal regeneration, modulating neuroinflammation, and enhancing functional recovery. We summarize the findings of previous studies and systematically illustrate the potential of low-intensity transcranial ultrasound in regulating cell death mechanisms, enhancing neural repair and regeneration, and alleviating symptoms associated with neurodegenerative diseases. Preclinical findings indicate that low-intensity transcranial ultrasound can enhance the release of neurotrophic factors (e.g., brainderived neurotrophic factor), promote autophagy to clear protein aggregates, modulate microglial activation, and temporarily open the blood–brain barrier to facilitate targeted drug delivery. Existing clinical trial data show that low-intensity transcranial ultrasound can reduce amyloid-β plaques, improve motor and cognitive deficits, and promote remyelination in various disease models. Early clinical trials suggest that low-intensity transcranial ultrasound may enhance cognitive scores in Alzheimer’s disease and alleviate motor symptoms in Parkinson’s disease, all while demonstrating a favorable safety profile. Past studies support the notion that by integrating safety, precision, and reversibility, low-intensity transcranial ultrasound can transform the treatment landscape for neurodegenerative disease. However, more advancements are necessary for future clinical application of low-intensity transcranial ultrasound, including optimizing parameters such as frequency, intensity, and duty cycle; considering individual anatomical differences; and confirming long-term efficacy. We believe establishing standardized protocols, conducting larger trials, and investigating the underlying mechanisms to clarify dose-response relationships and refine personalized application strategies are essential in this regard. Future research should focus on translating preclinical findings into clinical practice, addressing technical challenges, and exploring combination therapies with pharmacological or gene interventions.
    Related Articles | Metrics
    Regulatory role of neuronal guidance proteins in spinal cord injury
    Linyan Tang, Zhi Song, Jie Wang, Shenhua He, Chao Liu
    2026, 21 (6):  2295-2305.  doi: 10.4103/NRR.NRR-D-24-00564
    Abstract ( 100 )   PDF (2534KB) ( 25 )   Save
    Spinal cord injury is a severe neurological condition with limited neuronal regeneration and functional recovery. Currently, no effective treatments exist to improve spinal cord injury prognosis. Neuronal guidance proteins are a diverse group of molecules that play crucial roles in axon and dendrite growth during nervous system development. Increasing evidence highlights their regulatory functions in spinal cord injury. This review provides a brief overview of the modulation patterns of key neuronal guidance proteins in neuronal axon growth during nervous system formation and subsequently focuses on their roles in neuronal regeneration and functional recovery following spinal cord injury. Neuronal guidance proteins include, but are not limited to, semaphorins and their receptors, plexins; netrins and their receptors, deleted in colorectal cancer and UNC5; Eph receptors and their ligands, ephrins; Slit and its receptor, Robo; repulsive guidance molecules and their receptor, neogenin; Wnt proteins and their receptor, Frizzled; and protocadherins. Localized Netrin-1 at the injury site inhibits motor axon regeneration after adult spinal cord injury while promoting oligodendrocyte growth. Slit2 enhances synapse formation in the injured spinal cord of rats. EphA7 regulates acute apoptosis in the early pathophysiological stages of spinal cord injury, while ephrinA1 plays a role in the nervous system’s injury response, with its reduced expression leading to impaired motor function in rats. EphA3 is upregulated following spinal cord injury, promoting an inhibitory environment for axonal regeneration. After spinal cord injury, bidirectional activation of ephrinB2 and EphB2 in astrocytes and fibroblasts results in the formation of a dense astrocyte-meningeal fibroblast scar. EphB1/ephrinB1 signaling mediates pain processing in spinal cord injury by regulating calpain-1 and caspase-3 in neurons. EphB3 expression increases in white matter after spinal cord injury, further inhibiting axon regeneration. Sema3A, expressed by neurons and fibroblasts in the scar surrounding the injury, inhibits motor neuron and sensory nerve growth after spinal cord injury. Sema4D suppresses neuronal axon myelination and axon regeneration, while its inhibition significantly enhances axon regeneration and motor recovery. Sema7A is involved in glial scar formation and may influence serotonin channel remodeling, thereby affecting motor coordination. Given these findings, the local or systemic application of neuronal guidance proteins represents a promising avenue for spinal cord injury treatment.
    Related Articles | Metrics
    Synaptic mitochondria in aging and neurodegenerative diseases: Functional decline and vulnerability
    Karina A. Cicali, Angie K. Torres, Cheril Tapia-Rojas
    2026, 21 (6):  2306-2317.  doi: 10.4103/NRR.NRR-D-24-01571
    Abstract ( 101 )   PDF (2026KB) ( 82 )   Save
    Aging is a physiological and complex process produced by accumulative age-dependent cellular damage, which significantly impacts brain regions like the hippocampus, an essential region involved in memory and learning. A crucial factor contributing to this decline is the dysfunction of mitochondria, particularly those located at synapses. Synaptic mitochondria are specialized organelles that produce the energy required for synaptic transmission but are also important for calcium homeostasis at these sites. In contrast, non-synaptic mitochondria primarily involve cellular metabolism and long-term energy supply. Both pools of mitochondria differ in their form, proteome, functionality, and cellular role. The proper functioning of synaptic mitochondria depends on processes such as mitochondrial dynamics, transport, and quality control. However, synaptic mitochondria are particularly vulnerable to age-associated damage, characterized by oxidative stress, impaired energy production, and calcium dysregulation. These changes compromise synaptic transmission, reducing synaptic activity and cognitive decline during aging. In the context of neurodegenerative diseases such as Alzheimer’s, Parkinson’s, and Huntington’s, the decline of synaptic mitochondrial function is even more pronounced. These diseases are marked by pathological protein accumulation, disrupted mitochondrial dynamics, and heightened oxidative stress, accelerating synaptic dysfunction and neuronal loss. Due to their specialized role and location, synaptic mitochondria are among the first organelles to exhibit dysfunction, underscoring their critical role in disease progression. This review delves into the main differences at structural and functional levels between synaptic and nonsynaptic mitochondria, emphasizing the vulnerability of synaptic mitochondria to the aging process and neurodegeneration. These approaches highlight the potential of targeting synaptic mitochondria to mitigate age-associated cognitive impairment and synaptic degeneration. This review emphasizes the distinct vulnerabilities of hippocampal synaptic mitochondria, highlighting their essential role in sustaining brain function throughout life and their promise as therapeutic targets for safeguarding the cognitive capacities of people of advanced age.
    Related Articles | Metrics
    Cell apoptosis in ischemic stroke: Focus on lipid metabolism
    Rong Sun, Wenren Yang, Yuting Zhao, Fumei Zhang, Genping Wu, Aiping Wang, Ying Tian
    2026, 21 (6):  2315-2329.  doi: 10.4103/NRR.NRR-D-24-00301
    Abstract ( 171 )   PDF (2608KB) ( 34 )   Save
    Ischemic stroke is a severe neurological disease with high global mortality and disability rates. Atherosclerosis has been identified as the primary cause of ischemic stroke, while abnormal lipid levels are significant contributors to the development of this condition. Multiple pro-apoptotic mechanisms are involved in ischemic stroke caused by lipid metabolism disorders, while various lipids have a strong causal relationship with neuronal apoptosis. However, studies to date have focused on the individual roles of lipid metabolism and apoptosis in ischemic stroke, and an overview of how impaired lipid metabolism leads to apoptosis in ischemic stroke is still lacking in the literature. In this review, we summarize current research on lipids in ischemic stroke. We discuss the role of lipid metabolism in accelerating apoptosis in ischemic stroke as well as the associated mechanisms. Additionally, we highlight advances in drug development and the treatment of stroke, focusing on lipid metabolism. The purpose is to provide novel ideas and strategies for the treatment and prevention of ischemic stroke.
    Related Articles | Metrics
    Neuromodulatory role and therapeutic potential of N6 -methyladenosine RNA methylation in neurodegenerative diseases
    Jinyu Zhang, Wenjing Ma, Ranxu Liu, Xiaoheng Li, Zengqiang Yuan, Jinbo Cheng
    2026, 21 (6):  2330-2349.  doi: 10.4103/NRR.NRR-D-24-01648
    Abstract ( 88 )   Save
    N6 -methyladenosine RNA methylation, an essential post-transcriptional modification, dynamically regulates RNA metabolism and plays a crucial role in neuronal function. Growing evidence suggests that dysregulated N6 -methyladenosine modification contributes to the pathogenesis of neurodegenerative diseases, including Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, and amyotrophic lateral sclerosis. However, the precise mechanisms by which N6 -methyladenosine modification influences these conditions remain unclear. This review summarizes the role of m6 A modification and its associated regulators in neurodegeneration, focusing on their involvement in key pathological processes. In Alzheimer’s disease, m6 A modification contributes to synaptic dysfunction, mitochondrial damage, and neuronal apoptosis. Evidence from APP/PS1, 5XFAD, tau transgenic, and Drosophila models demonstrates that regulators such as methyltransferase-like 3 and fat mass and obesityassociated protein influence Alzheimer’s disease progression through neuroinflammation, circRNA dysregulation, and autophagy-related mechanisms. In Parkinson’s disease, altered N6 -methyladenosine regulator expression affects dopaminergic neuron survival and stress responses by modulating mRNA stability and autophagy-related lncRNAs. In multiple sclerosis and amyotrophic lateral sclerosis, N6 - methyladenosine affects immune activation, myelin repair, and the regulation of disease-associated genes such as TDP-43. Beyond N6 -methyladenosine, other RNA methylation modifications—such as m1 A, m5 C, m7 G, uracil, and pseudouridine—are implicated in neurodegenerative diseases through their regulation of mitochondrial function, RNA metabolism, and neuronal stress responses. Additionally, N6 - methyladenosine exhibits cell type–specific functions: in microglia, it regulates inflammatory activation and phagocytic function; in astrocytes, it modulates metabolic homeostasis and glutamate-associated neurotoxicity; in neurons, it affects synaptic function and neurodegeneration-related gene expression; and in adult neural stem cells, it controls differentiation, neurogenesis, and cognitive plasticity. Recently, several small-molecule inhibitors targeting methyltransferase-like 3 or fat mass and obesityassociated protein have been developed to modulate N6 -methyladenosine modification, providing new opportunities for disease intervention, with the targeting of N⁶-methyladenosine-related pathways emerging as a promising therapeutic strategy. However, challenges persist in optimizing the specificity and delivery of these therapeutic approaches.
    Related Articles | Metrics
    The role of autophagy in spinal cord injury: Mechanisms, crosstalk, and therapeutic strategies
    Rui Wang, Zhen Niu, Runze Tian, Aini Chen, Huangmei Liao, Rui Kuang, Ying Feng, Guangyu Chin, Jiesheng Xie, Ping Zhu, Chi Teng Vong, Ge Li
    2026, 21 (6):  2350-2369.  doi: 10.4103/NRR.NRR-D-24-01467
    Abstract ( 83 )   PDF (2871KB) ( 138 )   Save
    Spinal cord injury is a neurological disorder resulting from trauma, typically affecting sensory and motor function at the injury site, even leading to paralysis and internal dysfunction. The treatment of spinal cord injury mainly relies on pharmacological and surgical interventions; however, significant challenges remain in the protection and repair of neural tissues. Autophagy, an intracellular process responsible for the degradation and recycling of macromolecular components, plays a vital role in spinal cord injury, alleviating the severity of injury by inhibiting cell apoptosis and inflammatory responses. In this review, we provide an overview of the physiological mechanisms underlying autophagy and spinal cord injury and detail the crosstalk between autophagy and other modes of cell death in spinal cord injury. In addition, we discuss the potential of targeting autophagy as a therapeutic strategy for spinal cord injury through approaches that focus on promoting or inhibiting this process, targeting specific autophagic substrates or pathways, and combining autophagy modulation with other neuroprotective or restorative interventions. In summary, this review proposes that strict regulation of autophagy may represent a viable strategy for the treatment of spinal cord injury.
    Related Articles | Metrics
    Metabolic breakdown: Linking insulin resistance and mitochondrial dysfunction to neurodegeneration in Alzheimer’s disease
    Simona Lanzillotta, Lucrezia Romana Rolfi , Barbara Zulli , Eugenio Barone
    2026, 21 (6):  2370-2383.  doi: 10.4103/NRR.NRR-D-25-00144
    Abstract ( 64 )   PDF (2307KB) ( 29 )   Save
    The increasing prevalence of metabolic disorders and neurodegenerative diseases has uncovered shared pathophysiological pathways, with insulin resistance and mitochondrial dysfunction emerging as critical contributors to cognitive decline. Insulin resistance impairs neuronal metabolism and synaptic function, fostering neurodegeneration as observed in Alzheimer’s disease and Down syndrome. Indeed, Down syndrome, characterized by the triplication of the APP gene, represents a valuable genetic model for studying early-onset Alzheimer’s disease and accelerated aging. Building on the link between metabolic dysfunctions and neurodegeneration, innovative strategies addressed brain insulin resistance as a key driver of cognitive decline. Intranasal insulin has shown promise in improving cognition in early Alzheimer’s disease and type 2 diabetes, supporting the concept that restoring insulin sensitivity can mitigate neurodegeneration. However, insulin-based therapies risk desensitizing insulin signaling, potentially worsening the disease. Incretins, particularly glucagonlike peptide 1 receptor agonists, offer neuroprotective benefits by enhancing insulin sensitivity, metabolism, and synaptic plasticity while reducing oxidative distress and neuroinflammation. This review focuses on current knowledge on the metabolic and molecular interactions between insulin resistance, mitochondrial dynamics (including their roles in energy metabolism), and oxidative distress regulation, as these are pivotal in both Alzheimer’s disease and Down syndrome. By addressing these interconnected mechanisms, innovative treatments may emerge for both metabolic and neurodegenerative disorders.
    Related Articles | Metrics
    Dual role of microglia in glaucoma: Regulation of neuroinflammation and neuroregeneration
    Panpan Li, Xin Shi, Verena Prokosch
    2026, 21 (6):  2384-2394.  doi: 10.4103/NRR.NRR-D-24-00876
    Abstract ( 80 )   PDF (2165KB) ( 32 )   Save
    Globally, glaucoma stands as a primary cause of irreversible blindness, marked by intricate pathophysiological processes in which neuroinflammation plays a pivotal role. As the principal immune cells within the central nervous system, microglia play a dual function in the progression of glaucoma. Under standard physiological states, microglia safeguard the retina by offering neurotrophic support and removing cellular debris. In the pathological progression of glaucoma, microglia become activated and release significant levels of inflammatory factors, resulting in retinal ganglion cell injury, cell death, and impaired neuroregeneration. This review focuses on examining the dual functions of microglia in glaucoma, evaluating their influence on retinal neurodegeneration and repair, and suggesting that modulating microglial activity could serve as a promising therapeutic strategy. Understanding the mechanisms of microglial action in glaucoma is crucial for unveiling the complex pathophysiological processes of the disease and developing new therapeutic strategies.
    Related Articles | Metrics
    Link between blood–brain barrier disruption and microglial activation
    Arjun Sapkota, Sebok K. Halder, Richard Milner
    2026, 21 (6):  2395-2396.  doi: 10.4103/NRR.NRR-D-25-00103
    Abstract ( 111 )   PDF (873KB) ( 34 )   Save
    Cells of the central nervous system (CNS) are privileged in lying behind the blood–brain barrier (BBB). Unlike blood vessels in other organs, CNS blood vessels are unique in displaying high electrical resistance and low permeability. With this unique structure and function, the BBB prevents potentially harmful blood components such as serum proteins, inflammatory cytokines, and inflammatory leukocytes from entering the hallowed space of the CNS and wreaking havoc. In addition to these “tightness” properties, the BBB has an array of specialized transporters designed to import essential nutrients, such as amino acids and glucose into the CNS. It also has transporters that remove unwanted chemicals or drugs from the CNS called multidrug resistance proteins, such as P-glycoprotein. At the structural level, the BBB consists of a lining of endothelial cells firmly attached to a basement membrane (BM) containing high levels of the extracellular matrix (ECM) proteins collagen IV, laminin, fibronectin, and perlecan. Pericytes and astrocyte foot processes also play an important role in inducing and maintaining BBB properties. At the molecular level, the BBB relies on adhesion molecules (primarily integrins) that bind endothelial cells to the BM as well as adherens and tight junction protein complexes that form between adjacent endothelial cells (Zlokovic, 2008). The importance of the BBB is illustrated by the fact that its disruption is instrumental in the initiation and/or maintenance in almost all neurological diseases, including ischemic stroke, multiple sclerosis, vascular dementia, and Alzheimer’s disease. Aside from deteriorating during disease conditions, accumulating evidence suggests that BBB integrity also declines as a function of age (Senatorov et al., 2019).
    Related Articles | Metrics
    Frontal synaptic plasticity: A new key to homeostatic sleep regulation
    Yusuke Iino , Shoi Shi
    2026, 21 (6):  2397-2398.  doi: 10.4103/NRR.NRR-D-25-00231
    Abstract ( 76 )   PDF (664KB) ( 84 )   Save
    Sleep is a fundamental biological process essential for maintaining brain function, cognitive performance, and overall health. Despite over a century of research, the mechanisms underlying sleep homeostasis—the process by which the need for sleep accumulates during wakefulness and dissipates during sleep—remain incompletely understood. This article explores the latest advancements in sleep research, focusing on the role of synaptic plasticity in sleep homeostasis, as illuminated by Sawada et al. (2024).
    Related Articles | Metrics
    Decoding microglial aging through multi-model approaches
    Martin Škandík , Bertrand Joseph
    2026, 21 (6):  2399-2400.  doi: 10.4103/NRR.NRR-D-25-00229
    Abstract ( 65 )   PDF (2269KB) ( 27 )   Save
    In recent years, rising life expectancy has led to a significant increase in the prevalence of neurodegenerative disorders, including Alzheimer’s disease (AD), Parkinson’s disease, and age-related cognitive decline. Additionally, other neurological conditions such as glioblastoma, the most common and aggressive brain tumor in adults have been more frequently reported in aging populations. The brain itself is highly vulnerable to age-related changes, particularly disruptions in homeostatic regulation, which further contribute to its functional decline and heightened susceptibility to disease. This has led to a surge of interest in understanding the cellular and molecular mechanisms driving these changes, with a heightened focus directed at microglia, the resident immune sentinels of the brain. Microglia under physiological conditions actively survey the brain environment, clear invading pathogens, remove dead or dying neurons, and promote synaptic remodeling and neuroprotection. They support neuronal functions and overall brain activity in close cooperation with other glial cells, particularly astrocytes, involved in synaptic signal transmission (Malvaso et al., 2023). However, microglia exhibit significant heterogeneity between species and even between brain regions that must be considered when interpreting research findings.
    Related Articles | Metrics
    NLRP3 inflammasome: A link between systemic infection and Alzheimer’s disease
    Tatiana Barichello , Felipe Dal-Pizzol
    2026, 21 (6):  2401-2402.  doi: 10.4103/NRR.NRR-D-25-00073
    Abstract ( 59 )   PDF (6358KB) ( 44 )   Save
    Neuroinflammation is a crucial factor in the progression of various diseases, ranging from immune-related conditions such as sepsis to neurodegenerative disorders such as Alzheimer’s disease (AD) (Ravichandran and Heneka, 2024). This perspective article, which draws on insights from diverse fields including neuroscience, i m m u n o l o g y, a n d p at h o l o g y, p ro v i d e s a critical analysis of ongoing research efforts in inflammasome biology, with specific emphasis on Nod-like receptor (NLR) and pyrin domaincontaining protein 3 (NLRP3). This article takes an interdisciplinary approach, placing emphasis on the relationship between sepsis and AD, in an attempt to advance our understanding of these multifaceted diseases and the role of infection on neuroinflammation and AD pathology.
    Related Articles | Metrics
    Dynamic regulation of the developmental establishment of the adult hippocampal neural stem cell pool
    Feng Zhang, Guo-li Ming, Hongjun Song
    2026, 21 (6):  2403-2404.  doi: 10.4103/NRR.NRR-D-24-01581
    Abstract ( 88 )   PDF (1070KB) ( 67 )   Save
    The adult subventricular zone of the lateral ventricles and the subgranular zone in the hippocampal dentate gyrus (DG) are the two brain regions where neurogenesis occurs throughout life in the adult mammalian brain (Ming and Song, 2011). Adult quiescent hippocampal neural stem cells (NSCs) are bona fide stem cells and, when activated, give rise to newborn granule neurons in the adult brain, which play vital roles in learning, memory, mood, and affective cognition (Bonaguidi et al., 2011; Ming and Song, 2011). Dysregulation of this process is often associated with brain disorders in both patients and various animal models (Christian et al., 2014). The proper establishment of a quiescent adult NSC pool is essential to sustain life-long continuous neurogenesis (Urban et al., 2019). Clonal lineagetracing in mice has identified a common neural precursor population that originates from the dentate neuroepithelium and migrates to primitive DG during the embryonic stage, and continuously contributes to the generation of dentate granule neurons from early embryonic stages to adulthood (Berg et al., 2019; Figure 1A). Furthermore, a subpopulation of proliferating DG NSCs undergoes a transition to quiescence during the early postnatal stage and adopts adult NSC-like quiescent properties to establish the quiescent adult DG NSC pool in mice (Berg et al., 2019; Bond et al., 2021; Figure 1A). Importantly, disturbances in the quiescence acquisition and fate determination of DG NSCs are closely associated with abnormal postnatal DG neurogenesis and neuronal circuitry formation during development, which would impair hippocampal function (Zhang et al., 2023, 2024; Jimenez-Cyrus et al., 2024). A better understanding of the fundamental mechanisms underlying adult neurogenesis and the establishment of a quiescent NSC pool is necessary to harness this striking intrinsic regenerative capacity in the adult mammalian brain for regeneration and repair. While the mechanisms regulating the activation and quiescence maintenance of adult DG NSCs have been widely investigated (Urban et al., 2019), research into cellular and molecular mechanisms governing the establishment of the adult NSC pool during development remains in its infancy.
    Related Articles | Metrics
    Energy for myelination: Implications for metabolic disturbances in multiple sclerosis pathology
    Milton Guilherme Forestieri Fernandes, Jack P. Antel, Timothy E. Kennedy
    2026, 21 (6):  2405-2406.  doi: 10.4103/NRR.NRR-D-24-01570
    Abstract ( 81 )   PDF (394KB) ( 62 )   Save
    Myelin, made by oligodendrocytes (OLs) in the central nervous system (CNS), is essential for neural transmission. In particular, myelin facilitates communication across the long connections between different brain regions that form the white matter. Myelinated segments also provide metabolic intermediates to axons, supporting their demanding energetic needs. Genetic disorders that disrupt myelin formation result in progressive neurologic degeneration, referred to as leukodystrophies. Multiple sclerosis (MS) is considered an acquired disease, reflecting both genetic and environmental factors.
    Related Articles | Metrics
    Proteolytic shedding of the prion protein: Uncovering “new” biological implications of a conserved cleavage event
    Feizhi Song, Valerija Kovac, Behnam Mohammadi, Josephine E. Pippi, Vladka Curin Serbec, Markus Glatzel, Hermann C. Altmeppen
    2026, 21 (6):  2407-2408.  doi: 10.4103/NRR.NRR-D-25-00013
    Abstract ( 76 )   PDF (610KB) ( 17 )   Save
    Novel insights into complex biological processes very often critically depend on the establishment of new potent read-out tools and improved protocols. A lot has been learned over the past four decades on physiological functions and, importantly, disease-related roles of the prion protein (PrP), a relatively broadly expressed membrane-anchored glycoprotein with high levels in several cell types of the nervous and immune system and with well-established key roles in different progressive and fatal neurodegenerative protein misfolding diseases (proteopathies). However, while several controversies and unclarities remain even for these widely accepted involvements, currently unexplored (and unexpected) facets and functions may still wait to be discovered. New light might be shed into these aspects by a better understanding of potential intrinsic roles of previously largely unconsidered post-translationally generated forms or fragments of PrP, for instance those resulting from endogenous proteolytic cleavage (Mohammadi et al., 2022; Vanni et al., 2022). In fact, membranebound full-length (FL) PrP, the form most research of the past has focused on, may not even represent the majority of total PrP in the brain (Vanni et al., 2022). A nearly FL form released from cells by a constitutive and very membrane-proximate proteolytic cleavage event (“shedding”) makes up for a rather small yet relevant fraction and is of emerging interest. This physiological, anchorless, and predominantly double-glycosylated form, now called “shed PrP” (sPrP), has repeatedly been reported in the past (e.g., Parizek et al., 2001), for instance, in cell culture media supernatants or body fluids, yet its mechanistic origin and biological relevance remained obscure for a long time. The latter, to a great deal, is due to technical challenges differentiating this fragment from excess FL-PrP present in most biological specimens (e.g., tissue homogenates; but even in body fluids or cell culture supernatants, FLPrP is present on cellular membrane debris and physiologically released extracellular vesicles (EVs)). Both forms are of similar molecular weight and share structure and sequence and, hence, epitopes for most available antibodies used for detection in standard laboratory techniques (Mohammadi et al., 2022). Besides recently improved protocols to differentiate and quantify the abundance of different PrP “proteoforms” by immunoblotting (Vanni et al., 2022), cleavage site-directed antibodies previously presented for the reliable detection of rodent sPrP have become a convenient tool to systematically, highly specifically, and comparably comfortably assess sPrP with various methods, as these antibodies are “blind” for the just few amino acids (plus the C-terminally attached glycosylphosphatidylinositol anchor) longer FL form (Linsenmeier et al., 2021; Mohammadi et al., 2022). However, given that many key aspects regarding PrP shedding, such as cleavage site and responsible protease, remained uncharacterized for the human body, our groups recently set out to unravel many unknowns in this regard. We eventually succeeded in identifying the shedding site (Y226↓Q227) in human PrP and recently presented an in-depth characterization of respective antibodies exclusively detecting sPrP (Song et al., 2024). As expected from previous mouse data, we revealed that the shedding of human PrP is likewise strictly dependent on the metalloprotease ADAM10, and we did not find any evidence for alternative proteolytic cleavages in the vicinity of Y226 (Song et al., 2024).
    Related Articles | Metrics
    Improving recovery from traumatic spinal cord injury: Targeting remyelination versus white matter remodeling
    Bethany R. Kondiles , Wolfram Tetzlaff
    2026, 21 (6):  2409-2410.  doi: 10.4103/NRR.NRR-D-24-01628
    Abstract ( 62 )   PDF (979KB) ( 17 )   Save
    The inter-related pathological cascades following a traumatic spinal cord injury (tSCI) disrupt multiple cell types and physiological processes. Subsequently, motor and sensory functions are disrupted by breakdowns in cellular interactions and circuitry. Therapeutic interventions seek to modify some aspects of the injury course to enable the re-establishment of functional circuitry. Interventions often target one cell type (e.g., promoting neuroprotection or neural regeneration) or one process (e.g., modulating inflammation, affecting astrocytic, microglial, or macrophage responses.) Many axons in the spinal cord are myelinated, and after injury oligodendrocyte death causes demyelination. Promoting remyelination of spared or new axons to re-establish conduction seems a logical choice as a therapeutic target. However, “remyelination” refers to a binary process: the presence or absence of myelin regeneration. “White matter remodeling” considers the plasticity of and interactions between the multiple cell types essential for signal conduction. As the field develops more combinatorial approaches, wherein interventions target multiple cell types and processes, incorporating the concept of white matter remodeling, as opposed to remyelination, considers how to re-establish the requisite cellular circuitry necessary for regaining function.
    Related Articles | Metrics
    Glia and semaphorins in neurodegenerative diseases: The frontier for new therapeutics
    Sofia Nutarelli, Claudia Palazzo, Maria Teresa Viscomi
    2026, 21 (6):  2411-2412.  doi: 10.4103/NRR.NRR-D-25-00223
    Abstract ( 65 )   PDF (472KB) ( 27 )   Save
    The optimal development, function, and maintenance of the central nervous system (CNS) are determined by the dynamic and continuous crosstalk between its components. Neurons and glial cells, the cellular constituents of the CNS, orchestrate a wide range of essential activities (Allen and Lyons, 2018). Notably, glial cells, which outnumber neurons, constitute the major population within the CNS. This population comprises astrocytes, microglia, oligodendrocytes, and ependymal cells, each fulfilling specialized functions that contribute to neural homeostasis and overall CNS integrity. Astrocytes are pivotal in preserving structural and functional integrity through the regulation of synaptic function, the clearance of neurotransmitters, and ion balance. Moreover, they provide metabolic support to neurons. Microglia are resident immune cells that provide continuous surveillance within the CNS, regulating brain development and maintenance of neuronal networks. Oligodendrocytes are responsible for myelination, thereby modulating the speed of action potential conduction and optimizing neural communication. Lastly, ependymal cells, which form the epithelial lining of the brain’s ventricular system, are essential for the production of cerebrospinal fluid, its regulation, and the clearance of waste, thus playing a pivotal role in brain metabolism.
    Related Articles | Metrics
    Three-dimensional patientderived cell models represent an emerging frontier in the study of neurodegenerative diseases
    Rachel J. Boyd, Vasiliki Mahairaki
    2026, 21 (6):  2413-2414.  doi: 10.4103/NRR.NRR-D-25-00178
    Abstract ( 84 )   PDF (589KB) ( 33 )   Save
    Neurodegenerative disorders represent an increasingly pertinent public health crisis. As a greater proportion of the population ages, neurodegenerative disorders and other diseases of aging place undue burdens on patients, caregivers, and healthcare workers. Alzheimer’s disease (AD) and Parkinson’s disease represent the two most common neurodegenerative disorders in the population, affecting over 65 million people, worldwide. These diseases and others often arise due to complex interactions between polygenic risk and environmental exposures, which is often reflected in variable clinical presentation and response to therapeutic interventions.
    Related Articles | Metrics
    NAD substrate deficiency is an inherent and targetable risk factor for late-onset Alzheimer’s disease
    Kai-Christian Sonntag , Bruce M. Cohen
    2026, 21 (6):  2415-2416.  doi: 10.4103/NRR.NRR-D-25-00048
    Abstract ( 49 )   PDF (2004KB) ( 27 )   Save
    Sporadic or late-onset Alzheimer’s disease (LOAD) occurs in 1 of 10 people over 65 years of age and comprises 95% of all AD patients. Unlike earlyonset AD, which is caused by defined single gene mutations, the mechanisms and events underlying risk for LOAD are not fully understood and no substantial disease-modifying interventions are currently available. Age is the most prominent risk factor for LOAD, and interacting age-related and LOAD-associated factors contribute to its pathogenesis. Among these factors are changes in bioenergetic cell functions, which metabolize substrates and produce energy stored in adenosine triphosphate. Our findings and the work of others have suggested that disturbances of these bioenergetic functions are both inherent and acquired during the aging process and contribute to LOAD dementia later in life (Ryu et al., 2021a, b; Cohen and Sonntag, 2024). Identifying these abnormal bioenergetic functions may lead to the development of agents or approaches to reduce the risk for LOAD.
    Related Articles | Metrics
    Shifting focus to preclinical stages: Locus coeruleus tau pathology as a driver and therapeutic target in Alzheimer’s disease
    Qi Yuan , Tamunotonye Omoluabi, Brandon F. Hannam
    2026, 21 (6):  2417-2418.  doi: 10.4103/NRR.NRR-D-25-00140
    Abstract ( 59 )   PDF (1556KB) ( 9 )   Save
    Alzheimer’s disease (AD) remains an incurable neurodegenerative disorder with devastating societal and personal impacts. Despite decades of intensive research, therapeutic efforts targeting the clinical stages of AD have largely failed to halt or reverse disease progression. This has prompted a critical shift in focus toward the earlier, preclinical stages of AD, where interventions may hold greater promise for altering the disease trajectory. Theoretical frameworks of preclinical AD, such as those proposed by Sperling et al. (2011), describe a continuum spanning over a decade or more, characterized by three progressive stages: asymptomatic amyloidosis, the onset of neurodegeneration, and subtle cognitive impairments. While this model emphasizes amyloid-β as the initiating pathology, mounting evidence challenges this amyloid-centric paradigm, positioning early tau pathology as a primary driver of early neurodegenerative changes.
    Related Articles | Metrics
    Targeting gangliosides to treat Alzheimer’s and Parkinson’s diseases: A disruptive approach with the first-in-class peptide AmyP53
    Jacques Fantini , Nouara Yahi
    2026, 21 (6):  2419-2420.  doi: 10.4103/NRR.NRR-D-25-00076
    Abstract ( 65 )   PDF (818KB) ( 24 )   Save
    Neurodegenerative diseases are a growing burden on healthcare systems. Patients with Alzheimer’s or Parkinson’s diseases (AD or PD) are desperately waiting for innovative solutions that are slow to come, despite several decades of research worldwide. In 2021 and again in 2023, two monoclonal antibodies, aducanumab and lecanemab, have been approved by the U.S. Food and Drug Administration, and a third, donanemab, is currently under review.  However, these treatments have very limited efficacy on cognitive functions and are accompanied by major side effects: amyloidrelated imaging abnormalities, microhemorrhages, and accelerated brain volume loss (Høilund-Carlsen et al., 2024). The paradigm underlying these treatments is the amyloid cascade leading to the accumulation of amyloid plaques in the brain of patients (Fantini et al., 2020). Even if this strategy remains favored by most pharmaceutical companies, it suffers from major contradictions that could eventually lead to its abandonment. If the involvement of the Alzheimer’s amyloid-β (Aβ) protein remains undeniable, the debate has been open for several years to unequivocally identify the neurotoxic form of this protein. In fact, the presence of amyloid plaques is not systematically associated with AD, and we know of at least one deletion in the Aβ protein (Osaka mutation noted E22del or E22Δ) associated with the disease but without amyloid plaque (Fantini et al., 2020).
    Related Articles | Metrics
    Intermittent hypoxic perconditioning improves cognitive function in a mouse model of vascular cognitive impairment and dementia with comorbidities by recovering cerebral blood flow
    Feiyang Jin, Zhengming Tian, Yuying Guan, Yuning Li, Yakun Gu, Mengyuan Guo, Qianqian Shao, Yingxia Liu, Xiuhai Guo, Zhenzhen Quan, Jia Liu, Xunming Ji
    2026, 21 (6):  2421-2431.  doi: 10.4103/NRR.NRR-D-24-00716
    Abstract ( 101 )   PDF (6863KB) ( 34 )   Save
    Vascular cognitive impairment and dementia is a debilitating neurological disorder caused by chronic cerebral hypoperfusion, for which no effective causative treatments are currently available. Intermittent hypoxia has been shown to enhance cerebral blood flow in mice, but its efficacy in a model of vascular cognitive impairment and dementia remains unclear. In this study, we established a mouse model of vascular cognitive impairment and dementia by bilateral carotid artery stenosis. Intermittent hypoxia was induced before and after this stenosis. We found that intermittent hypoxia increased cerebral blood flow, oxygen saturation, and microcirculation in the prefrontal cortex and hippocampus in the model mice, without causing neurovascular damage. Additionally, intermittent hypoxia significantly improved cognitive function in the mouse model of vascular cognitive impairment and dementia, with perconditioning showing greater efficacy than preconditioning. Improvements in cerebral microcirculation and blood flow were positively correlated with cognitive recovery. Even in a mouse model of vascular cognitive impairment and dementia with comorbidities induced by a high-fat, high-fructose diet, intermittent hypoxic perconditioning demonstrated protective effects on cognitive function. Proteomic analysis indicated that mitochondrial protection is a key mechanism, particularly through upregulating NDUFB8 expression and increasing the activity of mitochondrial complex I. These findings suggest that intermittent hypoxia is a potential noninvasive strategy for the prevention and treatment of vascular cognitive impairment and dementia.
    Related Articles | Metrics
    Heterogeneity of the adult mammalian forebrain neurogenic ependyma: A comprehensive cellular map
    Xuejiao Yang, Yuchen Mu, Qianxiang Wu, Liqiang Zhou, Orion R. Fan, Quan Lin, Wenmin Zhu, Yi Eve Sun
    2026, 21 (6):  2432-2441.  doi: 10.4103/NRR.NRR-D-24-00789
    Abstract ( 120 )   Save
    The presence or absence of adult neural stem cells in the mammalian forebrain ependyma has been debated for two decades. In this study, we performed single-cell RNA sequencing to investigate the cellular composition of the ependymal surface of the adult mouse forebrain using whole mounts of lateral walls of lateral ventricles. We identified 12 different cell subtypes in the ependymal surface. Immunocytochemical analyses revealed that CD133+ multi-ciliated cells comprised 67.6% of ependymal cells, while the remaining 32.4% were CD133– . CD133+ ependymal cells can be further classified into FOXJ1+ /SOX2+ /ACTA2+ cells, FLT1+ /CD31+ /CLDN5+ endothelial-like cells, and PDGFRB+ /VTN+ /NG2+ pericyte-like cells, as well as endothelial–pericyte-like cells and Foxj1+ endotheliallike cells. CD133– ependymal cells can be further divided into endothelial-like cells, Foxj1+ ependymal cells, Foxj1+ endothelial-like cells, pericyte-like cells, endothelial-pericyte-like cells, VIM+ cells, and cells negative for all of these markers. This comprehensive profiling confirms the heterogeneity of the ependymal surface in the adult mouse forebrain. Debate regarding whether adult ependymal cells contain neural stem cells has arisen because different researchers have examined different populations of ependymal cells. Our study provides a new perspective for investigation of clinical endogenous neural stem cells, ultimately paving the way for stem cell therapies in neurological diseases.
    Related Articles | Metrics
    Machine learning identifies key cells and therapeutic targets during ferroptosis after spinal cord injury
    Yigang Lv, Zhen Li, Lusen Shi, Huan Jian, Fan Yang, Jichuan Qiu, Chao Li, Peng Xiao, Wendong Ruan, Hao Li, Xueying Li, Shiqing Feng, Hengxing Zhou
    2026, 21 (6):  2442-2453..  doi: 10.4103/NRR.NRR-D-24-00037
    Abstract ( 163 )   PDF (15073KB) ( 6 )   Save
    Ferroptosis, a type of cell death that mainly involves iron metabolism imbalance and lipid peroxidation, is strongly correlated with the phagocytic response caused by bleeding after spinal cord injury. Thus, in this study, bulk RNA sequencing data (GSE47681 and GSE5296) and single-cell RNA sequencing data (GSE162610) were acquired from gene expression databases. We then conducted differential analysis and immune infiltration analysis. Atf3 and Piezo1 were identified as key ferroptosis genes through random forest and least absolute shrinkage and selection operator algorithms. Further analysis of single-cell RNA sequencing data revealed a close relationship between ferroptosis and cell types such as macrophages/microglia and their intrinsic state transition processes. Differences in transcription factor regulation and intercellular communication networks were found in ferroptosis-related cells, confirming the high expression of Atf3 and Piezo1 in these cells. Molecular docking analysis confirmed that the proteins encoded by these genes can bind cycloheximide. In a mouse model of T8 spinal cord injury, low-dose cycloheximide treatment was found to improve neurological function, decrease levels of the pro-inflammatory cytokine inducible nitric oxide synthase, and increase levels of the anti-inflammatory cytokine arginase 1. Correspondingly, the expression of the ferroptosis-related gene Gpx4 increased in macrophages/microglia, while the expression of Acsl4 decreased. Our findings reveal the important role of ferroptosis in the treatment of spinal cord injury, identify the key cell types and genes involved in ferroptosis after spinal cord injury, and validate the efficacy of potential drug therapies, pointing to new directions in the treatment of spinal cord injury.
    Related Articles | Metrics
    Short-lived Niemann-Pick type C mice with accelerated brain aging as a novel model for Alzheimer’s disease research
    Vikas Anil Gujjala, Morteza Abyadeh, Isaiah Klimek, Alexander Tyshkovskiy , Naci Oz, José Pedro Castro, Vadim N. Gladyshev, Jason Newton, Alaattin Kaya
    2026, 21 (6):  2454-2467.  doi: 10.4103/NRR.NRR-D-24-01190
    Abstract ( 125 )   PDF (15882KB) ( 18 )   Save
    Alzheimer’s disease is initially thought to be caused by age-associated accumulation of plaques, in recent years, research has increasingly associated Alzheimer’s disease with lysosomal storage and metabolic disorders, and the explanation of its pathogenesis has shifted from amyloid and tau accumulation to oxidative stress and impaired lipid and glucose metabolism aggravated by hypoxic conditions. However, the underlying mechanisms linking those cellular processes and conditions to disease progression have yet to be defined. Here, we applied a disease similarity approach to identify unknown molecular targets of Alzheimer’s disease by using transcriptomic data from congenital diseases known to increase Alzheimer’s disease risk, namely Down syndrome, NiemannPick type C disease, and mucopolysaccharidoses I. We uncovered common pathways, hub genes, and miRNAs across in vitro and in vivo models of these diseases as potential molecular targets for neuroprotection and amelioration of Alzheimer’s disease pathology, many of which have never been associated with Alzheimer’s disease. We then investigated common molecular alterations in brain samples from a Niemann-Pick type C disease mouse model by juxtaposing them with brain samples of both human and mouse models of Alzheimer’s disease. Detailed phenotypic, molecular, chronological, and biological aging analyses revealed that the Npc1tm(I1061T)Dso mouse model can serve as a potential short-lived in vivo model for brain aging and Alzheimer’s disease research. This research represents the first comprehensive approach to congenital disease association with neurodegeneration and a new perspective on Alzheimer’s disease research while highlighting shortcomings and lack of correlation in diverse in vitro models. Considering the lack of an Alzheimer’s disease mouse model that recapitulates the physiological hallmarks of brain aging, the short-lived Npc1tm(I1061T)Dso mouse model can further accelerate the research in these fields and offer a unique model for understanding the molecular mechanisms of Alzheimer’s disease from a perspective of accelerated brain aging.
    Related Articles | Metrics
    Comprehensive clinical and genetic architecture of familial amyotrophic lateral sclerosis in China: A 15-year cohort study with 302 families
    Wei Zheng, Lu Xu, Jinling Cai, Jinwen Hou, Lu Chen, Nan Zhang, Siyan Zhan, Dongsheng Fan, Ji He
    2026, 21 (6):  2468-2475.  doi: 10.4103/NRR.NRR-D-24-00701
    Abstract ( 113 )   PDF (4780KB) ( 29 )   Save
    The growing recognition of the role of genetics in the development of amyotrophic lateral sclerosis is evident. However, there has yet to be a comprehensive analysis of the clinical characteristics and genetics of familial amyotrophic lateral sclerosis in an Asian population. This study aimed to provide an in-depth analysis of the clinical features and genetic spectrum of familial amyotrophic lateral sclerosis over 15 years in a clinic-based cohort of patients from the Chinese mainland. Enrollment of 302 amyotrophic lateral sclerosis families from 28 provinces was undertaken from January 2008 to September 2023. A groupbased trajectory model for disease progression based on amyotrophic lateral sclerosis Functional Rating Scale-Revised (ALSFRS-R) scores was validated using bootstrap internal validation in patients with familial amyotrophic lateral sclerosis, as well as patients with sporadic amyotrophic lateral sclerosis (matched at a 1:4 ratio, with replacement). DNA samples from 244 index patients were screened for variants in the pathogenic genes SOD1, FUS, TDP43, and C9ORF72, of which 146 were also subjected to genome-wide next-generation sequencing. Gene-level burden analysis was used to evaluate the distribution of rare variants in the cohort. We found that rapid dynamic disease progression was associated with an older age at onset, shorter diagnostic delay, lower body mass index, bulbar onset, and ≥ 1 affected first-degree relative. Certain attributes, such as age at onset and time from onset to diagnosis, had comparable impacts on the clinical progression trajectories of both familial amyotrophic lateral sclerosis and sporadic amyotrophic lateral sclerosis. Harboring pathogenic/likely pathogenic variants in amyotrophic lateral sclerosis-causative genes reduced the age of onset of familial amyotrophic lateral sclerosis. Among the patients with familial amyotrophic lateral sclerosis, 17.8% possessed ≥ 2 pathogenic/likely pathogenic variants. Sequencing kernel association test analysis showed that the SOD1 rare variant burden (P = 1.3e−15) was associated with a significant risk of familial amyotrophic lateral sclerosis. Our findings conclusively confirmed the clinical features and genetic spectrum of familial amyotrophic lateral sclerosis over 15 years in a clinical cohort from China, contributing to a deeper understanding of genotype–phenotype relationships in familial amyotrophic lateral sclerosis. This comprehensive evaluation of specific clinical characteristics, clinical prognosis, and genetic variants of amyotrophic lateral sclerosis based on detailed clinical and genetic information may lead to the development of genotype-specific treatment approaches.
    Related Articles | Metrics
    MicroRNA-301a knockout attenuates peripheral nerve regeneration by delaying Wallerian degeneration
    Lanya Fu, Xiaofang Hu, Jiawei Xu, Zhenlin Li, Jiale Cai, Xinrui Ma, Ying Zou, Ye He, Shuyi Xu, Yizhou Xu, Jiaqi Zhang, Yunlun Li, Jingmin Liu, Tsz Hei Fong, Xianghai Wang, Lixin Zhu, Dongfeng Chen, Aijun Liu, Xiaodong Ma, Jiasong Guo
    2026, 21 (6):  2476-2486.  doi: 10.4103/NRR.NRR-D-24-00081
    Abstract ( 160 )   PDF (10617KB) ( 6 )   Save
    Our recent study demonstrated that knockout of microRNA-301a attenuates migration and phagocytosis in macrophages. Considering that macrophages and Schwann cells synergistically clear the debris of degraded axons and myelin during Wallerian degeneration, which is a prerequisite for nerve regeneration, we hypothesized that microRNA-301a regulates Wallerian degeneration and nerve regeneration via impacts on Schwann cell migration and phagocytosis. Herein, we found low expression of microRNA-301a in intact sciatic nerves, with no impact of the microRNA-301a knockout on nerve structure and function. By contrast, we found significant upregulation of microRNA-301a in injured sciatic nerves. We established a sciatic nerve crush model in microRNA-301a knockout mice, which exhibited attenuated morphological and functional regeneration following sciatic nerve crush injury. The microRNA-301a knockout also led to significantly inhibited Wallerian degeneration in an in vivo sciatic nerve-transection model and in an in vitro nerve explant block model. Schwann cells with the microRNA-301a knockout showed inhibition of phagocytosis and migration, which was reversible under transfection with microRNA-301a mimics. Rescue experiments involving transfection of microRNA-301a-knockout Schwann cells with microRNA-301a mimics or treatment with the C–X–C motif receptor 4 inhibitor WZ811 indicated the mechanistic involvement of the Yin Yang 1/C–X–C motif receptor 4 pathway in the role of microRNA-301a. Combined with our previous findings in macrophages, we conclude that microRNA-301a plays a key role in peripheral nerve injury and repair by regulating the migratory and phagocytic capabilities of Schwann cells and macrophages via the Yin Yang 1/C–X–C motif receptor 4 pathway.
    Related Articles | Metrics
    Transforming growth factor beta–related proteins promote axonal regeneration of injured dorsal root ganglion neurons
    Yinying Shen, Peng Yang, Wenyu Dai, Xiaosong Gu , Sheng Yi
    2026, 21 (6):  2487-2496.  doi: 10.4103/NRR.NRR-D-24-01427
    Abstract ( 101 )   PDF (4057KB) ( 20 )   Save
    Dorsal root ganglia neurons gradually lose their axonal regeneration ability during development and aging. To explore molecules that enhance axonal regeneration, we screened growth factors with differential gene expression patterns in the dorsal root ganglias of young adult and aged animals following sciatic nerve injury. In young adult animals, two transforming growth factor beta-related factors, activin A and angiopoietin 2, were found to be upregulated post nerve injury. Treatment of isolated dorsal root ganglia explants and cultured dorsal root ganglia neurons of neonatal and young adult rats with recombinant activin A or angiopoietin 2 protein stimulated neurite outgrowth and axonal elongation. The administration of recombinant activin A or angiopoietin 2 protein to sciatic nerve crush-injured dorsal root ganglias also supported the growth of sensory neurons and facilitated nerve regeneration in both young adult and aged rats. Using RNA sequencing, we characterized genetic changes in dorsal root ganglia neurons following recombinant activin A or angiopoietin 2 treatment, revealing the unique mechanisms of these transforming growth factor beta–related factors. Recombinant activin A elicited changes in the gene expression of cytoskeleton-related Gper1 and activated extracellular signal-regulated kinase signaling, while angiopoietin 2 increased the expression of the transcription factor gene E2f2. Our identification of activin A and angiopoietin 2 as crucial promotional factors of axonal regeneration may guide future therapeutic strategies for the treatment of nerve injury.
    Related Articles | Metrics