Loading...

Table of Content

    15 April 2026, Volume 21 Issue 4 Previous Issue   
    For Selected: Toggle Thumbnails
    Pericyte-glial cell interactions: Insights into brain health and disease
    Ali Sepehrinezhad, Ali Gorji
    2026, 21 (4):  1253-1263.  doi: 10.4103/NRR.NRR-D-24-01472
    Abstract ( 26 )   PDF (5167KB) ( 23 )   Save
    Pericytes are multi-functional mural cells of the central nervous system that cover the capillary endothelial cells. Pericytes play a vital role in nervous system development, significantly influencing the formation, maturation, and maintenance of the central nervous system. An expanding body of studies has revealed that pericytes establish carefully regulated interactions with oligodendrocytes, microglia, and astrocytes. These communications govern numerous critical brain processes, including angiogenesis, neurovascular unit homeostasis, blood–brain barrier integrity, cerebral blood flow regulation, and immune response initiation. Glial cells and pericytes participate in dynamic and reciprocal interactions, with each influencing and adjusting the functionality of the other. Pericytes have the ability to control astrocyte polarization, trigger differentiation of oligodendrocyte precursor cells, and initiate immunological responses in microglia. Various neurological disorders that compromise the integrity of the blood–brain barrier can disrupt these communications, impair waste clearance, and hinder cerebral blood circulation, contributing to neuroinflammation. In the context of neurodegeneration, these disruptions exacerbate pathological processes, such as neuronal damage, synaptic dysfunction, and impaired tissue repair. This article explores the complex interactions between pericytes and various glial cells in both healthy and pathological states of the central nervous system. It highlights their essential roles in neurovascular function and disease progression, providing important insights that may enhance our understanding of the molecular mechanisms underlying these interactions and guide potential therapeutic strategies for neurodegenerative disorders in future research.
    Related Articles | Metrics
    Effects of noninvasive brain stimulation on motor functions in animal models of ischemia and trauma in the central nervous system
    Seda Demir, Gereon R. Fink, Maria A. Rueger, Stefan J. Blaschke
    2026, 21 (4):  1264-1276.  doi: 10.4103/NRR.NRR-D-24-01613
    Abstract ( 24 )   PDF (885KB) ( 15 )   Save
    Noninvasive brain stimulation techniques offer promising therapeutic and regenerative prospects in neurological diseases by modulating brain activity and improving cognitive and motor functions. Given the paucity of knowledge about the underlying modes of action and optimal treatment modalities, a thorough translational investigation of noninvasive brain stimulation in preclinical animal models is urgently needed. Thus, we reviewed the current literature on the mechanistic underpinnings of noninvasive brain stimulation in models of central nervous system impairment, with a particular emphasis on traumatic brain injury and stroke. Due to the lack of translational models in most noninvasive brain stimulation techniques proposed, we found this review to the most relevant techniques used in humans, i.e., transcranial magnetic stimulation and transcranial direct current stimulation. We searched the literature in PubMed, encompassing the MEDLINE and PMC databases, for studies published between January 1, 2020 and September 30, 2024. Thirty-five studies were eligible. Transcranial magnetic stimulation and transcranial direct current stimulation demonstrated distinct strengths in augmenting rehabilitation post-stroke and traumatic brain injury, with emerging mechanistic evidence. Overall, we identified neuronal, inflammatory, microvascular, and apoptotic pathways highlighted in the literature. This review also highlights a lack of translational surrogate parameters to bridge the gap between preclinical findings and their clinical translation.
    Related Articles | Metrics
    Regulatory T cells in neurological disorders and tissue regeneration: Mechanisms of action and therapeutic potentials
    Jing Jie, Xiaomin Yao, Hui Deng, Yuxiang Zhou, Xingyu Jiang, Xiu Dai, Yumin Yang, Pengxiang Yang
    2026, 21 (4):  1277-1291.  doi: 10.4103/NRR.NRR-D-24-01363
    Abstract ( 19 )   PDF (1745KB) ( 10 )   Save
    Regulatory T cells, a subset of CD4+ T cells, play a critical role in maintaining immune tolerance and tissue homeostasis due to their potent immunosuppressive properties. Recent advances in research have highlighted the important therapeutic potential of Tregs in neurological diseases and tissue repair, emphasizing their multifaceted roles in immune regulation. This review aims to summarize and analyze the mechanisms of action and therapeutic potential of Tregs in relation to neurological diseases and neural regeneration. Beyond their classical immune-regulatory functions, emerging evidence points to non-immune mechanisms of regulatory T cells, particularly their interactions with stem cells and other non-immune cells. These interactions contribute to optimizing the repair microenvironment and promoting tissue repair and nerve regeneration, positioning non-immune pathways as a promising direction for future research. By modulating immune and non-immune cells, including neurons and glia within neural tissues, Tregs have demonstrated remarkable efficacy in enhancing regeneration in the central and peripheral nervous systems. Preclinical studies have revealed that Treg cells interact with neurons, glial cells, and other neural components to mitigate inflammatory damage and support functional recovery. Current mechanistic studies show that Tregs can significantly promote neural repair and functional recovery by regulating inflammatory responses and the local immune microenvironment. However, research on the mechanistic roles of regulatory T cells in other diseases remains limited, highlighting substantial gaps and opportunities for exploration in this field. Laboratory and clinical studies have further advanced the application of regulatory T cells. Technical advances have enabled efficient isolation, ex vivo expansion and functionalization, and adoptive transfer of regulatory T cells, with efficacy validated in animal models. Innovative strategies, including gene editing, cell-free technologies, biomaterial-based recruitment, and in situ delivery have expanded the therapeutic potential of regulatory T cells. Gene editing enables precise functional optimization, while biomaterial and in situ delivery technologies enhance their accumulation and efficacy at target sites. These advancements not only improve the immune-regulatory capacity of regulatory T cells but also significantly enhance their role in tissue repair. By leveraging the pivotal and diverse functions of Tregs in immune modulation and tissue repair, regulatory T cells–based therapies may lead to transformative breakthroughs in the treatment of neurological diseases.
    Related Articles | Metrics
    Neuroglobin: A promising candidate to treat neurological diseases
    Ivan Millan Yañez, Isabel Torres-Cuevas, Marisol Corral-Debrinski
    2026, 21 (4):  1292-1303.  doi: 10.4103/NRR.NRR-D-24-01503
    Abstract ( 16 )   PDF (1291KB) ( 10 )   Save
    Neurodevelopmental and neurodegenerative illnesses constitute a global health issue and a foremost economic burden since they are a large cause of incapacity and death worldwide. Altogether, the burden of neurological disorders has increased considerably over the past 30 years because of population aging. Overall, neurological diseases significantly impair cognitive and motor functions and their incidence will increase as societies age and the world’s population continues to grow. Autism spectrum disorder, motor neuron disease, encephalopathy, epilepsy, stroke, ataxia, Alzheimer’s disease, amyotrophic lateral sclerosis, Huntington’s disease, and Parkinson’s disease represent a non-exhaustive list of neurological illnesses. These affections are due to perturbations in cellular homeostasis leading to the progressive injury and death of neurons in the nervous system. Among the common features of neurological handicaps, we find protein aggregation, oxidative stress, neuroinflammation, and mitochondrial impairment in the target tissues, e.g., the brain, cerebellum, and spinal cord. The high energy requirements of neurons and their inability to produce sufficient adenosine triphosphate by glycolysis, are responsible for their dependence on functional mitochondria for their integrity. Reactive oxygen species, produced along with the respiration process within mitochondria, can lead to oxidative stress, which compromises neuronal survival. Besides having an essential role in energy production and oxidative stress, mitochondria are indispensable for an array of cellular processes, such as amino acid metabolism, iron-sulfur cluster biosynthesis, calcium homeostasis, intrinsic programmed cell death (apoptosis), and intraorganellar signaling. Despite the progress made in the last decades in the understanding of a growing number of genetic and molecular causes of central nervous diseases, therapies that are effective to diminish or halt neuronal dysfunction/death are rare. Given the genetic complexity responsible for neurological disorders, the development of neuroprotective strategies seeking to preserve mitochondrial homeostasis is a realistic challenge to lastingly diminish the harmful evolution of these pathologies and so to recover quality of life. A promising candidate is the neuroglobin, a globin superfamily member of 151 amino acids, which is found at high levels in the brain, the eye, and the cerebellum. The protein, which localizes to mitochondria, is involved in electron transfer, oxygen storage and defence against oxidative stress; hence, possessing neuroprotective properties. This review surveys up-to-date knowledge and emphasizes on existing investigations regarding neuroglobin physiological functions, which remain since its discovery in 2000 under intense debate and the possibility of using neuroglobin either by gene therapy or its direct delivery into the brain to treat neurological disorders.
    Related Articles | Metrics
    Trends in the application of chondroitinase ABC in injured spinal cord repair
    Zhongqing Ji, Jiangfeng Zhu, Jinming Liu, Bin Wei, Yixin Shen, Yanan Hu
    2026, 21 (4):  1304-1321.  doi: 10.4103/NRR.NRR-D-24-01354
    Abstract ( 18 )   PDF (2932KB) ( 4 )   Save
    Spinal cord injuries have overwhelming physical and occupational implications for patients. Moreover, the extensive and long-term medical care required for spinal cord injury significantly increases healthcare costs and resources, adding a substantial burden to the healthcare system and patients’ families. In this context, chondroitinase ABC, a bacterial enzyme isolated from Proteus vulgaris that is modified to facilitate expression and secretion in mammals, has emerged as a promising therapeutic agent. It works by degrading chondroitin sulfate proteoglycans, cleaving the glycosaminoglycanchains of chondroitin sulfate proteoglycans into soluble disaccharides or tetrasaccharides. Chondroitin sulfate proteoglycans are potent axon growth inhibitors and principal constituents of the extracellular matrix surrounding glial and neuronal cells attached to glycosaminoglycan chains. Chondroitinase ABC has been shown to play an effective role in promoting recovery from acute and chronic spinal cord injury by improving axonal regeneration and sprouting, enhancing the plasticity of perineuronal nets, inhibiting neuronal apoptosis, and modulating immune responses in various animal models. In this review, we introduce the classification and pathological mechanisms of spinal cord injury and discuss the pathophysiological role of chondroitin sulfate proteoglycans in spinal cord injury. We also highlight research advancements in spinal cord injury treatment strategies, with a focus on chondroitinase ABC, and illustrate how improvements in chondroitinase ABC stability, enzymatic activity, and delivery methods have enhanced injured spinal cord repair. Furthermore, we emphasize that combination treatment with chondroitinase ABC further enhances therapeutic efficacy. This review aimed to provide a comprehensive understanding of the current trends and future directions of chondroitinase ABC -based spinal cord injury therapies, with an emphasis on how modern technologies are accelerating the optimization of chondroitinase ABC development.
    Related Articles | Metrics
    Mitochondrial damage-associated molecular patterns: Neuroimmunomodulators in central nervous system pathophysiology
    Noah A. H. Brooks, Ishvin Riar, Andis Klegeris
    2026, 21 (4):  1322-1338.  doi: 10.4103/NRR.NRR-D-24-01459
    Abstract ( 14 )   PDF (2046KB) ( 4 )   Save
    Neuroinflammation contributes to a wide range of neurodegenerative diseases including Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, and multiple sclerosis. It is driven by non-neuronal glial cells, mainly microglia and astrocytes. Microglia are the resident immune cells of the central nervous system, while astrocytes are the main support cells for neuronal functions but can also participate in neuroimmune responses. Both these glial cell types can become reactive upon detection of certain endogenous intracellular molecules that appear in the extracellular space under specific circumstances; these can be pathology-associated abnormal structures, such as amyloid β proteins, or damage-associated molecular patterns released from injured cells, including their mitochondria. Once in the extracellular space, damage-associated molecular patterns act as ligands for specific pattern recognition receptors expressed by glia inducing their reactivity and neuroimmune responses. This review considers the following mitochondrial damage-associated molecular patterns: heme, cytochrome c, cardiolipin, adenosine triphosphate, mitochondrial DNA, mitochondrial transcription factor A, N-formyl peptides, and the tricarboxylic acid cycle metabolites: succinate, fumarate, and itaconate. We describe their well-established functions as damage-associated molecular patterns of the peripheral tissues before summarizing available evidence indicating these molecules may also play significant roles in the neuroimmune processes of the central nervous system. We highlight the pattern recognition receptors that mitochondrial damage-associated molecular patterns interact with and the cellular signaling mechanisms they modulate. Our review demonstrates that some mitochondrial damage-associated molecular patterns, such as cytochrome c, adenosine triphosphate, and mitochondrial transcription factor A, have already demonstrated significant effects on the central nervous system. In contrast, others including cardiolipin, mitochondrial DNA, N-formyl peptides, succinate, fumarate, and itaconate, will require additional studies corroborating their roles as damageassociated molecular patterns in the central nervous system. For all of the reviewed mitochondrial damage-associated molecular patterns, there is a shortage of studies using human cells and tissues, which is identified as a significant knowledge gap. We also assess the need for targeted research on the effects of mitochondrial damage-associated molecular patterns in the central nervous system pathologies where their roles are understudied. Such studies could identify novel treatment strategies for multiple neurodegenerative diseases, which are characterized by chronic neuroinflammation and currently lack effective therapies.
    Related Articles | Metrics
    Unfolded protein response in endoplasmic reticulum stress associated with retinal degenerative diseases: A promising therapeutic target
    Hongbing Zhang, Yalin Mu, Hongsong Li, Xiaogang Li
    2026, 21 (4):  1339-1352.  doi: 10.4103/NRR.NRR-D-24-01124
    Abstract ( 20 )   PDF (3623KB) ( 7 )   Save
    The unfolded protein response is a cellular pathway activated to maintain proteostasis and prevent cell death when the endoplasmic reticulum is overwhelmed by unfolded proteins. However, if the unfolded protein response fails to restore endoplasmic reticulum homeostasis, it can trigger proinflammatory and pro-death signals, which are implicated in various malignancies and are currently being investigated for their role in retinal degenerative diseases. This paper reviews the role of the unfolded protein responsein addressing endoplasmic reticulumstress in retinal degenerative diseases. The accumulation of ubiquitylated misfolded proteins can lead to rapid destabilization of the proteome and cellular demise. Targeting endoplasmic reticulum stress to alleviate retinal pathologies involves multiple strategies, including the use of chemical chaperones such as 4-phenylbutyric acid and tauroursodeoxycholic acid, which enhance protein folding and reduce endoplasmic reticulum stress. Small molecule modulators that influence endoplasmic reticulum stress sensors, including those that increase the expression of the endoplasmic reticulum stress regulator X-box binding protein 1, are also potential therapeutic agents. Additionally, inhibitors of the RNAse activity of inositol-requiring transmembrane kinase/endoribonuclease 1, a key endoplasmic reticulum stress sensor, represent another class of drugs that could prevent the formation of toxic aggregates. The activation of nuclear receptors, such as PPAR and FXR, may also help mitigate ER stress. Furthermore, enhancing proteolysis through the induction of autophagy or the inhibition of deubiquitinating enzymes can assist in clearing misfolded proteins. Combination treatments that involve endoplasmicreticulum-stress-targeting drugs and gene therapies are also being explored. Despite these potential therapeutic strategies, significant challenges remain in targeting endoplasmic reticulum stress for the treatment of retinal degeneration, and further research is essential to elucidate the mechanisms underlying human retinal diseases and to develop effective, well-tolerated drugs. The use of existing drugs that target inositol-requiring transmembrane kinase/endoribonuclease 1 and X-box binding protein 1 has been associated with adverse side effects, which have hindered their clinical translation. Moreover, signaling pathways downstream of endoplasmic reticulum stress sensors can contribute to therapy resistance. Addressing these limitations is crucial for developing drugs that can be effectively used in treating retinal dystrophies. In conclusion, while the unfolded protein response is a promising therapeutic target in retinal degenerative diseases, additional research and development efforts are imperative to overcome the current limitations and improve patient outcomes.
    Related Articles | Metrics
    Techniques and factors for reducing chronic neuropathic pain: A review
    Damien P. Kuffler
    2026, 21 (4):  1353-1358.  doi: 10.4103/NRR.NRR-D-22-00015
    Abstract ( 13 )   PDF (482KB) ( 5 )   Save
    Nerve trauma commonly results in chronic neuropathic pain. This is by triggering the release of proinflammatory mediators from local and invading cells that induce inflammation and nociceptive neuron hyperexcitability. Even without apparent inflammation, injury sites are associated with increased inflammatory markers. This review focuses on how it might be possible to reduce neuropathic pain by reducing inflammation. Physiologically, pain is resolved by a combination of the out-migration of pro-inflammatory cells from the injury site, the down-regulation of the genes underlying the inflammation, up-regulating genes for anti-inflammatory mediators, and reducing nociceptive neuron hyperexcitability. While various techniques reduce chronic neuropathic pain, the best are effective on < 50% of patients, no technique reliably or permanently eliminates neuropathic pain. This is because most techniques are predominantly aimed at reducing pain, not inflammation. In addition, while single factors reduce pain, increasing evidence indicates significant and longer-lasting pain relief requires multiple factors acting simultaneously. Therefore, it is not surprising that extensive data indicate that the application of platelet-rich plasma provides more significant and longer-lasting pain suppression than other techniques, although its analgesia is neither complete nor permanent. However, several case reports indicate that platelet-rich plasma can induce permanent neuropathic pain elimination when the platelet concentration is significantly increased and is applied to longer nerve lengths. This review examines the primary triggers of the development and maintenance of neuropathic pain and techniques that reduce chronic neuropathic pain. The application of plateletrich plasma holds great promise for providing complete and permanent chronic neuropathic pain elimination.
    Related Articles | Metrics
    Different roles of astrocytes in the blood–brain barrier during the acute and recovery phases of stroke
    Jialin Cheng , Yuxiao Zheng , Fafeng Cheng, Chunyu Wang, Jinhua Han, Haojia Zhang, Xin Lan, Chuxin Zhang, Xueqian Wang , Qingguo Wang , Changxiang Li
    2026, 21 (4):  1359-1372.  doi: 10.4103/NRR.NRR-D-24-01417
    Abstract ( 17 )   PDF (17873KB) ( 3 )   Save
    Ischemic stroke, a frequently occurring form of stroke, is caused by obstruction of cerebral blood flow, which leads to ischemia, hypoxia, and necrosis of local brain tissue. After ischemic stroke, both astrocytes and the blood–brain barrier undergo morphological and functional transformations. However, the interplay between astrocytes and the blood–brain barrier has received less attention. This comprehensive review explores the physiological and pathological morphological and functional changes in astrocytes and the blood–brain barrier in ischemic stroke. Post-stroke, the structure of endothelial cells and peripheral cells undergoes alterations, causing disruption of the blood–brain barrier. This disruption allows various pro-inflammatory factors and chemokines to cross the blood– brain barrier. Simultaneously, astrocytes swell and primarily adopt two phenotypic states: A1 and A2, which exhibit different roles at different stages of ischemic stroke. During the acute phase, A1 reactive astrocytes secrete vascular endothelial growth factor, matrix metalloproteinases, lipid carrier protein-2, and other cytokines, exacerbating damage to endothelial cells and tight junctions. Conversely, A2 reactive astrocytes produce pentraxin 3, Sonic hedgehog, angiopoietin-1, and other protective factors for endothelial cells. Furthermore, astrocytes indirectly influence blood–brain barrier permeability through ferroptosis and exosomes. In the middle and late (recovery) stages of ischemic stroke, A1 and A2 astrocytes show different effects on glial scar formation. A1 astrocytes promote glial scar formation and inhibit axon growth via glial fibrillary acidic protein, chondroitin sulfate proteoglycans, and transforming growth factor-β. In contrast, A2 astrocytes facilitate axon growth through platelet-derived growth factor, playing a crucial role in vascular remodeling. Therefore, enhancing our understanding of the pathological changes and interactions between astrocytes and the blood–brain barrier is a vital therapeutic target for preventing further brain damage in acute stroke. These insights may pave the way for innovative therapeutic strategies for ischemic stroke.
    Related Articles | Metrics
    Neuronal ion channel modulation by Drimys winteri compounds: Opening a new chemical space to neuropharmacology
    Macarena E. Meza, Oscar Ramirez-Molina, Oscar Flores, Katherine Fariña-Oliva, Pamela A. Godoy, Jorge Fuentealba, Gonzalo E. Yévenes
    2026, 21 (4):  1373-1382.  doi: 10.4103/NRR.NRR-D-24-01194
    Abstract ( 13 )   PDF (1861KB) ( 5 )   Save
    Numerous pathological states of the nervous system involve alterations in neuronal excitability and synaptic dysfunction, which depend on the function of ion channels. Due to their critical involvement in health and disease, the search for new compounds that modulate these proteins is still relevant. Traditional medicine has long been a rich source of neuroactive compounds. For example, the indigenous Mapuche people have used the leaves and bark of the Drimys winteri tree for centuries to treat various diseases. Consequently, several studies have investigated the biological effects of compounds in Drimys winteri, highlighting sesquiterpenes such as α-humulene, drimenin, polygodial, and α-, β-, γ-eudesmol. However, there is currently no literature review focusing on the ability of these sesquiterpenes to modulate ion channels. This review summarizes the current knowledge about neuroactive compounds found in Drimys winteri, with special emphasis on their direct actions on neuronal ion channels. Several Drimys winteri sesquiterpenes modulate a diverse array of neuronal ion channels, including transient receptor potential channels, gamma-aminobutyric acid A receptors, nicotinic acetylcholine receptors, and voltage-dependent Ca2+ and Na+ channels. Interestingly, the modulation of these molecular targets by Drimys winteri sesquiterpenes correlates with their therapeutic actions. The promiscuous pharmacological profile of Drimys winteri sesquiterpenes suggests they modulate multiple protein targets in vivo, making them potentially useful for treating complex, multifactorial diseases. Further studies at the molecular level may aid in developing multitargeted drugs with enhanced therapeutic effects.
    Related Articles | Metrics
    Damage and repair in retinal degenerative diseases: Molecular basis through clinical translation
    Ziting Zhang, Junfeng Ma, Wahid Shah, Xin Quan, Tao Ding, Yuan Gao
    2026, 21 (4):  1383-1395.  doi: 10.4103/NRR.NRR-D-24-01016
    Abstract ( 30 )   PDF (2614KB) ( 7 )   Save
    Retinal ganglion cells are the bridging neurons between the eye and the central nervous system, transmitting visual signals to the brain. The injury and loss of retinal ganglion cells are the primary pathological changes in several retinal degenerative diseases, including glaucoma, ischemic optic neuropathy, diabetic neuropathy, and optic neuritis. In mammals, injured retinal ganglion cells lack regenerative capacity and undergo apoptotic cell death within a few days of injury. Additionally, these cells exhibit limited regenerative ability, ultimately contributing to vision impairment and potentially leading to blindness. Currently, the only effective clinical treatment for glaucoma is to prevent vision loss by lowering intraocular pressure through medications or surgery; however, this approach cannot halt the effect of retinal ganglion cell loss on visual function. This review comprehensively investigates the mechanisms underlying retinal ganglion cell degeneration in retinal degenerative diseases and further explores the current status and potential of cell replacement therapy for regenerating retinal ganglion cells. As our understanding of the complex processes involved in retinal ganglion cell degeneration deepens, we can explore new treatment strategies, such as cell transplantation, which may offer more effective ways to mitigate the effect of retinal degenerative diseases on vision.
    Related Articles | Metrics
    Mitophagy: A key regulator in the pathophysiology and treatment of spinal cord injury
    Qiuyang Gu, Shengye Yuan, Yumei An, Wenyue Sun, Mingyuan Xu, Mengchun Xue, Xianzhe Li, Chao Liu, Haiyan Shan, Mingyang Zhang
    2026, 21 (4):  1396-1408.  doi: 10.4103/NRR.NRR-D-24-01029
    Abstract ( 20 )   PDF (20589KB) ( 5 )   Save
    Mitophagy is closely associated with the pathogenesis of secondary spinal cord injury. Abnormal mitophagy may contribute significantly to secondary spinal cord injury, leading to the impaired production of adenosine triphosphate, ion imbalance, the excessive production of reactive oxygen species, neuroinflammation, and neuronal cell death. Therefore, maintaining an appropriate balance of mitophagy is crucial when treating spinal cord injury, as both excessive and insufficient mitophagy can impede recovery. In this review, we summarize the pathological changes associated with spinal cord injury, the mechanisms of mitophagy, and the direct and indirect relationships between mitophagy and spinal cord injury. We also consider therapeutic approaches that target mitophagy for the treatment of spinal cord injury, including ongoing clinical trials and other innovative therapies, such as use of stem cells, nanomaterials, and small molecule polymers. Finally, we highlight the current challenges facing this field and suggest potential directions for future research. The aim of our review is to provide a theoretical reference for future studies targeting mitophagy in the treatment of spinal cord injury.
    Related Articles | Metrics
    Inherent potential of mitochondria-targeted interventions for chronic neurodegenerative diseases
    Min Zhou, Min Zheng, Siyao Liang, Maomao Li, Jiarui Ma, Shiyu Zhang, Xinyao Song, Yonglin Hu, Yuhong Lyu, Xingkun Ou, Changwu Yue
    2026, 21 (4):  1409-1427.  doi: 10.4103/NRR.NRR-D-24-01507
    Abstract ( 13 )   PDF (12991KB) ( 2 )   Save
    The cure rate for chronic neurodegenerative diseases remains low, creating an urgent need for improved intervention methods. Recent studies have shown that enhancing mitochondrial function can mitigate the effects of these diseases. This paper comprehensively reviews the relationship between mitochondrial dysfunction and chronic neurodegenerative diseases, aiming to uncover the potential use of targeted mitochondrial interventions as viable therapeutic options. We detail five targeted mitochondrial intervention strategies for chronic neurodegenerative diseases that act by promoting mitophagy, inhibiting mitochondrial fission, enhancing mitochondrial biogenesis, applying mitochondria-targeting antioxidants, and transplanting mitochondria. Each method has unique advantages and potential limitations, making them suitable for various therapeutic situations. Therapies that promote mitophagy or inhibit mitochondrial fission could be particularly effective in slowing disease progression, especially in the early stages. In contrast, those that enhance mitochondrial biogenesis and apply mitochondria-targeting antioxidants may offer great benefits during the middle stages of the disease by improving cellular antioxidant capacity and energy metabolism. Mitochondrial transplantation, while still experimental, holds great promise for restoring the function of damaged cells. Future research should focus on exploring the mechanisms and effects of these intervention strategies, particularly regarding their safety and efficacy in clinical settings. Additionally, the development of innovative mitochondria-targeting approaches, such as gene editing and nanotechnology, may provide new solutions for treating chronic neurodegenerative diseases. Implementing combined therapeutic strategies that integrate multiple intervention methods could also enhance treatment outcomes.
    Related Articles | Metrics
    Stem cell repair strategies for epilepsy
    Xiao Ma , Zitong Wang , Yinuo Niu, Jie Zhao, Xiaorui Wang, Xuan Wang, Fang Yang, Dong Wei, Zhongqing Sun , Wen Jiang
    2026, 21 (4):  1428-1446.  doi: 10.4103/NRR.NRR-D-24-01337
    Abstract ( 17 )   PDF (2806KB) ( 15 )   Save
    Epilepsy is a serious neurological disorder; however, the effectiveness of current medications is often suboptimal. Recently, stem cell technology has demonstrated remarkable therapeutic potential in addressing various neurological diseases, igniting interest in its applicability for epilepsy treatment. This comprehensive review summarizes different therapeutic approaches utilizing various types of stem cells. Preclinical experiments have explored the use and potential therapeutic effects of mesenchymal stem cells, including genetically modified variants. Clinical trials involving patientderived mesenchymal stem cells have shown promising results, with reductions in the frequency of epileptic seizures and improvements in neurological, cognitive, and motor functions reported. Another promising therapeutic strategy involves neural stem cells. These cells can be cultured outside the body and directed to differentiate into specific cell types. The transplant of neural stem cells has the potential to replace lost inhibitory interneurons, providing a novel treatment avenue for epilepsy. Embryonic stem cells are characterized by their significant capacity for self-renewal and their ability to differentiate into any type of somatic cell. In epilepsy treatment, embryonic stem cells can serve three primary functions: neuron regeneration, the maintenance of cellular homeostasis, and restorative activity. One notable strategy involves differentiating embryonic stem cells into γ-aminobutyric acidergic neurons for transplantation into lesion sites. This approach is currently undergoing clinical trials and could be a breakthrough in the treatment of refractory epilepsy. Induced pluripotent stem cells share the same genetic background as the donor, thereby reducing the risk of immune rejection and addressing ethical concerns. However, research on induced pluripotent stem cell therapy remains in the preclinical stage. Despite the promise of stem cell therapies for epilepsy, several limitations must be addressed. Safety concerns persist, including issues such as tumor formation, and the low survival rate of transplanted cells remains a significant challenge. Additionally, the high cost of these treatments may be prohibitive for some patients. In summary, stem cell therapy shows considerable promise in managing epilepsy, but further research is needed to overcome its existing limitations and enhance its clinical applicability.
    Related Articles | Metrics
    Brain-derived extracellular vesicles: A promising avenue for Parkinson’s disease pathogenesis, diagnosis, and treatment
    Shurui Zhang, Jingwen Li, Xinyu Hu, Hanshu Liu , Qinwei Yu , Guiying Kuang , Long Liu , Danfang Yu , Zhicheng Lin, Nian Xiong
    2026, 21 (4):  1447-1467.  doi: 10.4103/NRR.NRR-D-24-01262
    Abstract ( 14 )   PDF (4123KB) ( 6 )   Save
    The misfolding, aggregation, and deposition of alpha-synuclein into Lewy bodies are pivotal events that trigger pathological changes in Parkinson’s disease. Extracellular vesicles are nanosized lipidbilayer vesicles secreted by cells that play a crucial role in intercellular communication due to their diverse cargo. Among these, brain-derived extracellular vesicles, which are secreted by various brain cells such as neurons, glial cells, and Schwann cells, have garnered increasing attention. They serve as a promising tool for elucidating Parkinson’s disease pathogenesis and for advancing diagnostic and therapeutic strategies. This review highlights the recent advancements in our understanding of brain-derived extracellular vesicles released into the blood and their role in the pathogenesis of Parkinson’s disease, with specific emphasis on their involvement in the aggregation and spread of alpha-synuclein. Brain-derived extracellular vesicles contribute to disease progression through multiple mechanisms, including autophagy-lysosome dysfunction, neuroinflammation, and oxidative stress, collectively driving neurodegeneration in Parkinson’s disease. Their application in Parkinson’s disease diagnosis is a primary focus of this review. Recent studies have demonstrated that brainderived extracellular vesicles can be isolated from peripheral blood samples, as they carry α-synuclein and other key biomarkers such as DJ-1 and various microRNAs. These findings highlight the potential of brain-derived extracellular vesicles, not only for the early diagnosis of Parkinson’s disease but also for disease progression monitoring and differential diagnosis. Additionally, an overview of explorations into the potential therapeutic applications of brain-derived extracellular vesicles for Parkinson’s disease is provided. Therapeutic strategies targeting brain-derived extracellular vesicles involve modulating the release and uptake of pathological alpha-synuclein -containing brain-derived extracellular vesicles to inhibit the spread of the protein. Moreover, brain-derived extracellular vesicles show immense promise as therapeutic delivery vehicles capable of transporting drugs into the central nervous system. Importantly, brain-derived extracellular vesicles also play a crucial role in neural regeneration by promoting neuronal protection, supporting axonal regeneration, and facilitating myelin repair, further enhancing their therapeutic potential in Parkinson’s disease and other neurological disorders. Further clarification is needed of the methods for identifying and extracting brain-derived extracellular vesicles, and large-scale cohort studies are necessary to validate the accuracy and specificity of these biomarkers. Future research should focus on systematically elucidating the unique mechanistic roles of brain-derived extracellular vesicles, as well as their distinct advantages in the clinical translation of methods for early detection and therapeutic development.
    Related Articles | Metrics
    Role of peroxisome proliferator-activated receptor alpha in neurodegenerative diseases and other neurological disorders: Clinical application prospects
    Zijun Wu, Yuying Zhao, Shujing Hao, Mengyao An, Chengcheng Song, Jing Li
    2026, 21 (4):  1468-1482.  doi: 10.4103/NRR.NRR-D-24-01371
    Abstract ( 15 )   PDF (4551KB) ( 4 )   Save
    Peroxisome proliferator-activated receptor alpha is a member of the nuclear hormone receptor superfamily and functions as a transcription factor involved in regulating cellular metabolism. Previous studies have shown that PPARα plays a key role in the onset and progression of neurodegenerative diseases. Consequently, peroxisome proliferator-activated receptor alpha agonists have garnered increasing attention as potential treatments for neurological disorders. This review aims to clarify the research progress regarding peroxisome proliferator-activated receptor alpha in nervous system diseases. Peroxisome proliferator-activated receptor alpha is present in all cell types within adult mouse and adult neural tissues. Although it is conventionally believed to be primarily localized in the nucleus, its function may be regulated by a dynamic balance between cytoplasmic and nuclear shuttling. Both endogenous and exogenous peroxisome proliferator-activated receptor alpha agonists bind to the peroxisome proliferator-activated response element to exert their biological effects. Peroxisome proliferator-activated receptor alpha plays a significant therapeutic role in neurodegenerative diseases. For instance, peroxisome proliferator-activated receptor alpha agonist gemfibrozil has been shown to reduce levels of soluble and insoluble amyloid-beta in the hippocampus of Alzheimer’s disease mouse models through the autophagy-lysosomal pathway. Additionally, peroxisome proliferator-activated receptor alpha is essential for the normal development and functional maintenance of the substantia nigra, and it can mitigate motor dysfunction in Parkinson’s disease mouse models. Furthermore, peroxisome proliferator-activated receptor alpha has been found to reduce neuroinflammation and oxidative stress in various neurological diseases. In summary, peroxisome proliferator-activated receptor alpha plays a crucial role in the onset and progression of multiple nervous system diseases, and peroxisome proliferator-activated receptor alpha agonists hold promise as new therapeutic agents for the treatment of neurodegenerative diseases, providing new options for patient care.
    Related Articles | Metrics
    Organelle symphony: Nuclear factor erythroid 2-related factor 2 and nuclear factor-kappa B in stroke pathobiology
    Ziliang Hu, Mingyue Zhao, Hangyu Shen, Liangzhe Wei, Jie Sun, Xiang Gao, Yi Huang
    2026, 21 (4):  1483-1496.  doi: 10.4103/NRR.NRR-D-24-01404
    Abstract ( 27 )   PDF (5175KB) ( 4 )   Save
    Strokes include both ischemic stroke, which is mediated by a blockade or reduction in the blood supply to the brain, and hemorrhagic stroke, which comprises intracerebral hemorrhage and subarachnoid hemorrhage and is characterized by bleeding within the brain. Stroke is a lifethreatening cerebrovascular condition characterized by intricate pathophysiological mechanisms, including oxidative stress, inflammation, mitochondrial dysfunction, and neuronal injury. Critical transcription factors, such as nuclear factor erythroid 2-related factor 2 and nuclear factor kappa B, play central roles in the progression of stroke. Nuclear factor erythroid 2-related factor 2 is sensitive to changes in the cellular redox status and is crucial in protecting cells against oxidative damage, inflammatory responses, and cytotoxic agents. It plays a significant role in post-stroke neuroprotection and repair by influencing mitochondrial function, endoplasmic reticulum stress, and lysosomal activity and regulating metabolic pathways and cytokine expression. Conversely, nuclear factor-kappaB is closely associated with mitochondrial dysfunction, the generation of reactive oxygen species, oxidative stress exacerbation, and inflammation. Nuclear factor-kappaB contributes to neuronal injury, apoptosis, and immune responses following stroke by modulating cell adhesion molecules and inflammatory mediators. The interplay between these pathways, potentially involving crosstalk among various organelles, significantly influences stroke pathophysiology. Advancements in single-cell sequencing and spatial transcriptomics have greatly improved our understanding of stroke pathogenesis and offer new opportunities for the development of targeted, individualized, cell typespecific treatments. In this review, we discuss the mechanisms underlying the involvement of nuclear factor erythroid 2-related factor 2 and nuclear factor-kappa B in both ischemic and hemorrhagic stroke, with an emphasis on their roles in oxidative stress, inflammation, and neuroprotection.
    Related Articles | Metrics
    Potential targets of microglia in the treatment of neurodegenerative disea
    Wenhui Zhao , Zhongxuan Liu , Jiannan Wu , Anran Liu , Junqiang Yan
    2026, 21 (4):  1497-1511.  doi: 10.4103/NRR.NRR-D-24-01343
    Abstract ( 18 )   PDF (1129KB) ( 8 )   Save
    For diverse neurodegenerative disorders, microglial cells are activated. Furthermore, dysfunctional and hyperactivated microglia initiate mitochondrial autophagy, oxidative stress, and pathological protein accumulation, ending with neuroinflammation that exacerbates damage to dopaminergic neurons and contributes significantly to the pathology of neurodegenerative disorder. Microglial overactivation is closely associated with the secretion of pro-inflammatory cytokines, the phagocytosis of injured neurons, and the modulation of neurotoxic environments. This review summarizes the role of microglia neurodegenerative diseases, such as Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, multiple system atrophy, amyotrophic lateral sclerosis, frontotemporal dementia, progressive supranuclear palsy, cortical degeneration, Lewy body dementia, and Huntington’s disease. It also discusses novel forms of cell death such as ferroptosis, cuproptosis, disulfidptosis, and parthanatos (poly(adenosine diphosphate ribose) polymerase 1-dependent cell death), as well as the impact of regulatory factors related to microglial inflammation on microglial activation and neuroinflammation. The aim is to identify potential targets for microglial cell therapy in neurodegenerative diseases.
    Related Articles | Metrics
    Generation and clearance of myelin debris after spinal cord injury
    Chaoyuan Li , Wenqi Luo , Irshad Hussain , Renrui Niu , Xiaodong He , Chunyu Xiang , Fengshuo Guo , Wanguo Liu, Rui Gu
    2026, 21 (4):  1512-1527.  doi: 10.4103/NRR.NRR-D-24-01405
    Abstract ( 22 )   PDF (1809KB) ( 10 )   Save
    Traumatic spinal cord injury often leads to the disintegration of nerve cells and axons, resulting in a substantial accumulation of myelin debris that can persist for years. The abnormal buildup of myelin debris at sites of injury greatly impedes nerve regeneration, making the clearance of debris within these microenvironments crucial for effective post-spinal cord injury repair. In this review, we comprehensively outline the mechanisms that promote the clearance of myelin debris and myelin metabolism and summarize their roles in spinal cord injury. First, we describe the composition and characteristics of myelin debris and explain its effects on the injury site. Next, we introduce the phagocytic cells involved in myelin debris clearance, including professional phagocytes (macrophages and microglia) and non-professional phagocytes (astrocytes and microvascular endothelial cells), as well as other cells that are also proposed to participate in phagocytosis. Finally, we focus on the pathways and associated targets that enhance myelin debris clearance by phagocytes and promote lipid metabolism following spinal cord injury. Our analysis indicates that myelin debris phagocytosis is not limited to monocyte-derived macrophages, but also involves microglia, astrocytes, and microvascular endothelial cells. By modulating the expression of genes related to phagocytosis and lipid metabolism, it is possible to modulate lipid metabolism disorders and influence inflammatory phenotypes, ultimately affecting the recovery of motor function following spinal cord injury. Additionally, therapies such as targeted mitochondrial transplantation in phagocytic cells, exosome therapy, and repeated trans-spinal magnetic stimulation can effectively enhance the removal of myelin debris, presenting promising potential for future applications.
    Related Articles | Metrics
    Neuropsychiatric symptoms and apolipoprotein E genotypes in neurocognitive disorders
    Madia Lozupone, Ivana Leccisotti , Anita Mollica , Giuseppe Berardino , Maria Claudia Moretti , Mario Altamura , Antonello Bellomo, Antonio Daniele , Vittorio Dibello , Vincenzo Solfrizzi , Emanuela Resta, Francesco Panza
    2026, 21 (4):  1528-1541.  doi: 10.4103/NRR.NRR-D-24-01274
    Abstract ( 10 )   PDF (3926KB) ( 3 )   Save
    Complex genetic relationships between neurodegenerative disorders and neuropsychiatric symptoms have been shown, suggesting shared pathogenic mechanisms and emphasizing the potential for developing common therapeutic targets. Apolipoprotein E (APOE) genotypes and their corresponding protein (ApoE) isoforms may influence the biophysical properties of the cell membrane lipid bilayer. However, the role of APOE in central nervous system pathophysiology extended beyond its lipid transport function. In the present review article, we analyzed the links existing between APOE genotypes and the neurobiology of neuropsychiatric symptoms in neurodegenerative and vascular diseases. APOE genotypes (APOE ε2, APOE ε3, and APOE ε4) were implicated in common mechanisms underlying a wide spectrum of neurodegenerative diseases, including sporadic Alzheimer’s disease, synucleinopathies such as Parkinson’s disease and Lewy body disease, stroke, and traumatic brain injury. These shared pathways often involved neuroinflammation, abnormal protein accumulation, or responses to acute detrimental events. Across these conditions, APOE variants are believed to contribute to the modulation of inflammatory responses, the regulation of amyloid and tau pathology, as well as the clearance of proteins such as α-synuclein. The bidirectional interactions among ApoE, amyloid and mitochondrial metabolism, immunomodulatory effects, neuronal repair, and remodeling underscored the complexity of ApoE’s role in neuropsychiatric symptoms associated with these conditions since from early phases of cognitive impairment such as mild cognitive impairment and mild behavioral impairment. Besides ApoE-specific isoforms’ link to increased neuropsychiatric symptoms in Alzheimer’s disease (depression, psychosis, aberrant motor behaviors, and anxiety, not apathy), the APOE ε4 genotype was also considered a significant genetic risk factor for Lewy body disease and its worse cognitive outcomes. Conversely, the APOE ε2 variant has been observed not to exert a protective effect equally in all neurodegenerative diseases. Specifically, in Lewy body disease, this variant may delay disease onset, paralleling its protective role in Alzheimer’s disease, although its role in frontotemporal dementia is uncertain. The APOE ε4 genotype has been associated with adverse cognitive outcomes across other various neurodegenerative conditions. In Parkinson’s disease, the APOE ε4 allele significantly impacted cognitive performance, increasing the risk of developing dementia, even in cases of pure synucleinopathies with minimal co-pathology from Alzheimer’s disease. Similarly, in traumatic brain injury, recovery rates varied, with APOE ε4 carriers demonstrating a greater risk of poor long-term cognitive outcomes and elevated levels of neuropsychiatric symptoms. Furthermore, APOE ε4 influenced the age of onset and severity of stroke, as well as the likelihood of developing stroke-associated dementia, potentially due to its role in compromising endothelial integrity and promoting blood–brain barrier dysfunction.
    Related Articles | Metrics
    Cell therapy rejuvenates the neuroglial-vascular unit
    Bandy Chen
    2026, 21 (4):  1542-1543.  doi: 10.4103/NRR.NRR-D-24-01359
    Abstract ( 13 )   PDF (2123KB) ( 2 )   Save
    The rise of the aging population parallels the rapidly increasing cases of neurological disorders. This puts pressure on scientists and physicians to find novel methods that can prevent and treat neurodegeneration. The brain is made up of a complex network of different cell types that work in tandem to maintain systemic homeostasis. These cells include vascular cells (endothelial cells, pericytes, and smooth muscle cells), glial cells (astrocytes, microglia, and oligodendrocytes), and neurons that have different functions to complement each other and form the neuroglial-vascular unit (NVU). These elements act in concert to orchestrate neurovascular coupling and maintain blood–brain barrier (BBB) integrity. Unlike other systems in the human body, the brain has limited regenerative capacity. To overcome this limitation, novel approaches in stem cell biology, immune cell engineering, and bioengineering work in tandem to repair, replace, and restore function in the central nervous system. Due to the diverse cell types of the central nervous system, cell therapy allows cell type-specific modifications to precisely target neural circuitries and advance personalized medicine. This puts cell therapy at the forefront as a potential treatment to rejuvenate the cerebral landscape. This perspective focuses on the impact of cell therapy through the lens of the NVU.
    Related Articles | Metrics
    New insights into the homeostatic role of Lrig1 in different neurogenic niches: Implications for neuronal regeneration
    Ana Paula De Vincenti, Fernanda Ledda, Gustavo Paratcha
    2026, 21 (4):  1544-1545.  doi: 10.4103/NRR.NRR-D-24-01333
    Abstract ( 23 )   PDF (990KB) ( 4 )   Save
    Stem cell proliferation is tightly regulated in developing and adult tissues through the coordinated action of cell-intrinsic and extracellular signals. Although many extracellular cues were identified, the cell-intrinsic mechanisms underlying the decision of a stem cell to proliferate, enter a dormant quiescent state or differentiate into a specific cell type remains incompletely understood. Several previous studies have shown that the leucine-rich repeats and immunoglobulin-like domains 1 (Lrig1) transmembrane protein is a tumor suppressor and a stem cell marker. Lrig1 is critically implicated in the cell-autonomous control of stem cell proliferation and/or quiescence in several tissues, including the epidermis, intestine, corneal epithelium, hard palate of the oral mucosa and brain (Herdenberg and Hedman, 2023). In most of these studies, Lrig1 controls adult tissue homeostasis by regulating proliferation and/or quiescence in different stem cell compartments. Thus, Lrig1 regulates proliferation by inhibiting epidermal growth factor (EGF) receptor (EGFR) signaling in highly proliferative stem cells. However, Lrig1 has also been reported as a signature molecule of diverse quiescent stem cell populations, including epidermal and intestinal stem cells. Although it is widely accepted that in these cell populations, Lrig1 induces quiescence by reducing EGFR activation, it has also been recently proposed that Lrig1 might regulate quiescence by enhancing bone morphogenetic protein (BMP) signaling, a well-known effector of stem cell-cycle exit and quiescence (Herdenberg et al., 2021). In relation to this, genetic ablation of Lrig1 in glioblastoma stem cells (GSCs) results in higher proliferation in response to EGFR and simultaneous poor signaling response to BMP that may underlie their lack of response to quiescence in vivo (Ferguson et al., 2022). Likewise, to normal adult neural stem cells (NSCs), GSCs can likely adopt a dynamic range of cellular states from quiescence to active proliferation. This feature enables GSCs to evade anti-mitotic therapies and lead to tumor recurrence through their reversible cellcycle arrest. Therefore, targeting Lrig1 to balance the proportion of quiescent and proliferative subpopulations of GSCs could contribute to the design of successful therapies to suppress tumor re-growth.
    Related Articles | Metrics
    Noradrenergic excitation of astrocytes supports cognitive reserve
    Robert Zorec , Alexei Verkhratsky
    2026, 21 (4):  1546-1547.  doi: 10.4103/NRR.NRR-D-24-01395
    Abstract ( 12 )   PDF (1202KB) ( 13 )   Save
    The concept of the brain cognitive reserve is derived from the well-acknowledged notion that the degree of brain damage does not always match the severity of clinical symptoms and neurological/ cognitive outcomes. It has been suggested that the size of the brain (brain reserve) and the extent of neural connections acquired through life (neural reserve) set a threshold beyond which noticeable impairments occur. In contrast, cognitive reserve refers to the brain’s ability to adapt and reorganize structurally and functionally to resist damage and maintain function, including neural reserve and brain maintenance, resilience, and compensation (Verkhratsky and Zorec, 2024). We propose that noradrenergic regulation of astrocytes plays a key role in defining cognitive reserve. Astrocytes support neural homeostasis through multiple mechanisms and contribute to the pathogenesis of all neurological disorders (Verkhratsky et al., 2023). Astrocytes are key components in maintaining cognitive reserve; they contribute to shaping cytoarchitecture of the nervous tissue and synaptic transmission, thus supporting neural reserve and neural maintenance; astrocytes are central elements of brain defense and regeneration thus supporting brain resilience and compensation. We present an overview of the role of the noradrenergic innervation of astrocytes in both normal and neurodegenerative states, then we point out that the demise of the noradrenergic system is an early event in many, if not all neurodegenerative diseases, acting through astrocytes and limiting their support of cognitive reserve.
    Related Articles | Metrics
    Chromatin accessibility regulates axon regeneration
    Isa Samad, Brett J. Hilton
    2026, 21 (4):  1548-1549.  doi: 10.4103/NRR.NRR-D-24-01307
    Abstract ( 15 )   PDF (1063KB) ( 0 )   Save
    Central nervous system (CNS) axons fail to regenerate following brain or spinal cord injury (SCI), which typically leads to permanent neurological deficits. Peripheral nervous system axons, however, can regenerate following injury. Understanding the mechanisms that underlie this difference is key to developing treatments for CNS neurological diseases and injuries characterized by axonal damage. To initiate repair after peripheral nerve injury, dorsal root ganglion (DRG) neurons mobilize a pro-regenerative gene expression program, which facilitates axon outgrowth. Chromatin accessibility actively regulates this genetic program by controlling how easily transcriptional machinery can bind to DNA (Palmisano et al., 2019; Cheng et al., 2023). Thus, the molecular machinery driving changes in chromatin accessibility is a critical therapeutic target to coax axon regeneration following injury or disease. Exploiting this machinery through genetic or pharmacological interventions in CNS neurons, which fail to activate or sustain the proregenerative program following injury, represents a promising strategy to enhance regeneration. In this perspective, we examine recent discoveries on how chromatin accessibility regulates axon regeneration. We start by describing recent work that focuses on how two different posttranslational modifications of histones, acetylation and methylation, influence gene expression and axon regeneration following injury. We then describe the major unaddressed questions in this field of research. Ultimately, we expect that deeper insights into this process will uncover therapeutics to elicit regeneration and repair of the CNS following injury or disease.
    Related Articles | Metrics
    Therapeutic potential of circular RNAs in neurovascular remodeling after stroke
    Zhenguo Yang, Chi Kwan Tsang
    2026, 21 (4):  1550-1551.  doi: 10.4103/NRR.NRR-D-24-01291
    Abstract ( 13 )   PDF (739KB) ( 2 )   Save
    Stroke-induced alterations in cerebral blood flow trigger neurovascular remodeling, as manifested by the blood–brain barrier dysfunction and subsequent neurovascular repair activities such as angiogenesis. This process involves neurovascular communication that facilitates the transport of mediators among cerebrovascular endothelial cells, pericytes, glial cells, and neurons, thereby transmitting signals from donor to recipient cells to elicit a collaborative response. Current research progress has implicated that circular RNAs (circRNAs) may play a crucial role in intercellular communication through extracellular vesicles (EVs). CircRNAs may function as messengers that are involved in the regulation of transcription and translation in both donor and recipient cells. These cellular functions of circRNAs can be mediated by the competitive binding of circRNAs to microRNAs (miRNAs) and RNA-binding proteins, which subsequently influence the biological functions of their targets. For example, our recent studies showed that circOGDH acts as a sponge for miR-5112, while circ-FoxO3 interacts with both mTOR and E2F1, thereby facilitating neurovascular remodeling (Liu et al., 2022; Yang et al., 2022). However, the precise roles of circRNAs in neurovascular remodeling and their specific functions in intercellular communications remain obscured. In this perspective, we will highlight the crucial emerging roles of circRNAs in relation to neurovascular remodeling and the therapeutic potential of targeting circRNAs in stroke.
    Related Articles | Metrics
    mTORC1 and mTORC2 synergy in human neural development, disease, and regeneration
    Navroop K. Dhaliwal, Julien Muffat, Yun Li
    2026, 21 (4):  1552-1553.  doi: 10.4103/NRR.NRR-D-24-00961
    Abstract ( 15 )   PDF (8160KB) ( 4 )   Save
    The mechanistic target of rapamycin (mTOR) is a serine/threonine kinase that plays a pivotal role in cellular growth, proliferation, survival, and metabolism. In the central nervous system (CNS), the mTOR pathway regulates diverse aspects of neural development and function. Genetic mutations within the mTOR pathway lead to severe neurodevelopmental disorders, collectively known as “mTORopathies” (Crino, 2020). Dysfunctions of mTOR, including both its hyperactivation and hypoactivation, have also been implicated in a wide spectrum of other neurodevelopmental and neurodegenerative conditions, highlighting its importance in CNS health. Molecularly, mTOR functions as the catalytic subunit of two distinct complexes: mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2). These two complexes are defined by their unique components and substrate specificities, which in turn activate distinct downstream signaling cascades that underpin their diverse roles in the CNS. mTORC1, characterized by the presence of RPTOR (Regulatory Associated Protein of MTOR complex 1), is known to govern protein synthesis and autophagy. In contrast, mTORC2, which contains RICTOR (RPTOR Independent Companion of MTOR complex 2), is less understood but is known to regulate the actin cytoskeleton and metabolism.
    Related Articles | Metrics
    Immunoproteasome as a therapeutic target in obesity-related brain inflammation and metabolic disorders
    Javiera Álvarez-Indo, Nicolás Albornoz, Andrea Soza , Patricia V. Burgos
    2026, 21 (4):  1554-1555.  doi: 10.4103/NRR.NRR-D-24-01358
    Abstract ( 11 )   PDF (704KB) ( 1 )   Save
    Obesity is widely recognized as a global epidemic, primarily driven by an imbalance between energy expenditure and caloric intake associated with a sedentary lifestyle. Diets high in carbohydrates and saturated fats, particularly palmitic acid, are potent inducers of chronic low-grade inflammation, largely due to disruptions in glucose metabolism and the onset of insulin resistance (Qiu et al., 2022). While many organs are affected, the brain, specifically the hypothalamus, is among the first to exhibit inflammation in response to an unhealthy diet, suggesting that obesity may, in fact, be a brain-centered disease with neuroinflammation as a central factor (Thaler et al., 2012).
    Related Articles | Metrics
    Adenosine: A key player in neuroinflammation
    Qilin Guo, Rhea Seth, Wenhui Huang
    2026, 21 (4):  1556-1557.  doi: 10.4103/NRR.NRR-D-24-01486
    Abstract ( 14 )   PDF (2369KB) ( 4 )   Save
    Neuroinflammation, the inflammatory response of the central nervous system (CNS), is a common feature of many neurological disorders such as sepsis-associated encephalopathy (SAE), multiple sclerosis (MS), and Parkinson’s disease (PD). Prior studies identified cytokines (e.g., tumor necrosis factor [TNF], interleukin [IL]-1, and IL-6) delivered by resident glial cells and brain-invading peripheral immune cells as the major contributor to neuroinflammation (Becher et al., 2017). In addition to pro-inflammatory cytokines, elevated levels of extracellular purine molecules such as adenosine triphosphate (ATP) and adenosine can be detected upon any pathological insults (e.g., injury, ischemia, and hypoxia), contributing to the progression of neurological disorders (Borea et al., 2017).
    Related Articles | Metrics
    Neuronal swelling implicated in functional recovery after spinal cord injury
    Qiang Li
    2026, 21 (4):  1558-1559.  doi: 10.4103/NRR.NRR-D-24-01556
    Abstract ( 15 )   PDF (659KB) ( 2 )   Save
    Spinal cord injury (SCI) often results in permanent dysfunction of locomotion, sensation, and autonomic regulation, imposing a substantial burden on both individuals and society (Anjum et al., 2020). SCI has a complex pathophysiology: an initial primary injury (mechanical trauma, axonal disruption, and hemorrhage) is followed by a progressive secondary injury cascade that involves ischemia, neuronal loss, and inflammation. Given the challenges in achieving regeneration of the injured spinal cord, neuroprotection has been at the forefront of clinical research. Yet, current neuroprotective therapeutic efficiency is limited (Anjum et al., 2020). To develop effective neuroprotective interventions for SCI patients, a deeper understanding of SCI pathophysiology is undoubtedly required.
    Related Articles | Metrics
    Peripheral nervous system and gut microbiota: Emerging evidence on increased mechanistic understanding to reveal innovative strategies for peripheral nerve regeneration
    Giulia Ronchi , Matilde Cescon, Giovanna Gambarotta, Kirsten Haastert-Talini
    2026, 21 (4):  1560-1561.  doi: 10.4103/NRR.NRR-D-24-01310
    Abstract ( 14 )   PDF (878KB) ( 3 )   Save
    The gut microbiota: The human body is colonized by a diverse and complex microbial community – including bacteria, viruses, archaea, and unicellular eukaryotes – that plays a central role in human wellbeing. Indeed, microbiota is crucial for several functions, including host metabolism, physiology, maintenance of the intestinal epithelial integrity, nutrition, and immune function, earning it the designation of a “vital organ” (Guinane and Cotter, 2013).
    Related Articles | Metrics
    Neurodegenerative processes of aging: A perspective of restoration through insulin-like growth factor-1
    Rosana Crespo, Claudia Hereñú
    2026, 21 (4):  1562-1563.  doi: 10.4103/NRR.NRR-D-24-01595
    Abstract ( 13 )   PDF (1207KB) ( 2 )   Save
    The aging process is an inexorable fact throughout our lives and is considered a major factor in developing neurological dysfunctions associated with cognitive, emotional, and motor impairments. Aging-associated neurodegenerative diseases are characterized by the progressive loss of neuronal structure and function. Numerous efforts and approaches are underway to enhance the quality of life and health span, including parabiosis with plasma pro-youthful factors, therapy with trophic factors, Klotho protein, caloric restriction, mitochondrial function, multivitamin supplementation, mesenchymal cells, and rejuvenation with Yamanaka genes, among several others (Kelly et al., 2024; Viña and Borras, 2024).
    Related Articles | Metrics
    Melatonin and mitochondrial stress: New insights into age-related neurodegeneration
    Silvia Carloni , Francesca Luchetti , Maria Gemma Nasoni, Walter Balduini , Walter Manucha , Russel J. Reiter
    2026, 21 (4):  1564-1565.  doi: 10.4103/NRR.NRR-D-24-01380
    Abstract ( 20 )   PDF (522KB) ( 7 )   Save
    Aging, mitochondria, and neurodegenerative diseases: Aging is often viewed as the buildup of changes that lead to the gradual transformations associated with getting older, along with a rising likelihood of disease and mortality. Although organism-wide deterioration is observed during aging, organs with high metabolic demand, such as the brain, are more vulnerable. Consequently, most neurodegenerative diseases occur in the aged population. Even in the healthy brain, the normal aging process is associated with several features of the neurodegenerative process, including neuroinflammation, and brain shrinkage, with a progressive decline in physiological functions (Lee and Kim, 2022).
    Related Articles | Metrics
    Imaging alpha-synuclein pathology in Parkinson’s disease
    Ruiqing Ni
    2026, 21 (4):  1566-1567.  doi: 10.4103/NRR.NRR-D-24-01348
    Abstract ( 12 )   PDF (2097KB) ( 2 )   Save
    Parkinson’s disease (PD) is the second most common neurodegenerative disorder. The clinical manifestations of PD include motor symptoms, such as bradykinesia, resting tremor, rigidity, and nonmotor symptoms, which include disturbances in sleep, gastrointestinal function, and olfaction. PD misdiagnosis rates have been reported to reach approximately 30%, partly owing to the heterogeneity of parkinsonism with non-PD pathologies, and the differential diagnosis of PD from neurodegenerative diseases such as multiple systemic atrophy (MSA) and progressive supranuclear palsy poses another unmet need. These nonmotor symptoms may emerge more than a decade prior to the onset of motor impairments. Pathologically, PD is characterized by the accumulation of Lewy bodies (LBs), which are composed of misfolded alpha-synuclein, and the early loss of dopaminergic neurons. Recent studies have introduced two biomarker-based systems for PD research: the SyNeurGe system and the neuronal alpha-synuclein disease integrated staging system (Höglinger et al., 2024; Simuni et al., 2024). Misfolded alpha-synuclein has been shown to spread between cells, serving as a template for further alpha-synuclein misfolding. Although the precise etiological role of alphasynuclein in PD has not been fully elucidated, a recent study has revealed heterogeneity in the trajectories of alpha-synuclein pathology in PD and the toxicity of alpha-synuclein especially soluble aggregates (Mastenbroek et al., 2024). Currently, there are several clinical trials targeting alphasynuclein as disease modifying treatments of PD, either by using active or passive immunotherapy, by preventing alpha-synuclein aggregation, or by disaggregation of existing complexes.
    Related Articles | Metrics
    Determinants of alpha-synuclein pathogenesis in Parkinson’s disease
    Oriol Bárcenas, Marc Estivill-Alonso, Salvador Ventura
    2026, 21 (4):  1568-1569.  doi: 10.4103/NRR.NRR-D-24-01357
    Abstract ( 20 )   PDF (722KB) ( 4 )   Save
    Alpha-synuclein and Parkinson’s disease: Neuronal damage and inflammation caused by the aggregation of alpha-synuclein (α-syn) are central to a group of disorders known as synucleopathies, which includes Parkinson’s disease (PD), dementia with Lewy bodies, and multiple system atrophy, among others. PD, the most common synucleinopathy, is the second most prevalent neurodegenerative disease after Alzheimer’s disease, and it is the fastest growing. Its primary hallmark is the degeneration of dopaminergic neurons in the substantia nigra pars compacta, disrupting the communication with the striatum. This has adverse motor and nonmotor effects, with the most prominent symptoms being tremors, rigidity, instability, and gait difficulties. While most patients have a late onset (60+ years), certain dominant genetic mutations in the gene encoding α-syn are associated with earlier onset. Despite the severity and prevalence of the disease, no treatments that halt or modify the pathology progression exist, with available therapies providing only symptomatic relief for motor symptoms (Vázquez-Vélez and Zoghbi, 2021).
    Related Articles | Metrics
    Molecular biomarkers in GNAO1 encephalopathies
    Vladimir L. Katanaev , Jana Valnohova
    2026, 21 (4):  1570-1571.  doi: 10.4103/NRR.NRR-D-24-01550
    Abstract ( 11 )   PDF (2246KB) ( 2 )   Save
    GNAO1-associated disorder is a rare disease and an example of developmental and epileptic encephalopathies. Caused by ca. 150 different dominant missense mutations in the gene encoding the major neuronal G protein Gαo, it spans a wide range of neurological clinical manifestations, that may include epileptic seizures, motor dysfunctions, developmental and intellectual delay, and other symptoms (Sáez González et al., 2023). As of today, tools to predict the disease severity and prognosis, as well as responsiveness to current and developing therapies, are missing. Our recent study (Solis et al., 2024) has peeled, layer by layer, cellular and molecular deficiencies of a large panel of pathogenic GNAO1 variants, seeking quantitative correlations with the clinical disease manifestations. This analysis resulted in the identification of two pathogenic characteristics that may serve as molecular biomarkers of the disease. The first is the drop in plasma membrane localization of certain pathogenic variants, which correlates with the existence of seizures in the anamnesis. The second is the strength of neomorphic interactions with the molecular chaperone Ric8B, which correlates with the overall disease severity. Identification of the predictive disease biomarkers sheds light on the molecular etiology of GNAO1 encephalopathy and is expected to improve patient care, follow-up, and treatment.
    Related Articles | Metrics
    Efferocytosis and retinal clean-up: Role of histone deacetylase 3 in ischemic retinopathy
    Abdelrahman Y. Fouda , Esraa Shosha
    2026, 21 (4):  1572-1573.  doi: 10.4103/NRR.NRR-D-24-01342
    Abstract ( 15 )   PDF (2301KB) ( 3 )   Save
    Ischemic retinopathy is a leading cause of blindness: Ischemic retinopathies including diabetic retinopathy (DR), retinopathy of prematurity, and retinal artery and vein occlusion are major causes of visual impairment. Ischemic retinopathy can be acute, such as in central or branch retinal artery occlusion, or chronic, such as with DR (Figure 1). Although the causes of retinopathies are diverse, one pathogenic event shared by these conditions is the myeloid cell response to retinal ischemia (Shahror et al., 2024a). The ischemia-induced neurovascular injury results in progressive cell death by apoptosis, causing neurodegeneration and loss of vascular cells. Concurrently, there is activation and proliferation of microglia, non-parenchymal macrophages (such as perivascular macrophages), and recruitment of blood-borne (infiltrating) monocytes. These activated cells (collectively termed “myeloid cells”) play either a protective or deleterious role after retinal injury depending on their molecular profile and activation state. Our publications and others recently introduced the concept that myeloid cells (microglia and macrophages) can be geared to a pro-resolving phenotype to protect the retina against injury (Shahror et al., 2024a). This protective effect can be conferred by the secretion of reparative cytokines. Moreover, the direct interaction of myeloid cells with dead cells in the inner retina following ischemic injury has been described in retinal ischemia-reperfusion injury, suggesting microglia/macrophage-mediated phagocytic clearance of dead cells or efferocytosis (Abcouwer et al., 2021).
    Related Articles | Metrics
    Recombinant tissue plasminogen activator protects neurons after intracerebral hemorrhage through activating the PI3K/AKT/mTOR pathway
    Jie Jing, Shiling Chen, Xuan Wu, Jingfei Yang, Xia Liu, Jiahui Wang, Jingyi Wang, Yunjie Li, Ping Zhang, Zhouping Tang
    2026, 21 (4):  1574-1585.  doi: 10.4103/NRR.NRR-D-23-01953
    Abstract ( 20 )   PDF (9319KB) ( 7 )   Save
    Recombinant tissue plasminogen activator is commonly used for hematoma evacuation in minimally invasive surgery following intracerebral hemorrhage. However, during minimally invasive surgery, recombinant tissue plasminogen activator may come into contact with brain tissue. Therefore, a thorough assessment of its safety is required. In this study, we established a mouse model of intracerebral hemorrhage induced by type VII collagenase. We observed that the administration of recombinant tissue plasminogen activator without hematoma aspiration significantly improved the neurological function of mice with intracerebral hemorrhage, reduced pathological damage, and lowered the levels of apoptosis and autophagy in the tissue surrounding the hematoma. In an in vitro model of intracerebral hemorrhage using primary cortical neurons induced by hemin, the administration of recombinant tissue plasminogen activator suppressed neuronal apoptosis, autophagy, and endoplasmic reticulum stress. Transcriptome sequencing analysis revealed that recombinant tissue plasminogen activator upregulated the phosphoinositide 3-kinase/RAC-alpha serine/threonine-protein kinase/mammalian target of rapamycin pathway in neurons. Moreover, the phosphoinositide 3-kinase inhibitor LY294002 abrogated the neuroprotective effects of recombinant tissue plasminogen activator in inhibiting excessive apoptosis, autophagy, and endoplasmic reticulum stress. Furthermore, to specify the domain of recombinant tissue plasminogen activator responsible for its neuroprotective effects, various inhibitors were used to target distinct domains. It has been revealed that the epidermal growth factor receptor inhibitor AG-1478 reversed the effect of recombinant tissue plasminogen activator on the phosphoinositide 3-kinase/RAC-alpha serine/threonineprotein kinase/mammalian target of rapamycin pathway. These findings suggest that recombinant tissue plasminogen activator exerts a direct neuroprotective effect on neurons following intracerebral hemorrhage, possibly through activation of the phosphoinositide 3-kinase/RAC-alpha serine/threonine-protein kinase/mammalian target of rapamycin pathway.
    Related Articles | Metrics
    Enhancing neural stem cell integration in the injured spinal cord through targeted PTEN modulation
    Simay Genişcan, Hee Hwan Park, Hyung Soon Kim, Seokjin Yoo, Hyunmi Kim, Byeong Seong Jang, Dong Hoon Hwang, Kevin K Park, Byung Gon Kim
    2026, 21 (4):  1586-1594.  doi: 10.4103/NRR.NRR-D-24-00455
    Abstract ( 14 )   PDF (6696KB) ( 14 )   Save
    Spinal cord injury results in permanent loss of neurological functions due to severance of neural networks. Transplantation of neural stem cells holds promise to repair disrupted connections. Yet, ensuring the survival and integration of neural stem cells into the host neural circuit remains a formidable challenge. Here, we investigated whether modifying the intrinsic properties of neural stem cells could enhance their integration post-transplantation. We focused on phosphatase and tensin homolog (PTEN), a well-characterized tumor suppressor known to critically regulate neuronal survival and axonal regeneration. By deleting Pten in mouse neural stem cells, we observed increased neurite outgrowth and enhanced resistance to neurotoxic environments in culture. Upon transplantation into injured spinal cords, Pten-deficient neural stem cells exhibited higher survival and more extensive rostrocaudal distribution. To examine the potential influence of partial PTEN suppression, rat neural stem cells were treated with short hairpin RNA targeting PTEN, and the PTEN knockdown resulted in significant improvements in neurite growth, survival, and neurosphere motility in vitro. Transplantation of shPTEN-treated neural stem cells into the injured spinal cord also led to an increase in graft survival and migration to an extent similar to that of complete deletion. Moreover, PTEN suppression facilitated neurite elongation from NSC-derived neurons migrating from the lesion epicenter. These findings suggest that modifying intrinsic signaling pathways, such as PTEN, within neural stem cells could bolster their therapeutic efficacy, offering potential avenues for future regenerative strategies for spinal cord injury.
    Related Articles | Metrics
    Low-density lipoprotein receptor–related protein 1 mediates α-synuclein transmission from the striatum to the substantia nigra in animal models of Parkinson’s disease
    Hanjiang Luo, Caixia Peng, Chengli Wu, Chengwei Liu, Qinghua Li, Shun Yu, Jia Liu, Min Chen
    2026, 21 (4):  1595-1606.  doi: 10.4103/NRR.NRR-D-23-01965
    Abstract ( 18 )   PDF (5170KB) ( 6 )   Save
    α-Synuclein accumulation and transmission are vital to the pathogenesis of Parkinson’s disease, although the mechanisms underlying misfolded α-synuclein accumulation and propagation have not been conclusively determined. The expression of low-density lipoprotein receptor–related protein 1, which is abundantly expressed in neurons and considered to be a multifunctional endocytic receptor, is elevated in the neurons of patients with Parkinson’s disease. However, whether there is a direct link between low-density lipoprotein receptor–related protein 1 and α-synuclein aggregation and propagation in Parkinson’s disease remains unclear. Here, we established animal models of Parkinson’s disease by inoculating monkeys and mice with α-synuclein pre-formed fibrils and observed elevated low-density lipoprotein receptor–related protein 1 levels in the striatum and substantia nigra, accompanied by dopaminergic neuron loss and increased α-synuclein levels. However, low-density lipoprotein receptor–related protein 1 knockdown efficiently rescued dopaminergic neurodegeneration and inhibited the increase in α-synuclein levels in the nigrostriatal system. In HEK293A cells overexpressing α-synuclein fragments, low-density lipoprotein receptor–related protein 1 levels were upregulated only when the N-terminus of α-synuclein was present, whereas an α-synuclein fragment lacking the N-terminus did not lead to low-density lipoprotein receptor–related protein 1 upregulation. Furthermore, the N-terminus of α-synuclein was found to be rich in lysine residues, and blocking lysine residues in PC12 cells treated with α-synuclein pre-formed fibrils effectively reduced the elevated low-density lipoprotein receptor–related protein 1 and α-synuclein levels. These findings indicate that low-density lipoprotein receptor–related protein 1 regulates pathological transmission of α-synuclein from the striatum to the substantia nigra in the nigrostriatal system via lysine residues in the α-synuclein N-terminus.
    Related Articles | Metrics
    Injury-induced KIF4A neural expression and its role in Schwann cell proliferation suggest a dual function for this kinesin in neural regeneration
    Patrícia D. Correia, Bárbara M. de Sousa, Jesús Chato-Astrain, Joana Paes de Faria, Veronica Estrada, João B. Relvas, Hans W. Müller , Víctor Carriel, Frank Bosse, Sandra I. Vieira
    2026, 21 (4):  1607-1620.  doi: 10.4103/NRR.NRR-D-24-00232
    Abstract ( 25 )   PDF (8294KB) ( 6 )   Save
    Contrary to the adult central nervous system, the peripheral nervous system has an intrinsic ability to regenerate that relies on the expression of regenerationassociated genes, such as some kinesin family members. Kinesins contribute to nerve regeneration through the transport of specific cargo, such as proteins and membrane components, from the cell body towards the axon periphery. We show here that KIF4A, associated with neurodevelopmental disorders and previously believed to be only expressed during development, is also expressed in the adult vertebrate nervous system and up-regulated in injured peripheral nervous system cells. KIF4A is detected both in the cell bodies and regrowing axons of injured neurons, consistent with its function as an axonal transporter of cargoes such as β1-integrin and L1CAM. Our study further demonstrates that KIF4A levels are greatly increased in Schwann cells from injured distal nerve stumps, particularly at a time when they are reprogrammed into an essential proliferative repair phenotype. Moreover, Kif4a mRNA levels were approximately ~6-fold higher in proliferative cultured Schwann cells compared with non-proliferative ones. A hypothesized function for Kif4a in Schwann cell proliferation was further confirmed by Kif4a knockdown, as this significantly reduced Schwann cell proliferation in vitro. Our findings show that KIF4A is expressed in adult vertebrate nervous systems and is up-regulated following peripheral injury. The timing of KIF4A up-regulation, its location during regeneration, and its proliferative role, all suggest a dual role for this protein in neuroregeneration that is worth exploring in the future.
    Related Articles | Metrics
    A radiomics approach for predicting gait freezing in Parkinson’s disease based on resting-state functional magnetic resonance imaging indices: A cross-sectional study
    Miaoran Guo, Hu Liu, Long Gao, Hongmei Yu, Yan Ren, Yingmei Li, Huaguang Yang, Chenghao Cao, Guoguang Fan
    2026, 21 (4):  1621-1627.  doi: 10.4103/NRR.NRR-D-23-01392
    Abstract ( 16 )   PDF (2917KB) ( 3 )   Save
    Freezing of gait is a significant and debilitating motor symptom often observed in individuals with Parkinson’s disease. Resting-state functional magnetic resonance imaging, along with its multi-level feature indices, has provided a fresh perspective and valuable insight into the study of freezing of gait in Parkinson’s disease. It has been revealed that Parkinson’s disease is accompanied by widespread irregularities in inherent brain network activity. However, the effective integration of the multi-level indices of resting-state functional magnetic resonance imaging into clinical settings for the diagnosis of freezing of gait in Parkinson’s disease remains a challenge. Although previous studies have demonstrated that radiomics can extract optimal features as biomarkers to identify or predict diseases, a knowledge gap still exists in the field of freezing of gait in Parkinson’s disease. This cross-sectional study aimed to evaluate the ability of radiomics features based on multi-level indices of resting-state functional magnetic resonance imaging, along with clinical features, to distinguish between Parkinson’s disease patients with and without freezing of gait. We recruited 28 patients with Parkinson’s disease who had freezing of gait (15 men and 13 women, average age 63 years) and 30 patients with Parkinson’s disease who had no freezing of gait (16 men and 14 women, average age 64 years). Magnetic resonance imaging scans were obtained using a 3.0T scanner to extract the mean amplitude of low-frequency fluctuations, mean regional homogeneity, and degree centrality. Neurological and clinical characteristics were also evaluated. We used the least absolute shrinkage and selection operator algorithm to extract features and established feedforward neural network models based solely on resting-state functional magnetic resonance imaging indicators. We then performed predictive analysis of three distinct groups based on resting-state functional magnetic resonance imaging indicators indicators combined with clinical features. Subsequently, we conducted 100 additional five-fold cross-validations to determine the most effective model for each classification task and evaluated the performance of the model using the area under the receiver operating characteristic curve. The results showed that when differentiating patients with Parkinson’s disease who had freezing of gait from those who did not have freezing of gait, or from healthy controls, the models using only the mean regional homogeneity values achieved the highest area under the receiver operating characteristic curve values of 0.750 (with an accuracy of 70.9%) and 0.759 (with an accuracy of 65.3%), respectively. When classifying patients with Parkinson’s disease who had freezing of gait from those who had no freezing of gait, the model using the mean amplitude of low-frequency fluctuation values combined with two clinical features achieved the highest area under the receiver operating characteristic curve of 0.847 (with an accuracy of 74.3%). The most significant features for patients with Parkinson’s disease who had freezing of gait were amplitude of low-frequency fluctuation alterations in the left parahippocampal gyrus and two clinical characteristics: Montreal Cognitive Assessment and Hamilton Depression Scale scores. Our findings suggest that radiomics features derived from resting-state functional magnetic resonance imaging indices and clinical information can serve as valuable indices for the identification of freezing of gait in Parkinson’s disease.
    Related Articles | Metrics
    Overexpression of the inwardly rectifying potassium channel Kir4.1 or Kir4.1 Tyr9 Asp in Müller cells exerts neuroprotective effects in an experimental glaucoma model
    Fang Li, Zhen Li, Shuying Li, Hong Zhou, Yunhui Guo, Yongchen Wang, Bo Lei, Yanying Miao, Zhongfeng Wang
    2026, 21 (4):  1628-1640.  doi: 10.4103/NRR.NRR-D-24-00461
    Abstract ( 16 )   PDF (7834KB) ( 2 )   Save
    Downregulation of the inwardly rectifying potassium channel Kir4.1 is a key step for inducing retinal Müller cell activation and interaction with other glial cells, which is involved in retinal ganglion cell apoptosis in glaucoma. Modulation of Kir4.1 expression in Müller cells may therefore be a potential strategy for attenuating retinal ganglion cell damage in glaucoma. In this study, we identified seven predicted phosphorylation sites in Kir4.1 and constructed lentiviral expression systems expressing Kir4.1 mutated at each site to prevent phosphorylation. Following this, we treated Müller glial cells in vitro and in vivo with the mGluR I agonist DHPG to induce Kir4.1 or Kir4.1 Tyr9 Asp overexpression. We found that both Kir4.1 and Kir4.1 Tyr9 Asp overexpression inhibited activation of Müller glial cells. Subsequently, we established a rat model of chronic ocular hypertension by injecting microbeads into the anterior chamber and overexpressed Kir4.1 or Kir4.1 Tyr9 Asp in the eye, and observed similar results in Müller cells in vivo as those seen in vitro. Both Kir4.1 and Kir4.1 Tyr9 Asp overexpression inhibited Müller cell activation, regulated the balance of Bax/Bcl-2, and reduced the mRNA and protein levels of pro-inflammatory factors, including interleukin-1β and tumor necrosis factor-α. Furthermore, we investigated the regulatory effects of Kir4.1 and Kir4.1 Tyr9 Asp overexpression on the release of pro-inflammatory factors in a co-culture system of Müller glial cells and microglia. In this co-culture system, we observed elevated adenosine triphosphate concentrations in activated Müller cells, increased levels of translocator protein (a marker of microglial activation), and elevated interleukin1β mRNA and protein levels in microglia induced by activated Müller cells. These changes could be reversed by Kir4.1 and Kir4.1 Tyr9 Asp overexpression in Müller cells. Kir4.1 overexpression, but not Kir4.1 Tyr9 Asp overexpression, reduced the number of proliferative and migratory microglia induced by activated Müller cells. Collectively, these results suggest that the tyrosine residue at position nine in Kir4.1 may serve as a functional modulation site in the retina in an experimental model of glaucoma. Kir4.1 and Kir4.1 Tyr9 Asp overexpression attenuated Müller cell activation, reduced ATP/P2X receptor–mediated interactions between glial cells, inhibited microglial activation, and decreased the synthesis and release of pro-inflammatory factors, consequently ameliorating retinal ganglion cell apoptosis in glaucoma.
    Related Articles | Metrics
    The Cullin3–Ring E3 ubiquitin ligase complex and USP14 regulate spastin-mediated microtubule severing and promotion of neurite outgrowth
    Zhenbin Cai, Hui Wu, Tao Jiang, Ao Ma, Zhichao Meng, Jiehao Zhu, Hongsheng Lin, Yaozhong Liang, Guowei Zhang, Minghui Tan
    2026, 21 (4):  1641-1651.  doi: 10.4103/NRR.NRR-D-25-00037
    Abstract ( 15 )   PDF (8359KB) ( 2 )   Save
    Post-translational modification of spastin enables precise spatiotemporal control of its microtubule severing activity. However, the detailed mechanism by which spastin turnover is regulated in the context of neurite outgrowth remains unknown. Here, we found that spastin interacted with ubiquitin and was significantly degraded by K48-mediated poly-ubiquitination. Cullin3 facilitated spastin degradation and ubiquitination. RING-box protein 1, but not RING-box protein 2, acted synergistically with Cullin3 protein to regulate spastin degradation. Overexpression of Culin3 or BRX1 markedly suppressed spastin expression, and inhibited spastin-mediated microtubule severing and promotion of neurite outgrowth. Moreover, USP14 interacted directly with spastin to mediate its deubiquitination. USP14 overexpression significantly increased spastin expression and suppressed its ubiquitination and degradation. Although co-expression of spastin and USP14 did not enhance microtubule severing, it did increase neurite length in hippocampal neurons. Taken together, these findings elucidate the intricate regulatory mechanisms of spastin turnover, highlighting the roles of the Cullin-3–Ring E3 ubiquitin ligase complex and USP14 in orchestrating its ubiquitination and degradation. The dynamic interplay between these factors governs spastin stability and function, ultimately influencing microtubule dynamics and neuronal morphology. These insights shed light on potential therapeutic targets for neurodegenerative disorders associated with spastin defects.
    Related Articles | Metrics
    Voltage-dependent anion channel 1 oligomerization regulates PANoptosis in retinal ischemia–reperfusion injury
    Hao Wan, Xiaoxia Ban, Ye He, Yandi Yang, Ximin Hu, Lei Shang, Xinxing Wan, Qi Zhang, Kun Xiong
    2026, 21 (4):  1652-1664.  doi: 10.4103/NRR.NRR-D-24-00674
    Abstract ( 35 )   PDF (18918KB) ( 3 )   Save
    Ischemia–reperfusion injury is a common pathophysiological mechanism in retinal degeneration. PANoptosis is a newly defined integral form of regulated cell death that combines the key features of pyroptosis, apoptosis, and necroptosis. Oligomerization of mitochondrial voltage-dependent anion channel 1 is an important pathological event in regulating cell death in retinal ischemia–reperfusion injury. However, its role in PANoptosis remains largely unknown. In this study, we demonstrated that voltage-dependent anion channel 1 oligomerization-mediated mitochondrial dysfunction was associated with PANoptosis in retinal ischemia–reperfusion injury. Inhibition of voltage-dependent anion channel 1 oligomerization suppressed mitochondrial dysfunction and PANoptosis in retinal cells subjected to ischemia–reperfusion injury. Mechanistically, mitochondria-derived reactive oxygen species played a central role in the voltagedependent anion channel 1-mediated regulation of PANoptosis by promoting PANoptosome assembly. Moreover, inhibiting voltage-dependent anion channel 1 oligomerization protected against PANoptosis in the retinas of rats subjected to ischemia–reperfusion injury. Overall, our findings reveal the critical role of voltage-dependent anion channel 1 oligomerization in regulating PANoptosis in retinal ischemia–reperfusion injury, highlighting voltage-dependent anion channel 1 as a promising therapeutic target.
    Related Articles | Metrics