Loading...

Table of Content

    15 December 2025, Volume 20 Issue 12 Previous Issue   
    For Selected: Toggle Thumbnails
    Calcium bridges built by mitochondria-associated endoplasmic reticulum membranes: potential targets for neural repair in neurological diseases
    Yichen Peng, Li Zhou, Yaju Jin, Danli Wu, Na Chen, Chengcai Zhang, Hongpeng Liu, Chunlan Li , Rong Ning, Xichen Yang, Qiuyue Mao, Jiaxin Liu, Pengyue Zhang
    2025, 20 (12):  3349-3369.  doi: NRR.NRR-D-24-00630
    Abstract ( 32 )   PDF (3023KB) ( 62 )   Save
    The exchange of information and materials between organelles plays a crucial role in regulating cellular physiological functions and metabolic levels. Mitochondria-associated endoplasmic reticulum membranes serve as physical contact channels between the endoplasmic reticulum membrane and the mitochondrial outer membrane, formed by various proteins and protein complexes. This microstructural domain mediates several specialized functions, including calcium (Ca2+) signaling, autophagy, mitochondrial morphology, oxidative stress response, and apoptosis. Notably, the dysregulation of Ca2+ signaling mediated by mitochondria-associated endoplasmic reticulum membranes is a critical factor in the pathogenesis of neurological diseases. Certain proteins or protein complexes within these membranes directly or indirectly regulate the distance between the endoplasmic reticulum and mitochondria, as well as the transduction of Ca2+ signaling. Conversely, Ca2+ signaling mediated by mitochondria-associated endoplasmic reticulum membranes influences other mitochondria-associated endoplasmic reticulum membraneassociated functions. These functions can vary significantly across different neurological diseases—such as ischemic stroke, traumatic brain injury, Alzheimer’s disease, Parkinson’s disease, amyotrophic lateral sclerosis, and Huntington’s disease—and their respective stages of progression. Targeted modulation of these disease-related pathways and functional proteins can enhance neurological function and promote the regeneration and repair of damaged neurons. Therefore, mitochondria-associated endoplasmic reticulum membranes-mediated Ca2+ signaling plays a pivotal role in the pathological progression of neurological diseases and represents a significant potential therapeutic target. This review focuses on the effects of protein complexes in mitochondria-associated endoplasmic reticulum membranes and the distinct roles of mitochondria-associated endoplasmic reticulum membranes-mediated Ca2+ signaling in neurological diseases, specifically highlighting the early protective effects and neuronal damage that can result from prolonged mitochondrial Ca2+ overload or deficiency. This article provides a comprehensive analysis of the various mechanisms of Ca2+ signaling mediated by mitochondria-associated endoplasmic reticulum membranes in neurological diseases, contributing to the exploration of potential therapeutic targets for promoting neuroprotection and nerve repair.
    Related Articles | Metrics
    Mitochondrial transplantation: a promising strategy for the treatment of retinal degenerative diseases
    Jing Chi, Bin Fan, Yulin Li, Qing Jiao, Guang-Yu Li
    2025, 20 (12):  3370-3387.  doi: 10.4103/NRR.NRR-D-24-00851
    Abstract ( 75 )   PDF (4216KB) ( 96 )   Save
    The retina, a crucial neural tissue, is responsible for transforming light signals into visual information, a process that necessitates a significant amount of energy. Mitochondria, the primary powerhouses of the cell, play an integral role in retinal physiology by fulfilling the high-energy requirements of photoreceptors and secondary neurons through oxidative phosphorylation. In a healthy state, mitochondria ensure proper visual function by facilitating efficient conversion and transduction of visual signals. However, in retinal degenerative diseases, mitochondrial dysfunction significantly contributes to disease progression, involving a decline in membrane potential, the occurrence of DNA mutations, increased oxidative stress, and imbalances in quality-control mechanisms. These abnormalities lead to an inadequate energy supply, the exacerbation of oxidative damage, and the activation of cell death pathways, ultimately resulting in neuronal injury and dysfunction in the retina. Mitochondrial transplantation has emerged as a promising strategy for addressing these challenges. This procedure aims to restore metabolic activity and function in compromised cells through the introduction of healthy mitochondria, thereby enhancing the cellular energy production capacity and offering new strategies for the treatment of retinal degenerative diseases. Although mitochondrial transplantation presents operational and safety challenges that require further investigation, it has demonstrated potential for reviving the vitality of retinal neurons. This review offers a comprehensive examination of the principles and techniques underlying mitochondrial transplantation and its prospects for application in retinal degenerative diseases, while also delving into the associated technical and safety challenges, thereby providing references and insights for future research and treatment.
    Related Articles | Metrics
    Optimizing non-invasive vagus nerve stimulation for treatment in stroke
    Sheharyar S. Baig, Samantha Dorney, Mudasar Aziz, Simon M. Bell, Ali N. Ali, Li Su, Jessica N. Redgrave, Arshad Majid
    2025, 20 (12):  3388-3399.  doi: 10.4103/NRR.NRR-D-24-00945
    Abstract ( 78 )   PDF (2021KB) ( 50 )   Save
    Stroke remains a leading cause of long-term disability worldwide. There is an unmet need for neuromodulatory therapies that can mitigate against neurovascular injury and potentially promote neurological recovery. Transcutaneous vagus nerve stimulation has been demonstrated to show potential therapeutic effects in both acute and chronic stroke. However, previously published research has only investigated a narrow range of stimulation settings and indications. In this review, we detail the ongoing studies of transcutaneous vagus nerve stimulation in stroke through systematic searches of registered clinical trials. We summarize the upcoming clinical trials of transcutaneous vagus nerve stimulation in stroke, highlighting their indications, parameter settings, scope, and limitations. We further explore the challenges and barriers associated with the implementation of transcutaneous vagus nerve stimulation in acute stroke and stroke rehabilitation, focusing on critical aspects such as stimulation settings, target groups, biomarkers, and integration with rehabilitation interventions.
    Related Articles | Metrics
    Bidirectional causality of physical exercise in retinal neuroprotection
    Stephen K. Agadagba, Suk-yu Yau, Ying Liang, Kristine Dalton, Benjamin Thompson
    2025, 20 (12):  3400-3415.  doi: 10.4103/NRR.NRR-D-24-00942
    Abstract ( 56 )   PDF (1813KB) ( 71 )   Save
    Physical exercise is recognized as an effective intervention to improve mood, physical performance, and general well-being. It achieves these benefits through cellular and molecular mechanisms that promote the release of neuroprotective factors. Interestingly, reduced levels of physical exercise have been implicated in several central nervous system diseases, including ocular disorders. Emerging evidence has suggested that physical exercise levels are significantly lower in individuals with ocular diseases such as glaucoma, age-related macular degeneration, retinitis pigmentosa, and diabetic retinopathy. Physical exercise may have a neuroprotective effect on the retina. Therefore, the association between reduced physical exercise and ocular diseases may involve a bidirectional causal relationship whereby visual impairment leads to reduced physical exercise and decreased exercise exacerbates the development of ocular disease. In this review, we summarize the evidence linking physical exercise to eye disease and identify potential mediators of physical exercise-induced retinal neuroprotection. Finally, we discuss future directions for preclinical and clinical research in exercise and eye health.
    Related Articles | Metrics
    Gut–brain axis and environmental factors in Parkinson’s disease: bidirectional link between disease onset and progression
    Soo Jung Park, Kyung Won Kim, Eun Jeong Lee
    2025, 20 (12):  3416-3429.  doi: 10.4103/NRR.NRR-D-24-00994
    Abstract ( 46 )   PDF (3025KB) ( 57 )   Save
    Parkinson’s disease has long been considered a disorder that primarily affects the brain, as it is defined by the dopaminergic neurodegeneration in the substantia nigra and the brain accumulation of Lewy bodies containing α-synuclein protein. In recent decades, however, accumulating research has revealed that Parkinson’s disease also involves the gut and uncovered an intimate and important bidirectional link between the brain and the gut, called the “gut–brain axis.” Numerous clinical studies demonstrate that gut dysfunction frequently precedes motor symptoms in Parkinson’s disease patients, with findings including impaired intestinal permeability, heightened inflammation, and distinct gut microbiome profiles and metabolites. Furthermore, α-synuclein deposition has been consistently observed in the gut of Parkinson’s disease patients, suggesting a potential role in disease initiation. Importantly, individuals with vagotomy have a reduced Parkinson’s disease risk. From these observations, researchers have hypothesized that α-synuclein accumulation may initiate in the gut and subsequently propagate to the central dopaminergic neurons through the gut–brain axis, leading to Parkinson’s disease. This review comprehensively examines the gut’s involvement in Parkinson’s disease, focusing on the concept of a gut-origin for the disease. We also examine the interplay between altered gut-related factors and the accumulation of pathological α-synuclein in the gut of Parkinson’s disease patients. Given the accessibility of the gut to both dietary and pharmacological interventions, targeting gut-localized α-synuclein represents a promising avenue for developing effective Parkinson’s disease therapies.
    Related Articles | Metrics
    Functional abnormalities of the glymphatic system in cognitive disorders
    Wuyue Shentu, Qi Kong, Yier Zhang, Wenyao Li, Qiulu Chen, Sicheng Yan, Junjun Wang, Qilun Lai, Qi Xu, Song Qiao
    2025, 20 (12):  3430-3447.  doi: 10.4103/NRR.NRR-D-24-01049
    Abstract ( 80 )   PDF (2084KB) ( 61 )   Save
    Various pathological mechanisms represent distinct therapeutic targets for cognitive disorders, but a balance between clearance and production is essential for maintaining the stability of the brain’s internal environment. Thus, the glymphatic system may represent a common pathway by which to address cognitive disorders. Using the established model of the glymphatic system as our foundation, this review disentangles and analyzes the components of its clearance mechanism, including the initial inflow of cerebrospinal fluid, the mixing of cerebrospinal fluid with interstitial fluid, and the outflow of the mixed fluid and the clearance. Each section summarizes evidence from experimental animal models and human studies, highlighting the normal physiological properties of key structures alongside their pathological manifestations in cognitive disorders. The same pathologic manifestations of different cognitive disorders appearing in the glymphatic system and the same upstream influences are main points of interest of this review. We conclude this article by discussing new findings and outlining the limitations identified in current research progress.
    Related Articles | Metrics
    Generation, interrogation, and future applications of microglia-containing brain organoids
    Julia Di Stefano, Federica Di Marco, Ilaria Cicalini, Una FitzGerald, Damiana Pieragostino, Marleen Verhoye, Peter Ponsaerts, Elise Van Breedam
    2025, 20 (12):  3448-3460.  doi: 10.4103/NRR.NRR-D-24-00921
    Abstract ( 55 )   PDF (6442KB) ( 49 )   Save
    Brain organoids encompass a large collection of in vitro stem cell–derived 3D culture systems that aim to recapitulate multiple aspects of in vivo brain development and function. First, this review provides a brief introduction to the current state-of-the-art for neuroectoderm brain organoid development, emphasizing their biggest advantages in comparison with classical two-dimensional cell cultures and animal models. However, despite their usefulness for developmental studies, a major limitation for most brain organoid models is the absence of contributing cell types from endodermal and mesodermal origin. As such, current research is highly investing towards the incorporation of a functional vasculature and the microglial immune component. In this review, we will specifically focus on the development of immune-competent brain organoids. By summarizing the different approaches applied to incorporate microglia, it is highlighted that immune-competent brain organoids are not only important for studying neuronal network formation, but also offer a clear future as a new tool to study inflammatory responses in vitro in 3D in a brainlike environment. Therefore, our main focus here is to provide a comprehensive overview of assays to measure microglial phenotype and function within brain organoids, with an outlook on how these findings could better understand neuronal network development or restoration, as well as the influence of physical stress on microglia-containing brain organoids. Finally, we would like to stress that even though the development of immunecompetent brain organoids has largely evolved over the past decade, their full potential as a pre-clinical tool to study novel therapeutic approaches to halt or reduce inflammationmediated neurodegeneration still needs to be explored and validated. 
    Related Articles | Metrics
    Role of astrocytes and microglia in hepatic encephalopathy associated with advanced chronic liver disease: lessons from animal studies
    Wouter Claeys, Anja Geerts, Lien Van Hoecke, Christophe Van Steenkiste, Roosmarijn E. Vandenbroucke
    2025, 20 (12):  3461-3475.  doi: 10.4103/NRR.NRR-D-24-00600
    Abstract ( 30 )   PDF (4023KB) ( 48 )   Save
    Hepatic encephalopathy, defined as neuropsychiatric dysfunction secondary to liver disease, is a frequent decompensating event in cirrhosis. Its clinical impact is highlighted by a notable increase in patient mortality rates and a concomitant reduction in overall quality of life. Systemically, liver disease, liver function failure, portosystemic shunting, and associated multi-organ dysfunction result in the increase of disease-causing neurotoxins in the circulation, which impairs cerebral homeostasis. Key circulating neurotoxins are ammonia and inflammatory mediators. In the brain, pathophysiology is less well understood, but is thought to be driven by glial cell dysfunction. Astrocytes are the only brain resident cells that have ammonia-metabolizing machinery and are therefore putatively most susceptible to ammonia elevation. Based on a large body of mostly in vitro evidence, ammonia-induced cellular and molecular disturbances include astrocyte swelling and oxidative stress. Microglia, the brain resident macrophages, have been linked to the translation of systemic inflammation to the brain microenvironment. Recent evidence from animal studies has provided novel insights into old and new downstream effects of astrocyte and microglial dysfunction such as toxin clearance disruption and myeloid cell attraction to the central nervous system parenchyma. Furthermore, state of the art research increasingly implicates neuronal dysfunction and possibly even irreversible neuronal cell death. Cell-type specific investigation in animal models highlights the need for critical revision of the contribution of astrocytes and microglia to wellestablished and novel cellular and molecular alterations in hepatic encephalopathy. In this review, we therefore give a current and comprehensive overview of causes, features, and consequences of astrocyte and microglial dysfunction in hepatic encephalopathy, including areas of interest for future investigation.
    Related Articles | Metrics
    Biomaterial-based strategies: a new era in spinal cord injury treatment
    Shihong Zhu, Sijun Diao, Xiaoyin Liu, Zhujun Zhang, Fujun Liu, Wei Chen, Xiyue Lu, Huiyang Luo, Xu Cheng, Qiang Liao, Zhongyu Li, Jing Chen
    2025, 20 (12):  3476-3500.  doi: 10.4103/NRR.NRR-D-24-00844
    Abstract ( 185 )   PDF (14777KB) ( 36 )   Save
    Enhancing neurological recovery and improving the prognosis of spinal cord injury have gained research attention recently. Spinal cord injury is associated with a complex molecular and cellular microenvironment. This complexity has prompted researchers to elucidate the underlying pathophysiological mechanisms and changes and to identify effective treatment strategies. Traditional approaches for spinal cord injury repair include surgery, oral or intravenous medications, and administration of neurotrophic factors; however, the efficacy of these approaches remains inconclusive, and serious adverse reactions continue to be a concern. With advancements in tissue engineering and regenerative medicine, emerging strategies for spinal cord injury repair now involve nanoparticle-based nanodelivery systems, scaffolds, and functional recovery techniques that incorporate biomaterials, bioengineering, stem cell, and growth factors as well as three-dimensional bioprinting. Ideal biomaterial scaffolds should not only provide structural support for neuron migration, adhesion, proliferation, and differentiation but also mimic the mechanical properties of natural spinal cord tissue. Additionally, these scaffolds should facilitate axon growth and neurogenesis by offering adjustable topography and a range of physical and biochemical cues. The three-dimensionally interconnected porous structure and appropriate physicochemical properties enabled by three-dimensional biomimetic printing technology can maximize the potential of biomaterials used for treating spinal cord injury. Therefore, correct selection and application of scaffolds, coupled with successful clinical translation, represent promising clinical objectives to enhance the treatment efficacy for and prognosis of spinal cord injury. This review elucidates the key mechanisms underlying the occurrence of spinal cord injury and regeneration post-injury, including neuroinflammation, oxidative stress, axon regeneration, and angiogenesis. This review also briefly discusses the critical role of nanodelivery systems used for repair and regeneration of injured spinal cord, highlighting the influence of nanoparticles and the factors that affect delivery efficiency. Finally, this review highlights tissue engineering strategies and the application of biomaterial scaffolds for the treatment of spinal cord injury. It discusses various types of scaffolds, their integrations with stem cells or growth factors, and approaches for optimization of scaffold design.
    Related Articles | Metrics
    Potential of ultrasound stimulation and sonogenetics in vision restoration: a narrative review
    Jie Ji, Chen Gong, Gengxi Lu, Junhang Zhang, Baoqiang Liu, Xunan Liu, Junhao Lin, Patrick Wang, Biju B. Thomas, Mark S. Humayun, Qifa Zhou
    2025, 20 (12):  3501-3516.  doi: 10.4103/NRR.NRR-D-24-00841
    Abstract ( 78 )   PDF (3437KB) ( 142 )   Save
    Vision restoration presents a considerable challenge in the realm of regenerative medicine, while recent progress in ultrasound stimulation has displayed potential as a non-invasive therapeutic approach. This narrative review offers a comprehensive overview of current research on ultrasound-stimulated neuromodulation, emphasizing its potential as a treatment modality for various nerve injuries. By examining of the efficacy of different types of ultrasound stimulation in modulating peripheral and optic nerves, we can delve into their underlying molecular mechanisms. Furthermore, the review underscores the potential of sonogenetics in vision restoration, which involves leveraging pharmacological and genetic manipulations to inhibit or enhance the expression of related mechanosensitive channels, thereby modulating the strength of the ultrasound response. We also address how methods such as viral transcription can be utilized to render specific neurons or organs highly responsive to ultrasound, leading to significantly improved therapeutic outcomes.
    Related Articles | Metrics
    Neuronal autosis: the selfdestructive side of autophagy involved in hypoxic-ischemic neuronal death
    Vanessa Ginet, Pauline Depierre, Julien Puyal
    2025, 20 (12):  3517-3518.  doi: 10.4103/NRR.NRR-D-24-00831
    Abstract ( 45 )   PDF (4516KB) ( 30 )   Save
    The challenge of protecting the brain resides in the unique characteristics of neurons, as they are postmitotic, long-lived, excitable, and polarized cells with long and fragile axons and dendrites. The complexity of the multiple potential cell death pathways further complicates this issue. In addition, the immature brain is prone to a “cell death continuum,” which involves intricate molecular interconnections between cell death processes. This makes finding safe and effective neuroprotective strategies to prevent damage to the developing brain a significant challenge in neonatology. The only approved treatment for term newborns with hypoxic-ischemic encephalopathy (HIE) is therapeutic hypothermia. However, access to this treatment and its effectiveness is limited to a few cases. Research is focused on developing new neuroprotective agents that can be combined with hypothermia to improve its therapeutic window and outcome.
    Related Articles | Metrics
    Autophagy regulation and function in axon regeneration: new insights from Caenorhabditis elegans
    Su-Hyuk Ko, Lizhen Chen
    2025, 20 (12):  3519-3520.  doi: 10.4103/NRR.NRR-D-24-00956
    Abstract ( 32 )   PDF (1724KB) ( 17 )   Save
    Autophagy plays a crucial role in axon regeneration by maintaining cellular homeostasis and promoting the clearance of damaged organelles and proteins, which is essential for the growth and repair of axons. In response to axonal injury, autophagy is upregulated to facilitate the removal of cellular debris and support the recycling of essential components needed for regeneration. This process, known as bulk autophagy, not only helps mitigate damage but also provides the necessary building blocks for axonal repair. Moreover, our previous work suggests that in addition to bulk autophagy, selective autophagy can specifically target and degrade inhibitors of axon regeneration, thereby promoting axon regrowth (Ko et al., 2020). Understanding the regulation and function of autophagy in axon regeneration could provide valuable insights for developing therapeutic strategies to target autophagy in neurodegenerative diseases and nerve injuries.
    Related Articles | Metrics
    Extracellular vesicles: multiple signaling capabilities and translation into promising therapeutic targets to promote neuronal plasticity
    Dirk M. Hermann , Bernd Giebel
    2025, 20 (12):  3521-3522.  doi: 10.4103/NRR.NRR-D-24-00980
    Abstract ( 32 )   PDF (457KB) ( 27 )   Save
    Extracellular vesicles (EVs) are cell-derived, lipid membrane-enclosed vesicles carrying a broad spectrum of biologically active molecules (including proteins, RNAs, and bioactive lipids) which play important roles in intercellular communication. EVs crucially control neuronal energy metabolism under physiological conditions, constrain oxidative stress and brain inflammatory responses, and promote neuronal survival and plasticity upon brain damage. Originating from the right cells, e.g., mesenchymal stromal cells (MSCs), EVs can exhibit striking neurological recovery-promoting activities in various brain disease models (Hermann et al., 2024). In rodent middle cerebral artery occlusion (MCAO) models, for example, intravenously administered MSC-derived EVs enhanced motor coordination recovery similar to parental MSCs by mechanisms involving long-term neuroprotection, neurogenesis, axonal sprouting, remyelination, and synaptic plasticity (Xin et al., 2013; Doeppner et al., 2015). In contrast to pharmacological compounds that target specific signaling pathways, EVs depending on their cellular origin exhibit multiple signaling abilities, enabling them to regulate several disease processes simultaneously in a clinically relevant way (Hermann et al., 2024). In the long term, EVs are expected to surpass and replace many current pharmaceuticals, as their multimodal mechanism of action can synergistically and contextually modify disease outcomes more effectively. The successful clinical translation will decisively depend on the selection of the right targets.
    Related Articles | Metrics
    Aptamers as new promising entities for therapeutics: our experience from the neurological diseases
    Macarena Hernández-Jiménez, Fernando de Castro
    2025, 20 (12):  3523-3524.  doi: 10.4103/NRR.NRR-D-24-00667
    Abstract ( 47 )   PDF (546KB) ( 17 )   Save
    Based on data from Global Burden of Disease, Injuries, and Risk Factors (2024), disorders of the central nervous system (CNS) are an important cause of death or long-term disability, representing the top-ranked contributor to global disability adjusted life years. This group of pathological conditions includes congenital and neurodevelopmental disorders, cerebrovascular diseases, and neurodegenerative diseases, and induce the disruption of brain growth, brain and/or spinal cord damage, and cognitive, sensory, or motor function impairment. Surprisingly, despite the huge impact of these diseases on the lives of patients, there is a lack of available therapeutic options and the investigation in the field represents real interest in the scientific community nowadays.
    Related Articles | Metrics
    Is it possible to maintain a plaquefree healthy brain by treadmill?
    Swarupa Pahan, Kalipada Pahan
    2025, 20 (12):  3525-3526.  doi: 10.4103/NRR.NRR-D-24-00936
    Abstract ( 45 )   PDF (634KB) ( 13 )   Save
    It is believed that overproduction and deposition of amyloid-β (Aβ) plaques in the hippocampus and cortex region of the brain cause neuronal dysfunction leading to cognitive impairments in Alzheimer’s disease (AD). Until now, there has been no effective treatment to reduce plaque load from the brain. Recent studies have shown that regular treadmill exercise lowers plaques from the brain of a mouse model of AD. Out of three proteolytic enzymes or secretases, while α-secretase activates the non-amyloidogenic pathway of amyloid precursor protein (APP) cleavage to inhibit Aβ plaque production, β- and γ-secretases indulge the APP cleavage in the amyloidogenic pathway for the formation of Aβ plaques. Accordingly, it is nice to see that treadmill run increases α-secretase and decreases β- and γ-secretases in AD mice. Interestingly, in normalaged mice as well, a treadmill run increases α-secretase and decreases β- and γ-secretases, indicating that in normal subjects as well, a treadmill run may keep the formation of Aβ plaques at bay. The prototype role of peroxisome proliferator-activated receptor α (PPARα), a nuclear hormone receptor, is to regulate energy homeostasis via catabolism of fatty acids. However, it has been shown that the success of treadmill run depends on PPARα as the treadmill remains ineffective in increasing α-secretase and reducing plaque burden in AD mice lacking PPARα. Here, we discuss about this drug-free safe approach and associated PPARα-dependent mechanisms for the inhibition of plaque pathogenesis.
    Related Articles | Metrics
    LPAR3: a shared target for neurodegenerative diseases?
    Susan Acton , Laurent Chesnel
    2025, 20 (12):  3527-3528.  doi: 10.4103/NRR.NRR-D-24-01024
    Abstract ( 37 )   PDF (1079KB) ( 10 )   Save
    Neurodegenerative diseases are often studied in isolation due to the vast differences in their underlying causes, the diversity in neuron types affected, and the variations in their clinical symptoms. However, there are common elements among these diseases, namely neuroinflammation, mitochondrial pathology, and an association with the gut–brain–immune axis that together suggest that these diseases are more similar to each other than they first seem (Zhang et al., 2023). Despite this, efforts have generally focused on developing therapies aimed at the source of the diseases while less focus has been on developing therapies that might impact a mutual downstream pathway. Reasons for this may include (1) the bias that each disease is unique and must be approached by targeting the initiating cause(s), (2) our lack of understanding of how all the common elements fit together among these diseases, and (3) the uncertainty that a shared element might be a successful target. Indeed, if it is challenging to target even one individual cause of one neurodegenerative disease, how could we expect to target a member of a downstream pathway for an effective treatment of multiple diseases? The discovery of an orally delivered bacteria that is broadly efficacious in neurodegenerative disease models through the production of an agonist of the lysophosphatidic acid receptor 3 (LPAR3), a G-protein coupled receptor involved in mitochondrial health, has presented a door to new possibilities (Acton et al., 2024).
    Related Articles | Metrics
    Alzheimer’s disease and the immune system: the emerging role of TEMRA cells
    Edric D. Winford, Adam D. Bachstetter
    2025, 20 (12):  3529-3530.  doi: 10.4103/NRR.NRR-D-24-00793
    Abstract ( 47 )   PDF (774KB) ( 27 )   Save
    Alzheimer’s disease (AD) is the most common form of dementia, affecting millions worldwide. It is characterized by progressive cognitive decline and changes in behavior and personality, attributed to neuropathological changes, such as amyloid-beta (Aβ) plaques and neurofibrillary tangles composed of hyperphosphorylated tau protein. While microglia, the resident immune cells in the brain, are well established as contributors in AD disease progression, recently, T cells in the periphery and the brain have also been highlighted as being important in the progression of AD, with CD8+ T cells reducing amyloid-β pathology and neurodegeneration by interacting with microglia (Su et al., 2023), while increasing tau-mediated neurodegeneration (Chen et al., 2023), showing the possibility of T cells to effect AD neuropathological changes, at least in animal models with specific and isolated AD-related pathologies.
    Related Articles | Metrics
    Alzheimer’s disease treatment landscape: current therapies and emerging mechanism-targeted approaches
    Eleen Yang, Khaled S. Abd-Elrahman
    2025, 20 (12):  3531-3532.  doi: 10.4103/NRR.NRR-D-24-00934
    Abstract ( 36 )   PDF (837KB) ( 30 )   Save
    Alzheimer ’s disease (AD) is an age-related neurodegenerative disease that involves a progressive decline in memory, thinking, and learning. AD has been studied rigorously for more than 50 years yet there is a marked lack of progress in pharmacological treatment. The challenges to developing AD therapeutics are rooted in the disease being ill-defined in terms of diagnosis, progression, and etiology. The neuropathological hallmarks of AD are extracellular amyloid-β (Aβ) plaques and intracellular neurofibrillary tangles of hyperphosphorylated tau. Generally, Aβ plaques disrupt neuronal connectivity, while neurofibrillary tangles interfere with intracellular function (Zhang et al., 2023). Therefore, developing innovative strategies to slow the formation or enhance the clearance of these toxic proteins has shown promise in disease-modifying approaches for AD.
    Related Articles | Metrics
    Age-dependent alpha-synuclein aggregation and Lewy body formation in Parkinson’s disease
    Chun Chau Sung, Kenny K. K. Chung
    2025, 20 (12):  3533-3534.  doi: 10.4103/NRR.NRR-D-24-00772
    Abstract ( 32 )   PDF (3351KB) ( 8 )   Save
    Parkinson’s disease (PD) is a common degenerative disorder that is becoming increasingly prevalent because of the global aging population. The exact cause of the disorder is unknown; however, recent studies have suggested that multiple factors may contribute to its pathogenesis. PD is characterized by a movement disorder that primarily affects motor control; pathologically, the disease is marked by the presence of Lewy bodies (LBs) in the brain. LBs were first described by Friederich Lewy as intraneuronal protein inclusion bodies observed in PD patients. The mechanisms of LB formation have been the focus of much PD research, as understanding the mechanisms of LB formation could provide potential treatments for PD.
    Related Articles | Metrics
    Cerebral cortex contributions to Parkinson’s disease tremor: what exactly generates the tremor?
    Manuel Bange , Hao Ding, Muthuraman Muthuraman
    2025, 20 (12):  3535-3536.  doi: 10.4103/NRR.NRR-D-24-00853
    Abstract ( 31 )   PDF (882KB) ( 7 )   Save
    Pathological tremor is one of the cardinal symptoms in Parkinson’s disease (PD). Tremor is comprised of involuntary, rhythmic, and oscillating movements that can vary according to the circumstances under which they occur, the body parts that are involved, and the frequency at which they present. For example, tremors can be mild to severe, are stress sensitive, and can affect arms, legs, or the head (Dirkx and Bologna, 2022). Furthermore, it can appear at rest (rest tremor), while holding a posture (postural tremor), or even during active movement (kinetic tremor). Among these variants, rest tremor is the most common manifestation, usually expressing itself as an asymmetric, pill-rolling movement of the hands at frequencies from 4 to 6 Hz which is inhibited during voluntary movement. Paradoxically, people with rest tremors may additionally experience a tremor that re-appears after a brief delay (on average 10 seconds) when maintaining a stable postural position, a phenomenon known as reemergent tremor (Jankovic et al., 1999). Given that the phenomenological and electrophysiological characteristics of rest and re-emergent tremor are similar and patients with re-emergent tremor do not differ clinically from patients with isolated rest tremor, re-emergent tremor was initially suggested to be a clinical variant of rest tremor and assumed to share the same central tremor circuit (Jankovic et al., 1999).
    Related Articles | Metrics
    Shedding light on the retina to see healthy and pathological aging
    Marília Inês Móvio, Maria Camila Almeida, Sergio T. Ferreira, Alexandre Hiroaki Kihara
    2025, 20 (12):  3537-3538.  doi: 10.4103/NRR.NRR-D-24-00809
    Abstract ( 25 )   PDF (750KB) ( 16 )   Save
    With the advances in medicine and technology, life expectancy has increased significantly in recent decades. While this is positive news, it also brings major challenges to public health systems. As people live longer, the number of cases of agerelated disorders, including neurodegenerative diseases such as Alzheimer’s disease (AD) and Parkinson’s disease (PD), has also grown. These debilitating conditions primarily affect the elderly and can have a devastating impact on the quality of life of patients and their families. Considering emerging treatments, one of the most challenging aspects of neurodegenerative diseases is their early diagnosis and continuous monitoring.
    Related Articles | Metrics
    A macro-transection model of brain trauma for neuromaterial testing with functional electrophysiological readouts
    Jessica Wiseman, Raja Haseeb Basit, Akihiro Suto, Sagnik Middya, Bushra Kabiri, Michael Evans, Vinoj George, Christopher Adams, George Malliaras, Divya Maitreyi Chari
    2025, 20 (12):  3539-3552.  doi: 10.4103/NRR.NRR-D-24-00422
    Abstract ( 73 )   PDF (14220KB) ( 17 )   Save
    Functional recovery in penetrating neurological injury is hampered by a lack of clinical regenerative therapies. Biomaterial therapies show promise as medical materials for neural repair through immunomodulation, structural support, and delivery of therapeutic biomolecules. However, a lack of facile and pathology-mimetic models for therapeutic testing is a bottleneck in neural tissue engineering research. We have deployed a two-dimensional, high-density multicellular cortical brain sheet to develop a facile model of injury (macrotransection/ scratch wound) in vitro. The model encompasses the major neural cell types involved in pathological responses post-injury. Critically, we observed hallmark pathological responses in injury foci including cell scarring, immune cell infiltration, precursor cell migration, and shortrange axonal sprouting. Delivering test magnetic particles to evaluate the potential of the model for biomaterial screening shows a high uptake of introduced magnetic particles by injury-activated immune cells, mimicking in vivo findings. Finally, we proved it is feasible to create reproducible traumatic injuries in the brain sheet (in multielectrode array devices in situ) characterized by focal loss of electrical spiking in injury sites, offering the potential for longer term, electrophysiology plus histology assays. To our knowledge, this is the first in vitro simulation of transecting injury in a two-dimensional multicellular cortical brain cell sheet, that allows for combined histological and electrophysiological readouts of damage/repair. The patho-mimicry and adaptability of this simplified model of brain injury could benefit the testing of biomaterial therapeutics in regenerative neurology, with the option for functional electrophysiological readouts.
    Related Articles | Metrics
    Plasma circulating cell–free DNA integrity and relative telomere length as diagnostic biomarkers for Parkinson’s disease and multiple system atrophy: a cross-sectional study
    Chao Ying, Chao Han, Yuan Li, Mingkai Zhang, Shuying Xiao, Lifang Zhao, Hui Zhang, Qian Yu, Jing An, Wei Mao, Yanning Cai
    2025, 20 (12):  3553-3563.  doi: 10.4103/NRR.NRR-D-24-00599
    Abstract ( 38 )   PDF (4097KB) ( 24 )   Save
    In clinical specialties focusing on neurological disorders, there is a need for comprehensive and integrated non-invasive, sensitive, and specific testing methods. Both Parkinson’s disease and multiple system atrophy are classified as α-synucleinopathies, characterized by abnormal accumulation of α-synuclein protein, which provides a shared pathological background for their comparative study. In addition, both Parkinson’s disease and multiple system atrophy involve neuronal death, a process that may release circulating cell–free DNA (cfDNA) into the bloodstream, leading to specific alterations. This premise formed the basis for investigating cell–free DNA as a potential biomarker. Cellfree DNA has garnered attention for its potential pathological significance, yet its characteristics in the context of Parkinson’s disease and multiple system atrophy are not fully understood. This study investigated the total concentration, nonapoptotic level, integrity, and cellfree DNA relative telomere length of cell-free DNA in the peripheral blood of 171 participants, comprising 76 normal controls, 62 patients with Parkinson’s disease, and 33 patients with multiple system atrophy. In our cohort, 75.8% of patients with Parkinson’s disease (stage 1–2 of Hoehn & Yahr) and 60.6% of patients with multiple system atrophy (disease duration less than 3 years) were in the early stages. The diagnostic potential of the cell-free DNA parameters was evaluated using receiver operating characteristic (ROC) analysis, and their association with disease prevalence was examined through logistic regression models, adjusting for confounders such as age, sex, body mass index, and education level. The results showed that cell-free DNA integrity was significantly elevated in both Parkinson’s disease and multiple system atrophy patients compared with normal controls (P < 0.001 for both groups), whereas cell-free DNA relative telomere length was markedly shorter (P = 0.003 for Parkinson’s disease and P = 0.010 for multiple system atrophy). Receiver operating characteristic analysis indicated that both cell-free DNA integrity and cell-free DNA relative telomere length possessed good diagnostic accuracy for differentiating Parkinson’s disease and multiple system atrophy from normal controls. Specifically, higher cell-free DNA integrity was associated with increased risk of Parkinson’s disease (odds ratio [OR]: 5.72; 95% confidence interval [CI]: 1.54–24.19) and multiple system atrophy (OR: 10.10; 95% CI: 1.55–122.98). Conversely, longer cell-free DNA relative telomere length was linked to reduced risk of Parkinson’s disease (OR: 0.16; 95% CI: 0.04–0.54) and multiple system atrophy (OR: 0.10; 95% CI: 0.01–0.57). These findings suggest that cell-free DNA integrity and cellfree DNA relative telomere length may serve as promising biomarkers for the early diagnosis of Parkinson’s disease and multiple system atrophy, potentially reflecting specific underlying pathophysiological processes of these neurodegenerative disorders.
    Related Articles | Metrics
    Nerve root magnetic stimulation regulates the synaptic plasticity of injured spinal cord by ascending sensory pathway
    Ya Zheng, Lingyun Cao, Dan Zhao, Qi Yang, Chunya Gu, Yeran Mao, Guangyue Zhu, Yulian Zhu, Jing Zhao, Dongsheng Xu
    2025, 20 (12):  3564-3573.  doi: 10.4103/NRR.NRR-D-24-00628
    Abstract ( 43 )   PDF (11914KB) ( 6 )   Save
    Promoting synaptic plasticity and inducing functional reorganization of residual nerve fibers hold clinical significance for restoring motor function following spinal cord injury. Neuromagnetic stimulation targeting the nerve roots has been shown to improve motor function by enhancing nerve conduction in the injured spinal cord and restoring the synaptic ultrastructure of both the sensory and motor cortex. However, our understanding of the neurophysiological mechanisms by which nerve root magnetic stimulation facilitates motor function recovery in the spinal cord is limited, and its role in neuroplasticity remains unclear. In this study, we established a model of spinal cord injury in adult male Sprague–Dawley rats by applying moderate compression at the T10 vertebra. We then performed magnetic stimulation on the L5 nerve root for 3 weeks, beginning on day 3 post-injury. At day 22 post-injury, we observed that nerve root magnetic stimulation downregulated the level of interleukin-6 in the injured spinal cord tissue of rats. Additionally, this treatment reduced neuronal damage and glial scar formation, and increased the number of neurons in the injured spinal cord. Furthermore, nerve root magnetic stimulation decreased the levels of acetylcholine, norepinephrine, and dopamine, and increased the expression of synaptic plasticity-related mRNA and proteins PSD95, GAP43, and Synapsin II. Taken together, these results showed that nerve root magnetic stimulation alleviated neuronal damage in the injured spinal cord, regulated synaptic plasticity, and suppressed inflammatory responses. These findings provide laboratory evidence for the clinical application of nerve root magnetic stimulation in the treatment of spinal cord injury.
    Related Articles | Metrics
    Systematic review of amyloid-beta clearance proteins from the brain to the periphery: implications for Alzheimer’s disease diagnosis and therapeutic targets
    Letian Huang, Mingyue Liu, Ze Li, Bing Li, Jiahe Wang, Ke Zhang
    2025, 20 (12):  3574-3590.  doi: 10.4103/NRR.NRR-D-24-00865
    Abstract ( 66 )   PDF (2826KB) ( 54 )   Save
    Amyloid-beta clearance plays a key role in the pathogenesis of Alzheimer’s disease. However, the variation in functional proteins involved in amyloid-beta clearance and their correlation with amyloid-beta levels remain unclear. In this study, we conducted meta-analyses and a systematic review using studies from the PubMed, Embase, Web of Science, and Cochrane Library databases, including journal articles published from inception to June 30, 2023. The inclusion criteria included studies comparing the levels of functional proteins associated with amyloid-beta clearance in the blood, cerebrospinal fluid, and brain of healthy controls, patients with mild cognitive impairment, and patients with Alzheimer’s disease. Additionally, we analyzed the correlation between these functional proteins and amyloid-beta levels in patients with Alzheimer’s disease. The methodological quality of the studies was assessed via the Newcastle‒Ottawa Scale. Owing to heterogeneity, we utilized either a fixed-effect or random-effect model to assess the 95% confidence interval (CI) of the standard mean difference (SMD) among healthy controls, patients with mild cognitive impairment, and patients with Alzheimer’s disease. The findings revealed significant alterations in the levels of insulin-degrading enzymes, neprilysin, matrix metalloproteinase-9, cathepsin D, receptor for advanced glycation end products, and P-glycoprotein in the brains of patients with Alzheimer’s disease, patients with mild cognitive impairment, and healthy controls. In cerebrospinal fluid, the levels of triggering receptor expressed on myeloid cells 2 and ubiquitin C-terminal hydrolase L1 are altered, whereas the levels of TREM2, CD40, CD40L, CD14, CD22, cathepsin D, cystatin C, and α2 M in peripheral blood differ. Notably, TREM2 and cathepsin D showed changes in both brain (SMD = 0.31, 95% CI: 0.16–0.47, P < 0.001, I 2 = 78.4%; SMD = 1.24, 95% CI: 0.01–2.48, P = 0.048, I 2 = 90.1%) and peripheral blood (SMD = 1.01, 95% CI: 0.35–1.66, P = 0.003, I 2 = 96.5%; SMD = 7.55, 95% CI: 3.92–11.18, P < 0.001, I 2 = 98.2%) samples. Furthermore, correlations were observed between amyloid-beta levels and the levels of TREM2 (r = 0.16, 95% CI: 0.04–0.28, P = 0.009, I 2 = 74.7%), neprilysin (r = –0.47, 95% CI: –0.80–0.14, P = 0.005, I 2 = 76.1%), and P-glycoprotein (r = –0.31, 95% CI: –0.51–0.11, P = 0.002, I 2 = 0.0%) in patients with Alzheimer’s disease. These findings suggest that triggering receptor expressed on myeloid cells 2 and cathepsin D could serve as potential diagnostic biomarkers for Alzheimer’s disease, whereas triggering receptor expressed on myeloid cells 2, neprilysin, and P-glycoprotein may represent potential therapeutic targets.
    Related Articles | Metrics
    A vascular endothelial growth factor–loaded chitosanhyaluronic acid hydrogel scaffold enhances the therapeutic effect of adipose-derived stem cells in the context of stroke
    Zhijian Zheng, Xiaohui Lin, Zijun Zhao, Qiang Lin, Ji Liu, Manli Chen, Wenwen Wu, Zhiyun Wu, Nan Liu, Hongbin Chen
    2025, 20 (12):  3591-3605. 
    Abstract ( 32 )   PDF (2908KB) ( 15 )   Save
    Adipose-derived stem cell, one type of mesenchymal stem cells, is a promising approach in treating ischemia-reperfusion injury caused by occlusion of the middle cerebral artery. However, its application has been limited by the complexities of the ischemic microenvironment. Hydrogel scaffolds, which are composed of hyaluronic acid and chitosan, exhibit excellent biocompatibility and biodegradability, making them promising candidates as cell carriers. Vascular endothelial growth factor is a crucial regulatory factor for stem cells. Both hyaluronic acid and chitosan have the potential to make the microenvironment more hospitable to transplanted stem cells, thereby enhancing the therapeutic effect of mesenchymal stem cell transplantation in the context of stroke. Here, we found that vascular endothelial growth factor significantly improved the activity and paracrine function of adipose-derived stem cells. Subsequently, we developed a chitosan-hyaluronic acid hydrogel scaffold that incorporated vascular endothelial growth factor and first injected the scaffold into an animal model of cerebral ischemiareperfusion injury. When loaded with adipose-derived stem cells, this vascular endothelial growth factor–loaded scaffold markedly reduced neuronal apoptosis caused by oxygen-glucose deprivation/reoxygenation and substantially restored mitochondrial membrane potential and axon morphology. Further in vivo experiments revealed that this vascular endothelial growth factor–loaded hydrogel scaffold facilitated the transplantation of adipose-derived stem cells, leading to a reduction in infarct volume and neuronal apoptosis in a rat model of stroke induced by transient middle cerebral artery occlusion. It also helped maintain mitochondrial integrity and axonal morphology, greatly improving rat motor function and angiogenesis. Therefore, utilizing a hydrogel scaffold loaded with vascular endothelial growth factor as a stem cell delivery system can mitigate the adverse effects of ischemic microenvironment on transplanted stem cells and enhance the therapeutic effect of stem cells in the context of stroke.
    Related Articles | Metrics
    Dynamic development of microglia and macrophages after spinal cord injury
    Hu-Yao Zhou, Xia Wang, Yi Li, Duan Wang, Xuan-Zi Zhou, Nong Xiao, Guo-Xing Li, Gang Li
    2025, 20 (12):  3606-3619.  doi: 10.4103/NRR.NRR-D-24-00063
    Abstract ( 69 )   PDF (122135KB) ( 14 )   Save
    Secondary injury following spinal cord injury is primarily characterized by a complex inflammatory response, with resident microglia and infiltrating macrophages playing pivotal roles. While previous studies have grouped these two cell types together based on similarities in structure and function, an increasing number of studies have demonstrated that microglia and macrophages exhibit differences in structure and function and have different effects on disease processes. In this study, we used single-cell RNA sequencing and spatial transcriptomics to identify the distinct evolutionary paths of microglia and macrophages following spinal cord injury. Our results showed that microglia were activated to a pro-inflammatory phenotype immediately after spinal cord injury, gradually transforming to an anti-inflammatory steady state phenotype as the disease progressed. Regarding macrophages, our findings highlighted abundant communication with other cells, including fibroblasts and neurons. Both pro-inflammatory and neuroprotective effects of macrophages were also identified; the pro-inflammatory effect may be related to integrin β2 (Itgb2) and the neuroprotective effect may be related to the oncostatin M pathway. These findings were validated by in vivo experiments. This research underscores differences in the cellular dynamics of microglia and macrophages following spinal cord injury, and may offer new perspectives on inflammatory mechanisms and potential therapeutic targets.
    Related Articles | Metrics
    Novel genes involved in vascular dysfunction of the middle temporal gyrus in Alzheimer’s disease: transcriptomics combined with machine learning analysis
    Meiling Wang, Aojie He, Yubing Kang, Zhaojun Wang, Yahui He, Kahleong Lim, Chengwu Zhang, Li Lu
    2025, 20 (12):  3620-3634.  doi: 10.4103/NRR.NRR-D-23-02004
    Abstract ( 70 )   PDF (9252KB) ( 8 )   Save
    Studies have shown that vascular dysfunction is closely related to the pathogenesis of Alzheimer’s disease. The middle temporal gyrus region of the brain is susceptible to pronounced impairment in Alzheimer’s disease. Identification of the molecules involved in vascular aberrance of the middle temporal gyrus would support elucidation of the mechanisms underlying Alzheimer’s disease and discovery of novel targets for intervention. We carried out single-cell transcriptomic analysis of the middle temporal gyrus in the brains of patients with Alzheimer’s disease and healthy controls, revealing obvious changes in vascular function. CellChat analysis of intercellular communication in the middle temporal gyrus showed that the number of cell interactions in this region was decreased in Alzheimer’s disease patients, with altered intercellular communication of endothelial cells and pericytes being the most prominent. Differentially expressed genes were also identified. Using the CellChat results, AUCell evaluation of the pathway activity of specific cells showed that the obvious changes in vascular function in the middle temporal gyrus in Alzheimer’s disease were directly related to changes in the vascular endothelial growth factor (VEGF)A–VEGF receptor (VEGFR) 2 pathway. AUCell analysis identified subtypes of endothelial cells and pericytes directly related to VEGFA–VEGFR2 pathway activity. Two subtypes of middle temporal gyrus cells showed significant alteration in AD: endothelial cells with high expression of Erb-B2 receptor tyrosine kinase 4 (ERBB4high) and pericytes with high expression of angiopoietin-like 4 (ANGPTL4high). Finally, combining bulk RNA sequencing data and two machine learning algorithms (least absolute shrinkage and selection operator and random forest), four characteristic Alzheimer’s disease feature genes were identified: somatostatin (SST), protein tyrosine phosphatase non-receptor type 3 (PTPN3), glutinase (GL3), and tropomyosin 3 (PTM3). These genes were downregulated in the middle temporal gyrus of patients with Alzheimer’s disease and may be used to target the VEGF pathway. Alzheimer’s disease mouse models demonstrated consistent altered expression of these genes in the middle temporal gyrus. In conclusion, this study detected changes in intercellular communication between endothelial cells and pericytes in the middle temporal gyrus and identified four novel feature genes related to middle temporal gyrus and vascular functioning in patients with Alzheimer’s disease. These findings contribute to a deeper understanding of the molecular mechanisms underlying Alzheimer’s disease and present novel treatment targets.
    Related Articles | Metrics
    Single-cell RNA sequencing reveals the heterogeneity and interactions of immune cells and Müller glia during zebrafish retina regeneration
    Hui Xu, Lining Cao, Yuxi Chen, Cuiping Zhou, Jie Xu, Zhuolin Zhang, Xiangyu Li, Lihua Liu, Jianfeng Lu
    2025, 20 (12):  3635-3648.  doi: 10.4103/NRR.NRR-D-23-02083
    Abstract ( 77 )   PDF (7430KB) ( 12 )   Save
    Inflammation plays a crucial role in the regeneration of fish and avian retinas. However, how inflammation regulates Müller glia (MG) reprogramming remains unclear. Here, we used single-cell RNA sequencing to investigate the cell heterogeneity and interactions of MG and immune cells in the regenerating zebrafish retina. We first showed that two types of quiescent MG (resting MG1 and MG2) reside in the uninjured retina. Following retinal injury, resting MG1 transitioned into an activated state expressing known reprogramming genes, while resting MG2 gave rise to rod progenitors. We further showed that retinal microglia can be categorized into three subtypes (microglia-1, microglia-2, and proliferative) and pseudotime analysis demonstrated dynamic changes in microglial status following retinal injury. Analysis of cell–cell interactions indicated extensive crosstalk between immune cells and MG, with many interactions shared among different immune cell types. Finally, we showed that inflammation activated Jak1–Stat3 signaling in MG, promoting their transition from a resting to an activated state. Our study reveals the cell heterogeneity and crosstalk of immune cells and MG in zebrafish retinal repair, and may provide valuable insights into future mammalian retina regeneration.
    Related Articles | Metrics