Loading...

Table of Content

    15 November 2025, Volume 20 Issue 11 Previous Issue   
    For Selected: Toggle Thumbnails
    Epigenetic regulation of the inflammatory response in stroke
    Jingyi Liang, Fei Yang, Zixiao Li, Qian Li
    2025, 20 (11):  3045-3062.  doi: 10.4103/NRR.NRR-D-24-00672
    Abstract ( 115 )   PDF (2705KB) ( 81 )   Save
    Stroke is classified as ischemic or hemorrhagic, and there are few effective treatments for either type. Immunologic mechanisms play a critical role in secondary brain injury following a stroke, which manifests as cytokine release, blood–brain barrier disruption, neuronal cell death, and ultimately behavioral impairment. Suppressing the inflammatory response has been shown to mitigate this cascade of events in experimental stroke models. However, in clinical trials of anti-inflammatory agents, longterm immunosuppression has not demonstrated significant clinical benefits for patients. This may be attributable to the dichotomous roles of inflammation in both tissue injury and repair, as well as the complex pathophysiologic inflammatory processes in stroke. Inhibiting acute harmful inflammatory responses or inducing a phenotypic shift from a pro-inflammatory to an anti-inflammatory state at specific time points after a stroke are alternative and promising therapeutic strategies. Identifying agents that can modulate inflammation requires a detailed understanding of the inflammatory processes of stroke. Furthermore, epigenetic reprogramming plays a crucial role in modulating post-stroke inflammation and can potentially be exploited for stroke management. In this review, we summarize current findings on the epigenetic regulation of the inflammatory response in stroke, focusing on key signaling pathways including nuclear factor-kappa B, Janus kinase/ signal transducer and activator of transcription, and mitogen-activated protein kinase as well as inflammasome activation. We also discuss promising molecular targets for stroke treatment. The evidence to date indicates that therapeutic targeting of the epigenetic regulation of inflammation can shift the balance from inflammation-induced tissue injury to repair following stroke, leading to improved post-stroke outcomes.
    Related Articles | Metrics
    Human induced pluripotent stem cell–derived therapies for regeneration after central nervous system injury
    Stephen Vidman, Yee Hang Ethan Ma, Nolan Fullenkamp, Giles W. Plant
    2025, 20 (11):  3063-3075.  doi: 10.4103/NRR.NRR-D-24-00901
    Abstract ( 69 )   PDF (972KB) ( 79 )   Save
    In recent years, the progression of stem cell therapies has shown great promise in advancing the nascent field of regenerative medicine. Considering the non-regenerative nature of the mature central nervous system, the concept that “blank” cells could be reprogrammed and functionally integrated into host neural networks remained intriguing. Previous work has also demonstrated the ability of such cells to stimulate intrinsic growth programs in post-mitotic cells, such as neurons. While embryonic stem cells demonstrated great potential in treating central nervous system pathologies, ethical and technical concerns remained. These barriers, along with the clear necessity for this type of treatment, ultimately prompted the advent of induced pluripotent stem cells. The advantage of pluripotent cells in central nervous system regeneration is multifaceted, permitting differentiation into neural stem cells, neural progenitor cells, glia, and various neuronal subpopulations. The precise spatiotemporal application of extrinsic growth factors in vitro, in addition to microenvironmental signaling in vivo, influences the efficiency of this directed differentiation. While the pluri- or multipotency of these cells is appealing, it also poses the risk of unregulated differentiation and teratoma formation. Cells of the neuroectodermal lineage, such as neuronal subpopulations and glia, have been explored with varying degrees of success. Although the risk of cancer or teratoma formation is greatly reduced, each subpopulation varies in effectiveness and is influenced by a myriad of factors, such as the timing of the transplant, pathology type, and the ratio of accompanying progenitor cells. Furthermore, successful transplantation requires innovative approaches to develop delivery vectors that can mitigate cell death and support integration. Lastly, host immune responses to allogeneic grafts must be thoroughly characterized and further developed to reduce the need for immunosuppression. Translation to a clinical setting will involve careful consideration when assessing both physiologic and functional outcomes. This review will highlight both successes and challenges faced when using human induced pluripotent stem cell-derived cell transplantation therapies to promote endogenous regeneration.
    Related Articles | Metrics
    Targeting capabilities of engineered extracellular vesicles for the treatment of neurological diseases
    Xinyu Yang, Xiangyu Gao, Xiaofan Jiang, Kangyi Yue, Peng Luo
    2025, 20 (11):  3076-3094.  doi: 10.4103/NRR.NRR-D-24-00462
    Abstract ( 190 )   PDF (3383KB) ( 105 )   Save
    Recent advances in research on extracellular vesicles have significantly enhanced their potential as therapeutic agents for neurological diseases. Owing to their therapeutic properties and ability to cross the blood–brain barrier, extracellular vesicles are recognized as promising drug delivery vehicles for various neurological conditions, including ischemic stroke, traumatic brain injury, neurodegenerative diseases, glioma, and psychosis. However, the clinical application of natural extracellular vesicles is hindered by their limited targeting ability and short clearance from the body. To address these limitations, multiple engineering strategies have been developed to enhance the targeting capabilities of extracellular vesicles, thereby enabling the delivery of therapeutic contents to specific tissues or cells. Therefore, this review aims to highlight the latest advancements in natural and targeting-engineered extracellular vesicles, exploring their applications in treating traumatic brain injury, ischemic stroke, Parkinson’s disease, Alzheimer’s disease, amyotrophic lateral sclerosis, glioma, and psychosis. Additionally, we summarized recent clinical trials involving extracellular vesicles and discussed the challenges and future prospects of using targeting-engineered extracellular vesicles for drug delivery in treating neurological diseases. This review offers new insights for developing highly targeted therapies in this field.
    Related Articles | Metrics
    Exploring the interaction between the gut microbiota and cyclic adenosine monophosphate-protein kinase A signaling pathway: a potential therapeutic approach for neurodegenerative diseases
    Fengcheng Deng, Dan Yang, Lingxi Qing, Yifei Chen, Jilian Zou, Meiling Jia, Qian Wang, Runda Jiang, Lihua Huang
    2025, 20 (11):  3095-3112.  doi: 10.4103/NRR.NRR-D-24-00607
    Abstract ( 102 )   PDF (4201KB) ( 124 )   Save
    The interaction between the gut microbiota and cyclic adenosine monophosphate (cAMP)- protein kinase A (PKA) signaling pathway in the host’s central nervous system plays a crucial role in neurological diseases and enhances communication along the gut–brain axis. The gut microbiota influences the cAMP-PKA signaling pathway through its metabolites, which activates the vagus nerve and modulates the immune and neuroendocrine systems. Conversely, alterations in the cAMP-PKA signaling pathway can affect the composition of the gut microbiota, creating a dynamic network of microbial-host interactions. This reciprocal regulation affects neurodevelopment, neurotransmitter control, and behavioral traits, thus playing a role in the modulation of neurological diseases. The coordinated activity of the gut microbiota and the cAMP-PKA signaling pathway regulates processes such as amyloid-β protein aggregation, mitochondrial dysfunction, abnormal energy metabolism, microglial activation, oxidative stress, and neurotransmitter release, which collectively influence the onset and progression of neurological diseases. This study explores the complex interplay between the gut microbiota and cAMP-PKA signaling pathway, along with its implications for potential therapeutic interventions in neurological diseases. Recent pharmacological research has shown that restoring the balance between gut flora and cAMP-PKA signaling pathway may improve outcomes in neurodegenerative diseases and emotional disorders. This can be achieved through various methods such as dietary modifications, probiotic supplements, Chinese herbal extracts, combinations of Chinese herbs, and innovative dosage forms. These findings suggest that regulating the gut microbiota and cAMP-PKA signaling pathway may provide valuable evidence for developing novel therapeutic approaches for neurodegenerative diseases.
    Related Articles | Metrics
    Understanding the link between type 2 diabetes mellitus and Parkinson’s disease: role of brain insulin resistance
    Theodora Ntetsika, Sergiu-Bogdan Catrina, Ioanna Markaki
    2025, 20 (11):  3113-3123.  doi: 10.4103/NRR.NRR-D-23-01910
    Abstract ( 33 )   PDF (7503KB) ( 6 )   Save
    Type 2 diabetes mellitus and Parkinson’s disease are chronic diseases linked to a growing pandemic that affects older adults and causes significant socio-economic burden. Epidemiological data supporting a close relationship between these two aging-related diseases have resulted in the investigation of shared pathophysiological molecular mechanisms. Impaired insulin signaling in the brain has gained increasing attention during the last decade and has been suggested to contribute to the development of Parkinson’s disease through the dysregulation of several pathological processes. The contribution of type 2 diabetes mellitus and insulin resistance in neurodegeneration in Parkinson’s disease, with emphasis on brain insulin resistance, is extensively discussed in this article and new therapeutic strategies targeting this pathological link are presented and reviewed
    Related Articles | Metrics
    Copper homeostasis and neurodegenerative diseases
    Yuanyuan Wang, Daidi Li, Kaifei Xu, Guoqing Wang, Feng Zhang
    2025, 20 (11):  3124-3143.  doi: 10.4103/NRR.NRR-D-24-00642
    Abstract ( 74 )   PDF (5601KB) ( 135 )   Save
    Copper, one of the most prolific transition metals in the body, is required for normal brain physiological activity and allows various functions to work normally through its range of concentrations. Copper homeostasis is meticulously maintained through a complex network of copper-dependent proteins, including copper transporters (CTR1 and CTR2), the two copper ion transporters the Cu -transporting ATPase 1 (ATP7A) and Cu-transporting beta (ATP7B), and the three copper chaperones ATOX1, CCS, and COX17. Disruptions in copper homeostasis can lead to either the deficiency or accumulation of copper in brain tissue. Emerging evidence suggests that abnormal copper metabolism or copper binding to various proteins, including ceruloplasmin and metallothionein, is involved in the pathogenesis of neurodegenerative disorders. However, the exact mechanisms underlying these processes are not known. Copper is a potent oxidant that increases reactive oxygen species production and promotes oxidative stress. Elevated reactive oxygen species levels may further compromise mitochondrial integrity and cause mitochondrial dysfunction. Reactive oxygen species serve as key signaling molecules in copper-induced neuroinflammation, with elevated levels activating several critical inflammatory pathways. Additionally, copper can bind aberrantly to several neuronal proteins, including alphasynuclein, tau, superoxide dismutase 1, and huntingtin, thereby inducing neurotoxicity and ultimately cell death. This study focuses on the latest literature evaluating the role of copper in neurodegenerative diseases, with a particular focus on copper-containing metalloenzymes and copper-binding proteins in the regulation of copper homeostasis and their involvement in neurodegenerative disease pathogenesis. By synthesizing the current findings on the functions of copper in oxidative stress, neuroinflammation, mitochondrial dysfunction, and protein misfolding, we aim to elucidate the mechanisms by which copper contributes to a wide range of hereditary and neuronal disorders, such as Wilson’s disease, Menkes’ disease, Alzheimer’s disease, Parkinson’s disease, amyotrophic lateral sclerosis, Huntington’s disease, and multiple sclerosis. Potential clinically significant therapeutic targets, including superoxide dismutase 1, D-penicillamine, and 5,7-dichloro2-[(dimethylamino)methyl]-8-hydroxyquinoline, along with their associated therapeutic agents, are further discussed. Ultimately, we collate evidence that copper homeostasis may function in the underlying etiology of several neurodegenerative diseases and offer novel insights into the potential prevention and treatment of these diseases based on copper homeostasis.
    Related Articles | Metrics
    Temporal dynamics of neonatal hypoxic–ischemic encephalopathy injuries on magnetic resonance imaging
    Holly Flyger, Samantha J. Holdsworth, Alistair J. Gunn, Laura Bennet, Hamid Abbasi
    2025, 20 (11):  3144-3150.  doi: 10.4103/NRR.NRR-D-24-00970
    Abstract ( 37 )   PDF (2298KB) ( 39 )   Save
    Moderate to severe perinatal hypoxic–ischemic encephalopathy occurs in ~1 to 3/1000 live births in high-income countries and is associated with a significant risk of death or neurodevelopmental disability. Detailed assessment is important to help identify highrisk infants, to help families, and to support appropriate interventions. A wide range of monitoring tools is available to assess changes over time, including urine and blood biomarkers, neurological examination, and electroencephalography. At present, magnetic resonance imaging is unique as although it is expensive and not suited to monitoring the early evolution of hypoxic–ischemic encephalopathy by a week of life it can provide direct insight into the anatomical changes in the brain after hypoxic–ischemic encephalopathy and so offers strong prognostic information on the long-term outcome after hypoxic– ischemic encephalopathy. This review investigated the temporal dynamics of neonatal hypoxic-ischemic encephalopathy injuries, with a particular emphasis on exploring the correlation between the prognostic implications of magnetic resonance imaging scans in the first week of life and their relationship to long-term outcome prediction, particularly for infants treated with therapeutic hypothermia. A comprehensive literature search, from 2016 to 2024, identified 20 pertinent articles. This review highlights that while the optimal timing of magnetic resonance imaging scans is not clear, overall, it suggests that magnetic resonance imaging within the first week of life provides strong prognostic accuracy. Many challenges limit the timing consistency, particularly the need for intensive care and clinical monitoring. Conversely, although most reports examined the prognostic value of scans taken between 4 and 10 days after birth, there is evidence from small numbers of cases that, at times, brain injury may continue to evolve for weeks after birth. This suggests that in the future it will be important to explore a wider range of times after hypoxic–ischemic encephalopathy to fully understand the optimal timing for predicting long-term outcomes.
    Related Articles | Metrics
    Advances in therapies using mesenchymal stem cells and their exosomes for treatment of peripheral nerve injury: state of the art and future perspectives
    Fatima Aldali, Chunchu Deng, Mingbo Nie, Hong Chen
    2025, 20 (11):  3151-3171.  doi: 10.4103/NRR.NRR-D-24-00235
    Abstract ( 112 )   PDF (2415KB) ( 99 )   Save
    “Peripheral nerve injury” refers to damage or trauma affecting nerves outside the brain and spinal cord. Peripheral nerve injury results in movements or sensation impairments, and represents a serious public health problem. Although severed peripheral nerves have been effectively joined and various therapies have been offered, recovery of sensory or motor functions remains limited, and efficacious therapies for complete repair of a nerve injury remain elusive. The emerging field of mesenchymal stem cells and their exosome-based therapies hold promise for enhancing nerve regeneration and function. Mesenchymal stem cells, as large living cells responsive to the environment, secrete various factors and exosomes. The latter are nano-sized extracellular vesicles containing bioactive molecules such as proteins, microRNA, and messenger RNA derived from parent mesenchymal stem cells. Exosomes have pivotal roles in cell-to-cell communication and nervous tissue function, offering solutions to changes associated with cell-based therapies. Despite ongoing investigations, mesenchymal stem cells and mesenchymal stem cell– derived exosome-based therapies are in the exploratory stage. A comprehensive review of the latest preclinical experiments and clinical trials is essential for deep understanding of therapeutic strategies and for facilitating clinical translation. This review initially explores current investigations of mesenchymal stem cells and mesenchymal stem cell–derived exosomes in peripheral nerve injury, exploring the underlying mechanisms. Subsequently, it provides an overview of the current status of mesenchymal stem cell and exosomebased therapies in clinical trials, followed by a comparative analysis of therapies utilizing mesenchymal stem cells and exosomes. Finally, the review addresses the limitations and challenges associated with use of mesenchymal stem cell–derived exosomes, offering potential solutions and guiding future directions.
    Related Articles | Metrics
    A systematic review of progenitor survival and maturation in Parkinsonian models
    Giulia Comini, Eilís Dowd
    2025, 20 (11):  3172-3178.  doi: 10.4103/NRR.NRR-D-24-00894
    Abstract ( 42 )   PDF (1619KB) ( 25 )   Save
    Stem cell–based brain repair is a promising emergent therapy for Parkinson’s disease based on years of foundational research using human fetal donors as a cell source. Unlike current therapeutic options for patients, this approach has the potential to provide longterm stem cell–derived reconstruction and restoration of the dopaminergic input to denervated regions of the brain allowing for restoration of certain functions to patients. The ultimate clinical success of stem cell–derived brain repair will depend on both the safety and efficacy of the approach and the latter is dependent on the ability of the transplanted cells to survive and differentiate into functional dopaminergic neurons in the Parkinsonian brain. Because the pre-clinical literature suggests that there is considerable variability in survival and differentiation between studies, the aim of this systematic review was to assess these parameters in human stem cell-derived dopaminergic progenitor transplant studies in animal models of Parkinson’s disease. A defined systematic search of the PubMed database was completed to identify relevant studies published up to March 2024. After screening, 76 articles were included in the analysis from which 178 separate transplant studies were identified. From these, graft survival could be assessed in 52 studies and differentiation in 129 studies. Overall, we found that graft survival ranged from < 1% to 500% of cells transplanted, with a median of 51% of transplanted cells surviving in the brain; while dopaminergic differentiation of the cells ranged from 0% to 46% of cells transplanted with a median of 3%. This systematic review suggests that there is considerable scope for improvement in the differentiation of stem cell–derived dopaminergic progenitors to maximize the therapeutic potential of this approach for patients.
    Related Articles | Metrics
    Oligodendroglial heterogeneity in health, disease, and recovery: deeper insights into myelin dynamics
    Pieter-Jan Serneels, Julie D. De Schutter, Lies De Groef, Lieve Moons, Steven Bergmans
    2025, 20 (11):  3179-3192.  doi: 10.4103/NRR.NRR-D-24-00694
    Abstract ( 37 )   PDF (6628KB) ( 49 )   Save
    Decades of research asserted that the oligodendroglial lineage comprises two cell types: oligodendrocyte precursor cells and oligodendrocytes. However, recent studies employing single-cell RNA sequencing techniques have uncovered novel cell states, prompting a revision of the existing terminology. Going forward, the oligodendroglial lineage should be delineated into five distinct cell states: oligodendrocyte precursor cells, committed oligodendrocyte precursor cells, newly formed oligodendrocytes, myelin-forming oligodendrocytes, and mature oligodendrocytes. This new classification system enables a deeper understanding of the oligodendroglia in both physiological and pathological contexts. Adopting this uniform terminology will facilitate comparison and integration of data across studies. This, including the consolidation of findings from various demyelinating models, is essential to better understand the pathogenesis of demyelinating diseases. Additionally, comparing injury models across species with varying regenerative capacities can provide insights that may lead to new therapeutic strategies to overcome remyelination failure. Thus, by standardizing terminology and synthesizing data from diverse studies across different animal models, we can enhance our understanding of myelin pathology in central nervous system disorders such as multiple sclerosis, Alzheimer’s disease, and amyotrophic lateral sclerosis, all of which involve oligodendroglial and myelin dysfunction.
    Related Articles | Metrics
    Induced pluripotent stem cell–related approaches to generate dopaminergic neurons for Parkinson’s disease
    Ling-Xiao Yi, Hui Ren Woon, Genevieve Saw, Li Zeng, Eng King Tan, Zhi Dong Zhou
    2025, 20 (11):  3193-3206.  doi: 10.4103/NRR.NRR-D-24-00771
    Abstract ( 45 )   PDF (11935KB) ( 17 )   Save
    The progressive loss of dopaminergic neurons in affected patient brains is one of the pathological features of Parkinson’s disease, the second most common human neurodegenerative disease. Although the detailed pathogenesis accounting for dopaminergic neuron degeneration in Parkinson’s disease is still unclear, the advancement of stem cell approaches has shown promise for Parkinson’s disease research and therapy. The induced pluripotent stem cells have been commonly used to generate dopaminergic neurons, which has provided valuable insights to improve our understanding of Parkinson’s disease pathogenesis and contributed to anti-Parkinson’s disease therapies. The current review discusses the practical approaches and potential applications of induced pluripotent stem cell techniques for generating and differentiating dopaminergic neurons from induced pluripotent stem cells. The benefits of induced pluripotent stem cell-based research are highlighted. Various dopaminergic neuron differentiation protocols from induced pluripotent stem cells are compared. The emerging three-dimension-based brain organoid models compared with conventional two-dimensional cell culture are evaluated. Finally, limitations, challenges, and future directions of induced pluripotent stem cell– based approaches are analyzed and proposed, which will be significant to the future application of induced pluripotent stem cell–related techniques for Parkinson’s disease.
    Related Articles | Metrics
    Iron-handling solute carrier SLC22A17 as a blood–brain barrier target after stroke
    Xueqi Ren, Wenlu Li
    2025, 20 (11):  3207-3208.  doi: 10.4103/NRR.NRR-D-24-00811
    Abstract ( 44 )   PDF (464KB) ( 10 )   Save
    The pathophysiology of ischemic stroke is complex and multifactorial, involving various forms of cell death such as apoptosis, autophagy, and necrosis. A recent study suggests that oxidative and inflammatory stress can induce ferroptosis, a specialized form of cell death characterized by the accumulation of lipid peroxides dependent on intracellular iron overload (Li and Jia, 2023). Although clinical trials have explored iron chelators—agents that bind to free iron and reduce its availability—as potential therapies for ischemic stroke to mitigate ferroptosis, a recent mechanistic study has focused on neuronal cells (Hanafy et al., 2019). Brain endothelial cells, which are essential for iron transport and homeostasis in the brain, particularly after stroke, express various iron transporters such as transferrin receptors, divalent metal transporter 1, and ferroportin to manage iron levels (Chen et al., 2022). This suggests that ferroptosis may play a significant role in brain vascular injury.
    Related Articles | Metrics
    Reassessing the AMPK-MTORC1 balance in autophagy in the central nervous system
    Marta García-Juan, Mario Villa, Irene Benito-Cuesta, Lara Ordóñez-Gutiérrez, Francisco Wandosell
    2025, 20 (11):  3209-3210.  doi: 10.4103/NRR.NRR-D-24-00733
    Abstract ( 111 )   PDF (549KB) ( 13 )   Save
    Autophagy is a cellular degradation and recycling system, indispensable for cellular and organ development, homeostasis, and function. This cellular process is evolutionarily highly conserved to quality control of many proteins and dysfunctional organelles, which finally recycle components as amino acids. This process is effective during normal physiology as part of anabolism and plays an additional important role during starvation (Dikic and Elazar, 2018). Different types of autophagy have been characterized based on their dynamic, mechanism of action, target substrates, and protein markers. Some of them are macroautophagy (hereafter called “autophagy”), microautophagy, and chaperone-mediated autophagy (Fleming et al., 2022).
    Related Articles | Metrics
    The complex role of protocadherin-19 in brain function: a focus on the oxytocin system
    Sara Mazzoleni, Marta Busnelli, Silvia Bassani
    2025, 20 (11):  3211-3212.  doi: 10.4103/NRR.NRR-D-24-00847
    Abstract ( 38 )   PDF (1041KB) ( 27 )   Save
    Mutations in the protocadherin-19 (PCDH19) gene (Xq22.1) cause the X-linked syndrome known as developmental and epileptic encephalopathy 9 (DEE9, OMIM # 300088) (Dibbens et al., 2008). DEE9 is characterized by early-onset clustering epilepsy associated with intellectual disability ranging from mild to profound, autism spectrum disorder, and other neuropsychiatric features including schizophrenia, anxiety, attentiondeficit/ hyperactivity, and obsessive or aggressive behaviors. While seizures may become less frequent in adolescence, psychiatric comorbidities persist and often worsen with age (Dibbens et al., 2008; Kolc et al., 2020).
    Related Articles | Metrics
    Vagus nerve stimulation in intracerebral hemorrhage: the need for further research
    Sheharyar S. Baig , Ali N. Ali, Arshad Majid
    2025, 20 (11):  3213-3214.  doi: 10.4103/NRR.NRR-D-24-00756
    Abstract ( 58 )   PDF (865KB) ( 22 )   Save
    Vagus nerve stimulation (VNS) and stroke: Stroke is the second leading cause of death and the third leading cause of disability worldwide (Baig et al., 2023). There have been significant paradigm shifts in the management of acute ischemic stroke through mechanical thrombectomy. In chronic ischemic stroke, invasive VNS paired with rehabilitation is associated with a significant increase in upper limb motor recovery and is FDA-approved (Baig et al., 2023). There are no treatments of similar efficacy in acute intracerebral hemorrhage (ICH) where several promising trials, e.g., TICH-2, STOP-AUST, and TRAIGE did not show improvements in functional outcomes (Puy et al., 2023).
    Related Articles | Metrics
    Inspires effective alternatives to backpropagation: predictive coding helps understand and build learning
    Zhenghua Xu, Miao Yu , Yuhang Song
    2025, 20 (11):  3215-3216.  doi: 10.4103/NRR.NRR-D-24-00629
    Abstract ( 61 )   PDF (2518KB) ( 85 )   Save
    Artificial neural networks are capable of machine learning by simulating the hierarchical structure of the human brain. To enable learning by brain and machine, it is essential to accurately identify and correct the prediction errors, referred to as credit assignment (Lillicrap et al., 2020). It is critical to develop artificial intelligence by understanding how the brain deals with credit assignment in neuroscience.
    Related Articles | Metrics
    Beginning from the end: the presynaptic terminal as a pathomechanism hub in frontotemporal dementia and amyotrophic lateral sclerosis
    Laura Huggon, Emma L. Clayton
    2025, 20 (11):  3217-3218.  doi: 10.4103/NRR.NRR-D-24-00639
    Abstract ( 41 )   PDF (968KB) ( 16 )   Save
    Frontotemporal dementia and amyotrophic lateral sclerosis: Frontotemporal dementia ( F T D ) a n d a m y o t r o p h i c l a t e r a l s c l e r o s i s (ALS) are neurodegenerative diseases with significant overlapping attributes. While these neurodegenerative diseases affect different neuronal populations (with FTD affecting neurons of the frontal and temporal lobes, and ALS affecting upper and lower motor neurons), these two diseases are complexly intertwined. FTD and ALS exist on a disease spectrum, with shared genetic causes, clinical presentations, and pathologies. The continuum of the genetic disease spectrum runs from those genetic mutations that cause ALS (SOD-1) through those that can cause ALS/FTD (TDP-43, FUS, CHCHD10, C9ORF72, TBK-1, UBQLN2, and CHMP2B) to those that cause FTD (MAPT and PGRN) (reviewed in Abramzon et al., 2020). Models of these genetic causes of disease have been instrumental in advancing our understanding of disease mechanisms.
    Related Articles | Metrics
    Misfolded amyloid-beta conformational variants (strains) as drivers of Alzheimer’s disease neuropathology
    Salvatore Saieva, Rodrigo Morales
    2025, 20 (11):  3219-3220.  doi: 10.4103/NRR.NRR-D-24-00699
    Abstract ( 37 )   PDF (1256KB) ( 22 )   Save
    Pathological and clinical variability in Alzheimer’s disease (AD): AD is clinically characterized by progressive memory loss and cognitive impairment. From a pathological point of view, the main features of AD are the deposition of amyloid plaques (composed of amyloid-beta, Aβ) and neurofibrillary tangles containing hyperphosphorylated Tau in the brain, accompanied by neuronal and synaptic loss, neuroinflammation and brain atrophy (Jellinger, 2022). Regardless of these common traits, growing evidence shows increased heterogeneity in the brain of AD patients considering both clinical manifestations and pathological features. Mounting evidence points to the variable conformations that misfolded Aβ can acquire (referred to as “Aβ strains”) as the source of this pathological and clinical variability. The existence of Aβ strains is relevant as they may also predict prognosis and responses to treatments. The concept of Aβ strains is analogous to that coined for infectious prion strains, which are further defined below (Jellinger, 2022).
    Related Articles | Metrics
    Proteostasis failure: at the intersection between aging and Alzheimer’s disease
    Natalia Poblete, Claudio Hetz
    2025, 20 (11):  3221-3222.  doi: 10.4103/NRR.NRR-D-24-00658
    Abstract ( 55 )   PDF (796KB) ( 26 )   Save
    Lifestyle and demographics of the world’s population are causing serious health problems impacting the brain, increasing the incidence of Alzheimer’s disease (AD) and other types of dementia. Although we have gained important insights into the pathogenic mechanisms of AD, only palliative care is available to patients. AD is characterized by the abnormal deposition of protein aggregates in the brain formed by amyloid β and hyper-phosphorylated, Tau in addition to neuroinflammation. The current failure to treat neurodegenerative diseases underlies the fact that the brain is highly plastic and clinical symptoms are manifested when extensive brain damage has progressed. Thus, new concepts need to be tested to develop preventive strategies that extend healthspan. For half of a century, biomedicine has focused on disease as a means to understand health, developing approaches to target pathogenic features. But health is not simply the absence of disease. It is an active process that enables the organism to cope with intrinsic and extrinsic changes (López-Otín and Kroemer, 2021). Thus, a sensible strategy should focus on extending brain healthspan to prevent disease, by improving synaptic function and connectivity, the initial pathological targets, before neurodegeneration and irreversible brain damage have occurred. The decay in the buffering capacity of the proteostasis network has been pointed out as a primary hallmark of aging (Kaushik and Cuervo, 2015), a phenomenon that may contribute to AD pathogenesis. Strategies to improve proteostasis have been tested in multiple models of neurodegenerative diseases, observing outstanding protective effects. The current challenge is to move forward those interventions into suitable strategies that can be tested in the clinic. Because proteostasis is essential to the normal functions of many organs, it is predicted that the chronic administration of small molecules to target proteostasis may have adverse effects in the long term. Gene therapy is emerging as an alternative because specific brain regions can be targeted locally without affecting the function of other organs. We recently tested a gene therapy strategy to improve proteostasis by artificially enhancing the repair programs to improve proteostasis in the context of AD. Our results indicated outstanding effects in improving cognition and reducing amyloid-β plaques in models of AD (Duran-Aniotz et al., 2023).
    Related Articles | Metrics
    Targeting Epac2 and GluA3- containing AMPARs: a novel therapeutic strategy for Alzheimer’s disease
    Tong Zhang, Martina Schmidt
    2025, 20 (11):  3223-3224.  doi: 10.4103/NRR.NRR-D-24-00751
    Abstract ( 31 )   PDF (565KB) ( 23 )   Save
    Alzheimer’s disease (AD) is a neurodegenerative disease that manifests progressive decline in memory and cognition. In the early stage of AD, memory retrieval is impaired preceding memory acquisition and consolidation (Roy et al., 2016). Prior to the onset of symptoms, pathological amyloid-β (Aβ) plagues and tau protein tangles accumulate in extracellular and intracellular spaces, respectively, leading to neurodegeneration. Among these hallmark pathologies, Aβ is proposed to be the primary etiology by triggering a cascade of pathogenic events, including neuroinflammation, oxidative stress, tau hyperphosphorylation, synaptic/ neuronal dysfunction, and neuronal death (Zhang et al., 2023b). In the early stage of AD, the Aβ accumulation appears in the medial temporal lobe of the brain, such as the hippocampus and entorhinal cortex, which are crucial structures for memory and cognitive functions. Consequently, anti-amyloid therapy is a primary strategy against AD. A recent phase 3 trial reported that lecanemab, a newly developed monoclonal antibody against Aβ protofibrils, greatly decreased the makers of Aβ, tau, neuroinflammation, and neurodegeneration in cerebrospinal fluid, and moderately improved cognitive decline in patients with mild AD after 18-month treatment (van Dyck et al., 2023). The success of lecanemab supports the Aβ hypothesis and suggests Aβ pathology as a target for AD treatment.
    Related Articles | Metrics
    Context-dependency in medicine: how neuronal excitability influences the impact of dopamine on cognition
    Mahboubeh Ahmadi, Nahid Rouhi, Javad Mirnajafi-Zadeh , Bechara J. Saab
    2025, 20 (11):  3225-3226.  doi: 10.4103/NRR.NRR-D-24-00704
    Abstract ( 79 )   PDF (578KB) ( 25 )   Save
    Dopamine, often termed the “feel-good” neurotransmitter, plays a crucial role in myriad physiological and psychological brain processes. While dopamine is primarily associated with pleasure, reward, and motivation, its effects can be quite complex; and this complexity is further compounded when examining how dopamine functions in typical versus disease-affected neural circuits.
    Related Articles | Metrics
    Role of resident memory T cells in neuroinflammatory and neurodegenerative diseases in the central nervous system
    Kimitoshi Kimura
    2025, 20 (11):  3227-3228.  doi: 10.4103/NRR.NRR-D-24-00760
    Abstract ( 48 )   PDF (440KB) ( 17 )   Save
    The immune system has been attracting increasing attention in the field of chronic neurological disorders in the central nervous system (CNS). Autoreactive T cells targeting CNS antigens play a crucial role in the development of various autoimmune diseases, such as multiple sclerosis (MS) and neuromyelitis optica spectrum disorder (NMOSD). Moreover, T cells are now recognized as a pivotal contributor to the pathology of neurodegenerative disorders, including Alzheimer’s disease (AD), Parkinson’s disease (PD), and multiple system atrophy. Among the diverse array of T cell subsets, resident memory T (Trm) cells are suspected to exert a substantial influence on the progression of these debilitating diseases. Trm cells are distinguished by their restricted localization within tissues and their unique transcriptomic signature. This perspective aims to elucidate the current understanding of Trm cells in CNS disorders and to explore future directions in this rapidly evolving field.
    Related Articles | Metrics
    Biochemical dissection of STAT3 signaling in amyotrophic lateral sclerosis
    Savina Apolloni , Nadia D’Ambrosi
    2025, 20 (11):  3229-3230.  doi: 10.4103/NRR.NRR-D-24-00862
    Abstract ( 65 )   PDF (493KB) ( 26 )   Save
    Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease characterized by the loss of upper and lower motor neurons, clinically marked by muscle atrophy and weakness. Although the clinical course is highly variable, the average time from the onset of symptoms to the need for respiratory support or death is 3–5 years. ALS is the most prevalent motor neuron disease in adults, occurring at a rate of 2 per 100,000 individuals and affecting 5.4 per 100,000 individuals overall. At present, there is no established effective treatment for ALS; riluzole (an antagonist of glutamate neurotransmission) and edaravone (a superoxide scavenger) are the only drugs approved for use in the treatment of ALS, and both produce only slight beneficial effects in a limited population of ALS patients. Tofersen, a recently US Food and Drug Administrationapproved antisense oligonucleotide, is only for patients carrying SOD1 mutation (Mead et al., 2023). Despite ALS has a significant genetic component with high heritability, many gene variants responsible for or contributing to the disease remain unidentified. The involvement of numerous cellular processes in ALS progression complicates the identification of causative factors. The complexity of the disease and the extensive genetic and phenotypic diversity among patients hinder the translation of findings from animal models to successful human clinical trials. 5%–10% of ALS cases are classified as familial ALS based on family history. The genes most associated with familial ALS include C9ORF72, TARDBP, FUS, and SOD1. Recent genetic studies have identified various mutations in sporadic ALS cases, suggesting potential genetic contributors to the disease. These risk genes implicate pathways such as apoptosis due to mitochondrial dysfunction, autophagy with disrupted protein homeostasis, inflammation (both peripheral and central), impaired intracellular trafficking, and excitotoxicity. These dysfunctional pathways affect multiple cell types, including upper (corticospinal) and lower (spinal) motor neurons, associated glial and Schwann cells, skeletal muscle fibers and their progenitors, and immune/inflammatory cells.  Among the genes most specifically associated with ALS and muscle atrophy retrieved by BenevolentAI Knowledge Graph, of particular interest is signal transducer and activator of transcription-3 (STAT3), which is among the five genes contributing to common ALS disease mechanisms such as autophagy, apoptosis, cytokine signaling and the FOXO signaling pathway, underscoring the central role of STAT3 signaling in the disease. Furthermore, beyond inflammation, autophagy, apoptosis, and the FOXO pathways, Janus kinase (JAK)/STAT signaling is heavily implicated in the distinctive pathophysiology of ALS. This includes aspects such as TAR DNA-binding protein 43 (TDP-43) protein aggregation, mitochondrial dysfunction, skeletal muscle denervation, and excitotoxicity. These disease processes are widely recognized as major contributors to ALS pathology, particularly mitochondrial dysfunction leading to ATP deficits and protein aggregation, with TDP-43 aggregates present in 95% of patients, along with muscle denervation and atrophy culminating in fatality. The revelation that all these pathways involve JAK/ STAT signaling implies that targeting this pathway could hold therapeutic promise (Richardson et al., 2023).
    Related Articles | Metrics
    Secretome of polarized macrophages: potential for targeting inflammatory dynamics in spinal cord injury
    Andreia Monteiro, Susana Monteiro, Nuno A. Silva
    2025, 20 (11):  3231-3232.  doi: 10.4103/NRR.NRR-D-24-00752
    Abstract ( 40 )   PDF (1705KB) ( 15 )   Save
    Spinal cord injury (SCI) involves an initial traumatic phase, followed by secondary events such as ischemia, increased blood–spinal cord barrier permeability, ionic disruption, glutamate excitotoxicity, and metabolic alterations. A persistent and exaggerated inflammatory response within the spinal cord accompanies these events (Lima et al., 2022). The complexity and interplay of these mechanisms exacerbate the initial injury, leading to a degenerative process at the injury site. While the initial trauma is unavoidable, the secondary injury begins within minutes and can last for months, creating an optimal window for therapeutic intervention.
    Related Articles | Metrics
    The Citron homology domain of MAP4Ks improves outcomes of traumatic brain injury
    Xiaoling Zhong, Wenjiao Tai, Meng-Lu Liu, Shuaipeng Ma, Tianjin Shen, Yuhua Zou, Chun-Li Zhang
    2025, 20 (11):  3233-3244.  doi: 10.4103/NRR.NRR-D-24-00113
    Abstract ( 64 )   PDF (5747KB) ( 107 )   Save
    The mitogen-activated protein kinase kinase kinase kinases (MAP4Ks) signaling pathway plays a pivotal role in axonal regrowth and neuronal degeneration following insults. Whether targeting this pathway is beneficial to brain injury remains unclear. In this study, we showed that adeno-associated virus-delivery of the Citron homology domain of MAP4Ks effectively reduces traumatic brain injury-induced reactive gliosis, tauopathy, lesion size, and behavioral deficits. Pharmacological inhibition of MAP4Ks replicated the ameliorative effects observed with expression of the Citron homology domain. Mechanistically, the Citron homology domain acted as a dominant-negative mutant, impeding MAP4K-mediated phosphorylation of the dishevelled proteins and thereby controlling the Wnt/β-catenin pathway. These findings implicate a therapeutic potential of targeting MAP4Ks to alleviate the detrimental effects of traumatic brain injury.
    Related Articles | Metrics
    Human neural stem cell–derived extracellular vesicles protect against ischemic stroke by activating the PI3K/ AKT/mTOR pathway
    Jiayi Wang, Mengke Zhao, Dong Fu, Meina Wang, Chao Han, Zhongyue Lv, Liang Wang, Jing Liu
    2025, 20 (11):  3245-3258.  doi: 10.4103/NRR.NRR-D-23-01144
    Abstract ( 66 )   PDF (7737KB) ( 9 )   Save
    Human neural stem cell–derived extracellular vesicles exhibit analogous functions to their parental cells, and can thus be used as substitutes for stem cells in stem cell therapy, thereby mitigating the risks of stem cell therapy and advancing the frontiers of stem cell–derived treatments. This lays a foundation for the development of potentially potent new treatment modalities for ischemic stroke. However, the precise mechanisms underlying the efficacy and safety of human neural stem cell–derived extracellular vesicles remain unclear, presenting challenges for clinical translation. To promote the translation of therapy based on human neural stem cell–derived extracellular vesicles from the bench to the bedside, we conducted a comprehensive preclinical study to evaluate the efficacy and safety of human neural stem cell–derived extracellular vesicles in the treatment of ischemic stroke. We found that administration of human neural stem cell– derived extracellular vesicles to an ischemic stroke rat model reduced the volume of cerebral infarction and promoted functional recovery by alleviating neuronal apoptosis. The human neural stem cell–derived extracellular vesicles reduced neuronal apoptosis by enhancing phosphorylation of phosphoinositide 3-kinase, mammalian target of rapamycin, and protein kinase B, and these effects were reversed by treatment with a phosphoinositide 3-kinase inhibitor. These findings suggest that human neural stem cell–derived extracellular vesicles play a neuroprotective role in ischemic stroke through activation of phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin signaling pathway. Finally, we showed that human neural stem cell–derived extracellular vesicles have a good in vivo safety profile. Therefore, human neural stem cell–derived extracellular vesicles are a promising potential agent for the treatment of ischemic stroke.
    Related Articles | Metrics
    Glutamatergic CYLD deletion leads to aberrant excitatory activity in the basolateral amygdala: association with enhanced cued fear expression
    Huidong Li, Faqin Li, Zhaoyi Chen, Erwen Wu, Xiaoxi Dai, Danni Li, Haojie An, Shiyi Zeng, Chunyan Wang, Li Yang, Cheng Long
    2025, 20 (11):  3259-3272.  doi: 10.4103/NRR.NRR-D-24-00054
    Abstract ( 27 )   PDF (12199KB) ( 5 )   Save
    Neuronal activity, synaptic transmission, and molecular changes in the basolateral amygdala play critical roles in fear memory. Cylindromatosis (CYLD) is a deubiquitinase that negatively regulates the nuclear factor kappa-B pathway. CYLD is well studied in non-neuronal cells, yet underinvestigated in the brain, where it is highly expressed. Emerging studies have shown involvement of CYLD in the remodeling of glutamatergic synapses, neuroinflammation, fear memory, and anxiety- and autism-like behaviors. However, the precise role of CYLD in glutamatergic neurons is largely unknown. Here, we first proposed involvement of CYLD in cued fear expression. We next constructed transgenic model mice with specific deletion of Cyld from glutamatergic neurons. Our results show that glutamatergic CYLD deficiency exaggerated the expression of cued fear in only male mice. Further, loss of CYLD in glutamatergic neurons resulted in enhanced neuronal activation, impaired excitatory synaptic transmission, and altered levels of glutamate receptors accompanied by over-activation of microglia in the basolateral amygdala of male mice. Altogether, our study suggests a critical role of glutamatergic CYLD in maintaining normal neuronal, synaptic, and microglial activation. This may contribute, at least in part, to cued fear expression.
    Related Articles | Metrics
    Exosomes originating from neural stem cells undergoing necroptosis participate in cellular communication by inducing TSC2 upregulation of recipient cells following spinal cord injury
    Shiming Li, Jianfeng Li, Guoliang Chen, Tao Lin, Penghui Zhang, Kuileung Tong, Ningning Chen, Shaoyu Liu
    2025, 20 (11):  3273-3286.  doi: 10.4103/NRR.NRR-D-24-00068
    Abstract ( 62 )   PDF (9418KB) ( 11 )   Save
    We previously demonstrated that inhibiting neural stem cells necroptosis enhances functional recovery after spinal cord injury. While exosomes are recognized as playing a pivotal role in neural stem cells exocrine function, their precise function in spinal cord injury remains unclear. To investigate the role of exosomes generated following neural stem cells necroptosis after spinal cord injury, we conducted singlecell RNA sequencing and validated that neural stem cells originate from ependymal cells and undergo necroptosis in response to spinal cord injury. Subsequently, we established an in vitro necroptosis model using neural stem cells isolated from embryonic mice aged 16–17 days and extracted exosomes. The results showed that necroptosis did not significantly impact the fundamental characteristics or number of exosomes. Transcriptome sequencing of exosomes in necroptosis group identified 108 differentially expressed messenger RNAs, 104 long non-coding RNAs, 720 circular RNAs, and 14 microRNAs compared with the control group. Construction of a competing endogenous RNA network identified the following hub genes: tuberous sclerosis 2 (Tsc2), solute carrier family 16 member 3 (Slc16a3), and forkhead box protein P1 (Foxp1). Notably, a significant elevation in TSC2 expression was observed in spinal cord tissues following spinal cord injury. TSC2-positive cells were localized around SRY-box transcription factor 2–positive cells within the injury zone. Furthermore, in vitro analysis revealed increased TSC2 expression in exosomal receptor cells compared with other cells. Further assessment of cellular communication following spinal cord injury showed that Tsc2 was involved in ependymal cellular communication at 1 and 3 days post-injury through the epidermal growth factor and midkine signaling pathways. In addition, Slc16a3 participated in cellular communication in ependymal cells at 7 days post-injury via the vascular endothelial growth factor and macrophage migration inhibitory factor signaling pathways. Collectively, these findings confirm that exosomes derived from neural stem cells undergoing necroptosis play an important role in cellular communication after spinal cord injury and induce TSC2 upregulation in recipient cells.
    Related Articles | Metrics
    Pharmacological targeting cGAS/STING/NF-κB axis by tryptanthrin induces microglia polarization toward M2 phenotype and promotes functional recovery in a mouse model of spinal cord injury
    Ziwei Fan, Mengxian Jia, Jian Zhou, Zhoule Zhu, Yumin Wu, Xiaowu Lin, Yiming Qian, Jiashu Lian, Xin Hua, Jianhong Dong, Zheyu Fang, Yuqing Liu, Sibing Chen, Xiumin Xue, Juanqing Yue, Minyu Zhu, Ying Wang, Zhihui Huang, Honglin Teng
    2025, 20 (11):  3287-3301.  doi: 10.4103/NRR.NRR-D-23-01256
    Abstract ( 88 )   PDF (23920KB) ( 28 )   Save
    The M1/M2 phenotypic shift of microglia after spinal cord injury plays an important role in the regulation of neuroinflammation during the secondary injury phase of spinal cord injury. Regulation of shifting microglia polarization from M1 (neurotoxic and proinflammatory type) to M2 (neuroprotective and anti-inflammatory type) after spinal cord injury appears to be crucial. Tryptanthrin possesses an anti-inflammatory biological function. However, its roles and the underlying molecular mechanisms in spinal cord injury remain unknown. In this study, we found that tryptanthrin inhibited microglia-derived inflammation by promoting polarization to the M2 phenotype in vitro. Tryptanthrin promoted M2 polarization through inactivating the cGAS/STING/NF-κB pathway. Additionally, we found that targeting the cGAS/STING/NF-κB pathway with tryptanthrin shifted microglia from the M1 to M2 phenotype after spinal cord injury, inhibited neuronal loss, and promoted tissue repair and functional recovery in a mouse model of spinal cord injury. Finally, using a conditional co-culture system, we found that microglia treated with tryptanthrin suppressed endoplasmic reticulum stress–related neuronal apoptosis. Taken together, these results suggest that by targeting the cGAS/STING/NF-κB axis, tryptanthrin attenuates microglia–derived neuroinflammation and promotes functional recovery after spinal cord injury through shifting microglia polarization to the M2 phenotype.
    Related Articles | Metrics
    Spatial transcriptomics combined with single-nucleus RNA sequencing reveals glial cell heterogeneity in the human spinal cord
    Yali Chen, Yiyong Wei, Jin Liu, Tao Zhu, Cheng Zhou, Donghang Zhang
    2025, 20 (11):  3302-3316.  doi: 10.4103/NRR.NRR-D-23-01876
    Abstract ( 112 )   PDF (14804KB) ( 14 )   Save
    Glial cells play crucial roles in regulating physiological and pathological functions, including sensation, the response to infection and acute injury, and chronic neurodegenerative disorders. Glial cells include astrocytes, microglia, and oligodendrocytes in the central nervous system, and satellite glial cells and Schwann cells in the peripheral nervous system. Despite the greater understanding of glial cell types and functional heterogeneity achieved through single-cell and single-nucleus RNA sequencing in animal models, few studies have investigated the transcriptomic profiles of glial cells in the human spinal cord. Here, we used high-throughput single-nucleus RNA sequencing and spatial transcriptomics to map the cellular and molecular heterogeneity of astrocytes, microglia, and oligodendrocytes in the human spinal cord. To explore the conservation and divergence across species, we compared these findings with those from mice. In the human spinal cord, astrocytes, microglia, and oligodendrocytes were each divided into six distinct transcriptomic subclusters. In the mouse spinal cord, astrocytes, microglia, and oligodendrocytes were divided into five, four, and five distinct transcriptomic subclusters, respectively. The comparative results revealed substantial heterogeneity in all glial cell types between humans and mice. Additionally, we detected sex differences in gene expression in human spinal cord glial cells. Specifically, in all astrocyte subtypes, the levels of NEAT1 and CHI3L1 were higher in males than in females, whereas the levels of CST3 were lower in males than in females. In all microglial subtypes, all differentially expressed genes were located on the sex chromosomes. In addition to sex-specific gene differences, the levels of MT-ND4, MT2A, MT-ATP6, MT-CO3, MT-ND2, MT-ND3, and MT-CO2 in all spinal cord oligodendrocyte subtypes were higher in females than in males. Collectively, the present dataset extensively characterizes glial cell heterogeneity and offers a valuable resource for exploring the cellular basis of spinal cordrelated illnesses, including chronic pain, amyotrophic lateral sclerosis, and multiple sclerosis.
    Related Articles | Metrics
    The sexually dimorphic expression of glutamate transporters and their implication in pain after spinal cord injury
    Jennifer M. Colón-Mercado, Aranza I. Torrado-Tapias , Iris K. Salgado , José M. Santiago , Samuel E. Ocasio Rivera , Dina P. Bracho-Rincon , Luis H. Pagan Rivera , Jorge D. Miranda
    2025, 20 (11):  3317-3329.  doi: 10.4103/NRR.NRR-D-24-00035
    Abstract ( 35 )   PDF (6846KB) ( 35 )   Save
    In addition to the loss of motor function, ~60% of patients develop pain after spinal cord injury. The cellular-molecular mechanisms are not well understood, but the data suggests that plasticity within the rostral, epicenter, and caudal penumbra of the injury site initiates a cellularmolecular interplay that acts as a rewiring mechanism leading to central neuropathic pain. Sprouting can lead to the formation of new connections triggering abnormal sensory transmission. The excitatory glutamate transporters are responsible for the reuptake of extracellular glutamate which makes them a critical target to prevent neuronal hyperexcitability and excitotoxicity. Our previous studies showed a sexually dimorphic therapeutic window for spinal cord injury after treatment with the selective estrogen receptor modulator tamoxifen. In this study, we investigated the anti-allodynic effects of tamoxifen in male and female rats with spinal cord injury. We hypothesized that tamoxifen exerts anti-allodynic effects by increasing the expression of glutamate transporters, leading to reduced hyperexcitability of the secondary neuron or by decreasing aberrant sprouting. Male and female rats received a moderate contusion to the thoracic spinal cord followed by subcutaneous slow-release treatment of tamoxifen or matrix pellets as a control (placebo). We used von Frey monofilaments and the “up-down method” to evaluate mechanical allodynia. Tamoxifen treatment decreased allodynia only in female rats with spinal cord injury revealing a sexdependent effect. The expression profile of glutamatergic transporters (excitatory amino acid transporter 1/glutamate aspartate transporter and excitatory amino acid transporter 2/glutamate transporter-1) revealed a sexual dimorphism in the rostral, epicenter, and caudal areas of the spinal cord with a pattern of expression primarily on astrocytes. Female rodents showed a significantly higher level of excitatory amino acid transporter-1 expression while male rodents showed increased excitatory amino acid transporter-2 expression compared with female rodents. Analyses of peptidergic (calcitonin gene-related peptide-α) and non-peptidergic (isolectin B4) fibers outgrowth in the dorsal horn after spinal cord injury showed an increased calcitonin gene-related peptide-α/ isolectin B4 ratio in comparison with sham, suggesting increased receptive fields in the dorsal horn. Although the behavioral assay shows decreased allodynia in tamoxifen-treated female rats, this was not associated with overexpression of glutamate transporters or alterations in the dorsal horn laminae fibers at 28 days post-injury. Our findings provide new evidence of the sexually dimorphic expression of glutamate transporters in the spinal cord. The dimorphic expression revealed in this study provides a therapeutic opportunity for treating chronic pain, an area with a critical need for treatment.
    Related Articles | Metrics
    The compound (E)-2-(3,4-dihydroxystyryl)-3-hydroxy4H-pyran-4-one alleviates neuroinflammation and cognitive impairment in a mouse model of Alzheimer’s disease
    Xueyan Liu, Wei Wu, Xuejuan Li, Chengyan Wang, Ke Chai, Fanru Yuan, Huijuan Zheng, Yuxing Yao, Chenlu Li, Zu-Cheng Ye, Daijun Zha
    2025, 20 (11):  3330-3344.  doi: 10.4103/NRR.NRR-D-23-01890
    Abstract ( 42 )   PDF (9912KB) ( 6 )   Save
    Previous studies have shown that the compound (E)-2-(3,4-dihydroxystyryl)-3-hydroxy-4H-pyran-4-one (D30), a pyromeconic acid derivative, possesses antioxidant and anti-inflammatory properties, inhibits amyloid-β aggregation, and alleviates scopolamine-induced cognitive impairment, similar to the phase III clinical drug resveratrol. In this study, we established a mouse model of Alzheimer’s disease via intracerebroventricular injection of fibrillar amyloid-β to investigate the effect of D30 on fibrillar amyloid-β–induced neuropathology. Our results showed that D30 alleviated fibrillar amyloid-β–induced cognitive impairment, promoted fibrillar amyloid-β clearance from the hippocampus and cortex, suppressed oxidative stress, and inhibited activation of microglia and astrocytes. D30 also reversed the fibrillar amyloid-β–induced loss of dendritic spines and synaptic protein expression. Notably, we demonstrated that exogenous fibrillar amyloid-β introduced by intracerebroventricular injection greatly increased galectin-3 expression levels in the brain, and this increase was blocked by D30. Considering the role of D30 in clearing amyloid-β, inhibiting neuroinflammation, protecting synapses, and improving cognition, this study highlights the potential of galectin-3 as a promising treatment target for patients with Alzheimer’s disease.
    Related Articles | Metrics