Pub Date : 2025-02-05DOI: 10.1515/revneuro-2024-0156
Farhad Mashayekhi, Zivar Salehi
There has been a significant amount of attention directed towards understanding brain development, shedding light on the underlying mechanisms. The proliferation and differentiation of brain stem cells have been a key focus. The process of neurolation occurs during the early stages of embryonic development, leading to the formation of the neural tube, a hollow nerve cord that gives rise to the central nervous system (CNS). There is a growing emphasis on the fluid-filled space inside the developing CNS and the potential role of cerebrospinal fluid (CSF) in brain development. The flow of CSF near the germinal epithelium significantly impacts the proliferation of cells in the cerebral cortex. CSF provides crucial support to the germinal epithelium, influencing the growth and differentiation of neural stem cells. It achieves this by releasing growth factors, cytokines, and morphogens that control the proliferation, survival, and migration of neuroepithelium. During development, the concentration of proteins in the CSF is notably higher compared to that in adults. Studies have indicated that removing CSF from the brain's ventricles during development causes an increase in neural cell deaths and a reduction in neural cell proliferation, ultimately leading to a thinner cerebral cortex. Additionally, many researches demonstrate that the composition of the CSF is essential for maintaining germinal matrix function and output, highlighting the critical role of CSF in brain development. It is concluded that CSF impacts the proliferation and differentiation of neural stem cells, which in turn plays a pivotal role in brain development.
{"title":"The essential role of cerebrospinal fluid in the brain; a comprehensive review.","authors":"Farhad Mashayekhi, Zivar Salehi","doi":"10.1515/revneuro-2024-0156","DOIUrl":"https://doi.org/10.1515/revneuro-2024-0156","url":null,"abstract":"<p><p>There has been a significant amount of attention directed towards understanding brain development, shedding light on the underlying mechanisms. The proliferation and differentiation of brain stem cells have been a key focus. The process of neurolation occurs during the early stages of embryonic development, leading to the formation of the neural tube, a hollow nerve cord that gives rise to the central nervous system (CNS). There is a growing emphasis on the fluid-filled space inside the developing CNS and the potential role of cerebrospinal fluid (CSF) in brain development. The flow of CSF near the germinal epithelium significantly impacts the proliferation of cells in the cerebral cortex. CSF provides crucial support to the germinal epithelium, influencing the growth and differentiation of neural stem cells. It achieves this by releasing growth factors, cytokines, and morphogens that control the proliferation, survival, and migration of neuroepithelium. During development, the concentration of proteins in the CSF is notably higher compared to that in adults. Studies have indicated that removing CSF from the brain's ventricles during development causes an increase in neural cell deaths and a reduction in neural cell proliferation, ultimately leading to a thinner cerebral cortex. Additionally, many researches demonstrate that the composition of the CSF is essential for maintaining germinal matrix function and output, highlighting the critical role of CSF in brain development. It is concluded that CSF impacts the proliferation and differentiation of neural stem cells, which in turn plays a pivotal role in brain development.</p>","PeriodicalId":49623,"journal":{"name":"Reviews in the Neurosciences","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-02-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143123135","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-02-05DOI: 10.1515/revneuro-2024-0165
Aurimas Mockevičius, Inga Griškova-Bulanova
Auditory steady-state response (ASSR) is a robust method to probe gamma (>30 Hz) activity in a controlled manner. While typically the magnitude and the phase synchronization over stimulus repetitions of ASSR is assessed, other measures are being investigated. One of them is phase-amplitude coupling (PAC), which reflects the interactions between lower frequency phase and higher frequency amplitude. Considering that the number of studies assessing PAC during auditory steady-state stimulation has grown recently, in the present work, we aimed to perform a comprehensive overview of PAC methodological approaches in ASSR studies. We sought to evaluate the studies according to PAC analysis issues emphasized in empirical and theoretical PAC studies. Our work showed considerable variability in the methodology among the reviewed studies. Furthermore, the reviewed works address methodological issues and confounding factors of PAC relatively poorly and are characterized by insufficient descriptions of the applied approaches. Our review shows that systematic research of PAC in the context of ASSR is imperative in order to properly evaluate the presence of PAC during the auditory steady-state stimulation.
{"title":"Phase-amplitude coupling during auditory steady-state stimulation: a methodological review.","authors":"Aurimas Mockevičius, Inga Griškova-Bulanova","doi":"10.1515/revneuro-2024-0165","DOIUrl":"https://doi.org/10.1515/revneuro-2024-0165","url":null,"abstract":"<p><p>Auditory steady-state response (ASSR) is a robust method to probe gamma (>30 Hz) activity in a controlled manner. While typically the magnitude and the phase synchronization over stimulus repetitions of ASSR is assessed, other measures are being investigated. One of them is phase-amplitude coupling (PAC), which reflects the interactions between lower frequency phase and higher frequency amplitude. Considering that the number of studies assessing PAC during auditory steady-state stimulation has grown recently, in the present work, we aimed to perform a comprehensive overview of PAC methodological approaches in ASSR studies. We sought to evaluate the studies according to PAC analysis issues emphasized in empirical and theoretical PAC studies. Our work showed considerable variability in the methodology among the reviewed studies. Furthermore, the reviewed works address methodological issues and confounding factors of PAC relatively poorly and are characterized by insufficient descriptions of the applied approaches. Our review shows that systematic research of PAC in the context of ASSR is imperative in order to properly evaluate the presence of PAC during the auditory steady-state stimulation.</p>","PeriodicalId":49623,"journal":{"name":"Reviews in the Neurosciences","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-02-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143123134","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-24DOI: 10.1515/revneuro-2024-0153
Pantea Allami, Niloufar Yazdanpanah, Nima Rezaei
Fast spiking parvalbumin (PV) interneuron is an inhibitory gamma-aminobutyric acid (GABA)ergic interneuron diffused in different brain networks, including the cortex and hippocampus. As a key component of brain networks, PV interneurons collaborate in fundamental brain functions such as learning and memory by regulating excitation and inhibition (E/I) balance and generating gamma oscillations. The unique characteristics of PV interneurons, like their high metabolic demands and long branching axons, make them too vulnerable to stressors. Neuroinflammation is one of the most significant stressors that have an adverse, long-lasting impact on PV interneurons. Neuroinflammation affects PV interneurons through specialized inflammatory pathways triggered by cytokines such as tumor necrosis factor (TNF) and interleukin 6 (IL-6). The crucial cells in neuroinflammation, microglia, also play a significant role. The destructive effect of inflammation on PV interneurons can have comprehensive effects and cause neurological disorders such as schizophrenia, Alzheimer's disease (AD), autism spectrum disorder (ASD), and bipolar disorder. In this article, we provide a comprehensive review of mechanisms in which neuroinflammation leads to PV interneuron hypofunction in these diseases. The integrated knowledge about the role of PV interneurons in cognitive networks of the brain and mechanisms involved in PV interneuron impairment in the pathology of these diseases can help us with better therapeutic interventions.
{"title":"The role of neuroinflammation in PV interneuron impairments in brain networks; implications for cognitive disorders.","authors":"Pantea Allami, Niloufar Yazdanpanah, Nima Rezaei","doi":"10.1515/revneuro-2024-0153","DOIUrl":"https://doi.org/10.1515/revneuro-2024-0153","url":null,"abstract":"<p><p>Fast spiking parvalbumin (PV) interneuron is an inhibitory gamma-aminobutyric acid (GABA)ergic interneuron diffused in different brain networks, including the cortex and hippocampus. As a key component of brain networks, PV interneurons collaborate in fundamental brain functions such as learning and memory by regulating excitation and inhibition (E/I) balance and generating gamma oscillations. The unique characteristics of PV interneurons, like their high metabolic demands and long branching axons, make them too vulnerable to stressors. Neuroinflammation is one of the most significant stressors that have an adverse, long-lasting impact on PV interneurons. Neuroinflammation affects PV interneurons through specialized inflammatory pathways triggered by cytokines such as tumor necrosis factor (TNF) and interleukin 6 (IL-6). The crucial cells in neuroinflammation, microglia, also play a significant role. The destructive effect of inflammation on PV interneurons can have comprehensive effects and cause neurological disorders such as schizophrenia, Alzheimer's disease (AD), autism spectrum disorder (ASD), and bipolar disorder. In this article, we provide a comprehensive review of mechanisms in which neuroinflammation leads to PV interneuron hypofunction in these diseases. The integrated knowledge about the role of PV interneurons in cognitive networks of the brain and mechanisms involved in PV interneuron impairment in the pathology of these diseases can help us with better therapeutic interventions.</p>","PeriodicalId":49623,"journal":{"name":"Reviews in the Neurosciences","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-01-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143025440","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-21DOI: 10.1515/revneuro-2024-0147
Nan Wang, Shanshan Zhu, Shuyang Chen, Ju Zou, Peng Zeng, Sijie Tan
Depression is a common mental disorder characterized by a high prevalence and significant adverse effects, making the searching for effective interventions an urgent priority. In recent years, physical activity (PA) has increasingly been recognized as a standard adjunctive treatment for mental disorders owing to its low cost, easy application, and high efficiency. Epidemiological data shows positive preventive and therapeutic effects of PA on mental illnesses such as depression. This article systematically describes the prophylactic and therapeutic effects of PA on depression and its biological basis. A comprehensive literature analysis reveals that PA significantly improves depressive symptoms by upregulating the expression of "exerkines" such as irisin, adiponectin, and BDNF to positively impacting neuropsychiatric conditions. In particular, lactate could also play a critical role in the ameliorating effects of PA on depression due to the findings about protein lactylation as a novel protein post-transcriptional modification. The literature also suggests that in terms of brain structure, PA may improve hippocampal volume, basal ganglia (neostriatum, caudate-crustal nucleus) and PFC density in patients with MDD. In summary, this study elucidates the multifaceted positive effects of PA on depression and its potential biological mechanisms with a particular emphasis on the roles of various exerkines. Future research may further investigate the effects of different types, intensities, and durations of PA on depression, as well as how to better integrate PA interventions into existing treatment strategies to achieve optimal outcomes in mental health interventions.
{"title":"Neurological mechanism-based analysis of the role and characteristics of physical activity in the improvement of depressive symptoms.","authors":"Nan Wang, Shanshan Zhu, Shuyang Chen, Ju Zou, Peng Zeng, Sijie Tan","doi":"10.1515/revneuro-2024-0147","DOIUrl":"https://doi.org/10.1515/revneuro-2024-0147","url":null,"abstract":"<p><p>Depression is a common mental disorder characterized by a high prevalence and significant adverse effects, making the searching for effective interventions an urgent priority. In recent years, physical activity (PA) has increasingly been recognized as a standard adjunctive treatment for mental disorders owing to its low cost, easy application, and high efficiency. Epidemiological data shows positive preventive and therapeutic effects of PA on mental illnesses such as depression. This article systematically describes the prophylactic and therapeutic effects of PA on depression and its biological basis. A comprehensive literature analysis reveals that PA significantly improves depressive symptoms by upregulating the expression of \"exerkines\" such as irisin, adiponectin, and BDNF to positively impacting neuropsychiatric conditions. In particular, lactate could also play a critical role in the ameliorating effects of PA on depression due to the findings about protein lactylation as a novel protein post-transcriptional modification. The literature also suggests that in terms of brain structure, PA may improve hippocampal volume, basal ganglia (neostriatum, caudate-crustal nucleus) and PFC density in patients with MDD. In summary, this study elucidates the multifaceted positive effects of PA on depression and its potential biological mechanisms with a particular emphasis on the roles of various exerkines. Future research may further investigate the effects of different types, intensities, and durations of PA on depression, as well as how to better integrate PA interventions into existing treatment strategies to achieve optimal outcomes in mental health interventions.</p>","PeriodicalId":49623,"journal":{"name":"Reviews in the Neurosciences","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-01-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143015172","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-21DOI: 10.1515/revneuro-2024-0086
Razieh Razavi, Ghazal Khajouei, Fatemeh Divsalar, Elmuez Dawi, Mahnaz Amiri
Essentially, the blood-brain barrier (BBB) serves as a line of demarcation between neural tissues and the bloodstream. A unique and protective characteristic of the blood-brain barrier is its ability to maintain cerebral homeostasis by regulating the flux of molecules and ions. The inability to uphold proper functioning in any of these constituents leads to the disruption of this specialized multicellular arrangement, consequently fostering neuroinflammation and neurodegeneration. Recent advancements in nanomedicine have been regarded as a promising avenue for improving the delivery of drugs to the central nervous system in the modern era. A major benefit of this innovation is that it allows drugs to accumulate selectively within the cerebral area by circumventing the blood-brain barrier. Although brain-targeted nanomedicines have demonstrated impressive achievements, certain limitations in targeting specificity still exist. In this examination, we scrutinize the distinctive physical and chemical attributes of nanoparticles (NPs) contributing to their facilitation in BBB traversal. We explore the various mechanisms governing NP passage over the BBB, encompassing paracellular conveyance, mediated transport, as well as adsorptive- and receptor-mediated transcytosis. The therapeutic success of NPs for the treatment of brain tumors has been extensively investigated through the use of various categories of NPs. Among these are polymeric nanoparticles, liposomes, solid lipid nanoparticles, dendrimers, metallic nanoparticles, quantum dots, and nanogels. The potential utility of nanoparticles goes beyond their ability to transport pharmaceuticals. They can serve as adept imaging contrast agents, capable of being linked with imaging probes. This will facilitate tumor visualization, delineate lesion boundaries and margins, and monitor drug delivery and treatment response. Versatile nanoparticles can be engineered to effectively target neoplastic lesions, serving dual roles in diagnostic imaging and therapeutic interventions. Subsequently, this discourse explores the constraints associated with nanoparticles in the context of treating brain tumors.
{"title":"Recent advances on brain drug delivery via nanoparticles: alternative future materials for neuroscience applications; a review.","authors":"Razieh Razavi, Ghazal Khajouei, Fatemeh Divsalar, Elmuez Dawi, Mahnaz Amiri","doi":"10.1515/revneuro-2024-0086","DOIUrl":"https://doi.org/10.1515/revneuro-2024-0086","url":null,"abstract":"<p><p>Essentially, the blood-brain barrier (BBB) serves as a line of demarcation between neural tissues and the bloodstream. A unique and protective characteristic of the blood-brain barrier is its ability to maintain cerebral homeostasis by regulating the flux of molecules and ions. The inability to uphold proper functioning in any of these constituents leads to the disruption of this specialized multicellular arrangement, consequently fostering neuroinflammation and neurodegeneration. Recent advancements in nanomedicine have been regarded as a promising avenue for improving the delivery of drugs to the central nervous system in the modern era. A major benefit of this innovation is that it allows drugs to accumulate selectively within the cerebral area by circumventing the blood-brain barrier. Although brain-targeted nanomedicines have demonstrated impressive achievements, certain limitations in targeting specificity still exist. In this examination, we scrutinize the distinctive physical and chemical attributes of nanoparticles (NPs) contributing to their facilitation in BBB traversal. We explore the various mechanisms governing NP passage over the BBB, encompassing paracellular conveyance, mediated transport, as well as adsorptive- and receptor-mediated transcytosis. The therapeutic success of NPs for the treatment of brain tumors has been extensively investigated through the use of various categories of NPs. Among these are polymeric nanoparticles, liposomes, solid lipid nanoparticles, dendrimers, metallic nanoparticles, quantum dots, and nanogels. The potential utility of nanoparticles goes beyond their ability to transport pharmaceuticals. They can serve as adept imaging contrast agents, capable of being linked with imaging probes. This will facilitate tumor visualization, delineate lesion boundaries and margins, and monitor drug delivery and treatment response. Versatile nanoparticles can be engineered to effectively target neoplastic lesions, serving dual roles in diagnostic imaging and therapeutic interventions. Subsequently, this discourse explores the constraints associated with nanoparticles in the context of treating brain tumors.</p>","PeriodicalId":49623,"journal":{"name":"Reviews in the Neurosciences","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-01-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143015173","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-21DOI: 10.1515/revneuro-2024-0113
Rad Ghannadzadeh Kermani Pour, Sara Kamali Zounouzi, Melina Farshbafnadi, Nima Rezaei
Recently, researchers have been interested in the potential connection between gut microbiota composition and various neuropsychological disorders. Dementia significantly affects the socioeconomics of families. Gut microbiota is considered as a probable factor in its pathogenesis. Multiple bacterial metabolites such as short-chain fatty acids, lipopolysaccharides, and various neurotransmitters that are responsible for the incidence and progression of dementia can be produced by gut microbiota. Various bacterial species such as Bifidobacterium breve, Akkermansia muciniphila, Streptococcus thermophilus, Escherichia coli, Blautia hydrogenotrophica, etc. are implicated in the pathogenesis of dementia. Gut microbiota can be a great target for imitating or inhibiting their metabolites as an adjunctive therapy based on their role in its pathogenesis. Therefore, some diets can prevent or decelerate dementia by altering the gut microbiota composition. Moreover, probiotics can modulate gut microbiota composition by increasing beneficial bacteria and reducing detrimental species. These therapeutic modalities are considered novel methods that are probably safe and effective. They can enhance the efficacy of traditional medications and improve cognitive function.
{"title":"The interplay between gut microbiota composition and dementia.","authors":"Rad Ghannadzadeh Kermani Pour, Sara Kamali Zounouzi, Melina Farshbafnadi, Nima Rezaei","doi":"10.1515/revneuro-2024-0113","DOIUrl":"https://doi.org/10.1515/revneuro-2024-0113","url":null,"abstract":"<p><p>Recently, researchers have been interested in the potential connection between gut microbiota composition and various neuropsychological disorders. Dementia significantly affects the socioeconomics of families. Gut microbiota is considered as a probable factor in its pathogenesis. Multiple bacterial metabolites such as short-chain fatty acids, lipopolysaccharides, and various neurotransmitters that are responsible for the incidence and progression of dementia can be produced by gut microbiota. Various bacterial species such as <i>Bifidobacterium breve</i>, <i>Akkermansia muciniphila</i>, <i>Streptococcus thermophilus</i>, <i>Escherichia coli</i>, <i>Blautia hydrogenotrophica</i>, etc. are implicated in the pathogenesis of dementia. Gut microbiota can be a great target for imitating or inhibiting their metabolites as an adjunctive therapy based on their role in its pathogenesis. Therefore, some diets can prevent or decelerate dementia by altering the gut microbiota composition. Moreover, probiotics can modulate gut microbiota composition by increasing beneficial bacteria and reducing detrimental species. These therapeutic modalities are considered novel methods that are probably safe and effective. They can enhance the efficacy of traditional medications and improve cognitive function.</p>","PeriodicalId":49623,"journal":{"name":"Reviews in the Neurosciences","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-01-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143015175","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-21DOI: 10.1515/revneuro-2024-0142
Hailiang Cui, Zhonghua Li, Hongyu Sun, Wanlin Zhao, He Ma, Li Hao, Zhenqiang Zhang, Christian Hölscher, Dongrui Ma, Zijuan Zhang
Cholecystokinin (CCK) is a major neuropeptide in the brain that functions as a neurotransmitter, hormone, and growth factor. The peptide and its receptors are widely expressed in the brain. CCK signaling modulates synaptic plasticity and can improve or impair memory formation, depending on the brain areas studies and the receptor subtype activated. Studies have shown in a series of animal models of neurodegenerative diseases that CCK receptor agonists show neuroprotective effects and can effectively alleviate oxidative stress, alleviate chronic inflammation of the central nervous system, improve neuronal synaptic plasticity, prevent neuronal loss, and improve cognitive dysfunction in Alzheimer's disease (AD) model mice and motor activity in animal models of Parkinson's disease. In addition, CCK plays important roles in the amygdala to regulate anxiety and depressive states. Activation of interneurons or inhibition of excitatory neurons can improve anxiety levels. This review summarizes the effects on memory formation and synaptic plasticity, the neuroprotective effects of cholecystokinin and its analogs in neurological diseases such as Alzheimer and Parkinson's disease, and the effects on anxiety and neuronal activity in the amygdala.
{"title":"The neuroprotective effects of cholecystokinin in the brain: antioxidant, anti-inflammatory, cognition, and synaptic plasticity.","authors":"Hailiang Cui, Zhonghua Li, Hongyu Sun, Wanlin Zhao, He Ma, Li Hao, Zhenqiang Zhang, Christian Hölscher, Dongrui Ma, Zijuan Zhang","doi":"10.1515/revneuro-2024-0142","DOIUrl":"https://doi.org/10.1515/revneuro-2024-0142","url":null,"abstract":"<p><p>Cholecystokinin (CCK) is a major neuropeptide in the brain that functions as a neurotransmitter, hormone, and growth factor. The peptide and its receptors are widely expressed in the brain. CCK signaling modulates synaptic plasticity and can improve or impair memory formation, depending on the brain areas studies and the receptor subtype activated. Studies have shown in a series of animal models of neurodegenerative diseases that CCK receptor agonists show neuroprotective effects and can effectively alleviate oxidative stress, alleviate chronic inflammation of the central nervous system, improve neuronal synaptic plasticity, prevent neuronal loss, and improve cognitive dysfunction in Alzheimer's disease (AD) model mice and motor activity in animal models of Parkinson's disease. In addition, CCK plays important roles in the amygdala to regulate anxiety and depressive states. Activation of interneurons or inhibition of excitatory neurons can improve anxiety levels. This review summarizes the effects on memory formation and synaptic plasticity, the neuroprotective effects of cholecystokinin and its analogs in neurological diseases such as Alzheimer and Parkinson's disease, and the effects on anxiety and neuronal activity in the amygdala.</p>","PeriodicalId":49623,"journal":{"name":"Reviews in the Neurosciences","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-01-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143015176","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-01-21DOI: 10.1515/revneuro-2024-0125
Nooshin Goudarzi, Zahra Taheri, Amir Mohammad Nezhad Salari, Kimia Kazemzadeh, Abbas Tafakhori
The recognition and classification of facial expressions using artificial intelligence (AI) presents a promising avenue for early detection and monitoring of neurodegenerative disorders. This narrative review critically examines the current state of AI-driven facial expression analysis in the context of neurodegenerative diseases, such as Alzheimer's and Parkinson's. We discuss the potential of AI techniques, including deep learning and computer vision, to accurately interpret and categorize subtle changes in facial expressions associated with these pathological conditions. Furthermore, we explore the role of facial expression recognition as a noninvasive, cost-effective tool for screening, disease progression tracking, and personalized intervention in neurodegenerative disorders. The review also addresses the challenges, ethical considerations, and future prospects of integrating AI-based facial expression analysis into clinical practice for early intervention and improved quality of life for individuals at risk of or affected by neurodegenerative diseases.
{"title":"Recognition and classification of facial expression using artificial intelligence as a key of early detection in neurological disorders.","authors":"Nooshin Goudarzi, Zahra Taheri, Amir Mohammad Nezhad Salari, Kimia Kazemzadeh, Abbas Tafakhori","doi":"10.1515/revneuro-2024-0125","DOIUrl":"https://doi.org/10.1515/revneuro-2024-0125","url":null,"abstract":"<p><p>The recognition and classification of facial expressions using artificial intelligence (AI) presents a promising avenue for early detection and monitoring of neurodegenerative disorders. This narrative review critically examines the current state of AI-driven facial expression analysis in the context of neurodegenerative diseases, such as Alzheimer's and Parkinson's. We discuss the potential of AI techniques, including deep learning and computer vision, to accurately interpret and categorize subtle changes in facial expressions associated with these pathological conditions. Furthermore, we explore the role of facial expression recognition as a noninvasive, cost-effective tool for screening, disease progression tracking, and personalized intervention in neurodegenerative disorders. The review also addresses the challenges, ethical considerations, and future prospects of integrating AI-based facial expression analysis into clinical practice for early intervention and improved quality of life for individuals at risk of or affected by neurodegenerative diseases.</p>","PeriodicalId":49623,"journal":{"name":"Reviews in the Neurosciences","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2025-01-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143015174","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-30DOI: 10.1515/revneuro-2024-0097
Faezeh Firuzpour, Kiarash Saleki, Cena Aram, Nima Rezaei
Glioblastoma multiforme (GBM) is the most fatal brain tumor with a poor prognosis with current treatments, mainly because of intrinsic resistance processes. GBM is also referred to as grade 4 astrocytoma, that makes up about 15.4 % of brain cancers globally as well as 60-75 % of astrocytoma. The most prevalent therapeutic choices for GBM comprise surgery in combination with radiotherapy and chemotherapy, providing patients with an average survival of 6-14 months. Nanocarriers provide various benefits such as enhanced drug solubility, biocompatibility, targeted activity, as well as minimized side effects. In addition, GBM treatment comes with several challenges such as the presence of the blood-brain barrier (BBB), blood-brain tumor barrier (BBTB), overexpressed efflux pumps, infiltration, invasion, drug resistance, as well as immune escape due to tumor microenvironment (TME) and cancer stem cells (CSC). Recent research has focused on nanocarriers due to their ability to self-assemble, improve bioavailability, provide controlled release, and penetrate the BBB. These nano-based components could potentially enhance drug accumulation in brain tumor tissues and reduce systemic toxicity, making them a compelling solution for GBM therapy. This review captures the complexities associated with multi-functional nano drug delivery systems (NDDS) in crossing the blood-brain barrier (BBB) and targeting cancer cells. In addition, it presents a succinct overview of various types of targeted multi-functional nano drug delivery system (NDDS) which has exhibited promising value for improving drug delivery to the brain.
{"title":"Nanocarriers in glioblastoma treatment: a neuroimmunological perspective.","authors":"Faezeh Firuzpour, Kiarash Saleki, Cena Aram, Nima Rezaei","doi":"10.1515/revneuro-2024-0097","DOIUrl":"https://doi.org/10.1515/revneuro-2024-0097","url":null,"abstract":"<p><p>Glioblastoma multiforme (GBM) is the most fatal brain tumor with a poor prognosis with current treatments, mainly because of intrinsic resistance processes. GBM is also referred to as grade 4 astrocytoma, that makes up about 15.4 % of brain cancers globally as well as 60-75 % of astrocytoma. The most prevalent therapeutic choices for GBM comprise surgery in combination with radiotherapy and chemotherapy, providing patients with an average survival of 6-14 months. Nanocarriers provide various benefits such as enhanced drug solubility, biocompatibility, targeted activity, as well as minimized side effects. In addition, GBM treatment comes with several challenges such as the presence of the blood-brain barrier (BBB), blood-brain tumor barrier (BBTB), overexpressed efflux pumps, infiltration, invasion, drug resistance, as well as immune escape due to tumor microenvironment (TME) and cancer stem cells (CSC). Recent research has focused on nanocarriers due to their ability to self-assemble, improve bioavailability, provide controlled release, and penetrate the BBB. These nano-based components could potentially enhance drug accumulation in brain tumor tissues and reduce systemic toxicity, making them a compelling solution for GBM therapy. This review captures the complexities associated with multi-functional nano drug delivery systems (NDDS) in crossing the blood-brain barrier (BBB) and targeting cancer cells. In addition, it presents a succinct overview of various types of targeted multi-functional nano drug delivery system (NDDS) which has exhibited promising value for improving drug delivery to the brain.</p>","PeriodicalId":49623,"journal":{"name":"Reviews in the Neurosciences","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2024-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142903886","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2024-12-18DOI: 10.1515/revneuro-2024-0128
Arian Daneshpour, Zoha Shaka, Nima Rezaei
Stroke is a severe neurological disease and a major worldwide issue, mostly manifesting as ischemic stroke (IS). In order to create effective treatments for IS, it is imperative to fully understand the underlying pathologies, as the existing therapeutic choices are inadequate. Recent investigations have shown the complex relationships between several programmed cell death (PCD) pathways, including necroptosis, ferroptosis, and pyroptosis, and their correlation with immune responses during IS. However, this relationship is still unclear. To address this gap, this review study explored the cellular interactions in the immune microenvironment of IS. Then, to validate prior findings and uncover biomarkers, the study investigated bioinformatics studies. Several pathways, including nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), Toll-like receptor 4 (TLR4), and receptor-interacting protein kinase (RIPK), were involved in PCD-immune interactions. The bioinformatics studies reported key biomarkers such as glutathione peroxidase 4 (GPX4), NOD-like receptor family pyrin domain containing 3 (NLRP3), gasdermin D (GSDMD), and TLR4, which have important implications in ferroptosis, cuproptosis, pyroptosis, and necroptosis respectively. These biomarkers were associated with PCD mechanisms such as oxidative stress and inflammatory reactions. The immune infiltration analysis consistently revealed a significant correlation between PCD pathways and detrimental immune cells, such as neutrophils and γδ T cells. Conversely, M2 macrophages and T helper cells showed protective effects. In conclusion, considering the intricate network of interactions between immune responses and PCD pathways, this study emphasized the necessity of a paradigm shift in therapeutic approaches to address the injuries that are related to this complex network.
脑卒中是一种严重的神经系统疾病,也是世界性的重大问题,主要表现为缺血性脑卒中(IS)。由于现有的治疗方法并不完善,要想有效治疗缺血性中风,就必须充分了解其潜在的病理机制。最近的研究表明,包括坏死、铁凋亡和热凋亡在内的几种程序性细胞死亡(PCD)通路之间存在复杂的关系,它们与 IS 期间的免疫反应也有关联。然而,这种关系仍不清楚。为了填补这一空白,本综述研究探讨了IS免疫微环境中的细胞相互作用。然后,为了验证之前的研究结果并发现生物标志物,该研究对生物信息学研究进行了调查。包括核因子卡巴轻链-活化B细胞增强因子(NF-κB)、Toll样受体4(TLR4)和受体相互作用蛋白激酶(RIPK)在内的几种通路参与了PCD与免疫的相互作用。生物信息学研究报告了一些关键的生物标志物,如谷胱甘肽过氧化物酶4(GPX4)、NOD样受体家族含吡林结构域3(NLRP3)、gasdermin D(GSDMD)和TLR4,它们分别在铁变性、杯突变性、热变性和坏死中具有重要影响。这些生物标志物与氧化应激和炎症反应等 PCD 机制有关。免疫浸润分析一致表明,PCD 途径与中性粒细胞和 γδ T 细胞等有害免疫细胞之间存在显著相关性。相反,M2 巨噬细胞和 T 辅助细胞则具有保护作用。总之,考虑到免疫反应和 PCD 通路之间错综复杂的相互作用网络,本研究强调有必要转变治疗方法的范式,以解决与这一复杂网络相关的损伤问题。
{"title":"Interplay of cell death pathways and immune responses in ischemic stroke: insights into novel biomarkers.","authors":"Arian Daneshpour, Zoha Shaka, Nima Rezaei","doi":"10.1515/revneuro-2024-0128","DOIUrl":"https://doi.org/10.1515/revneuro-2024-0128","url":null,"abstract":"<p><p>Stroke is a severe neurological disease and a major worldwide issue, mostly manifesting as ischemic stroke (IS). In order to create effective treatments for IS, it is imperative to fully understand the underlying pathologies, as the existing therapeutic choices are inadequate. Recent investigations have shown the complex relationships between several programmed cell death (PCD) pathways, including necroptosis, ferroptosis, and pyroptosis, and their correlation with immune responses during IS. However, this relationship is still unclear. To address this gap, this review study explored the cellular interactions in the immune microenvironment of IS. Then, to validate prior findings and uncover biomarkers, the study investigated bioinformatics studies. Several pathways, including nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), Toll-like receptor 4 (TLR4), and receptor-interacting protein kinase (RIPK), were involved in PCD-immune interactions. The bioinformatics studies reported key biomarkers such as glutathione peroxidase 4 (GPX4), NOD-like receptor family pyrin domain containing 3 (NLRP3), gasdermin D (GSDMD), and TLR4, which have important implications in ferroptosis, cuproptosis, pyroptosis, and necroptosis respectively. These biomarkers were associated with PCD mechanisms such as oxidative stress and inflammatory reactions. The immune infiltration analysis consistently revealed a significant correlation between PCD pathways and detrimental immune cells, such as neutrophils and γδ T cells. Conversely, M2 macrophages and T helper cells showed protective effects. In conclusion, considering the intricate network of interactions between immune responses and PCD pathways, this study emphasized the necessity of a paradigm shift in therapeutic approaches to address the injuries that are related to this complex network.</p>","PeriodicalId":49623,"journal":{"name":"Reviews in the Neurosciences","volume":" ","pages":""},"PeriodicalIF":3.4,"publicationDate":"2024-12-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142840134","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}