首页 > 最新文献

Progress in brain research最新文献

英文 中文
Stress in specific population: Cognitive decline in aging, occupational challenges, strategies for medical professionals. 特定人群的压力:老年认知能力下降,职业挑战,医疗专业人员的策略。
4区 医学 Q3 Neuroscience Pub Date : 2025-01-01 Epub Date: 2025-02-10 DOI: 10.1016/bs.pbr.2025.01.017
Gargi Gautam, Nasrollah Moradikor

Understanding cognitive decline and its contributing causes, such as stress, which presents differently in different groups, is crucial given the aging population's rapid growth. This chapter looks at how stress affects older persons' cognitive decline, with a particular emphasis on the difficulties faced by medical professionals in their line of work and how to lessen the consequences. The severity and course of cognitive decline differ from person to person and are impacted by factors such as lifestyle, medical history, and stress at work. The COVID-19 pandemic has made medical practitioners' already high demands even more precarious. Stress in underprivileged areas and among veterans emphasizes the negative effects of work-related stress on mental health even more. Techniques that improve psychological well-being and lessen burnout include resilience training, digital tools, supportive leadership, and mindfulness-based stress reduction (MBSR). Enhancing work-life balance and promoting a healthier work environment can be achieved by combining these interventions with organizational changes. Aging-related cognitive impairment necessitates a multimodal strategy that includes targeted stress reduction methods and organizational adjustments. Setting mental health as a top priority in healthcare settings promotes the wellbeing of staff members, enhances patient care, and improves healthcare results.

考虑到老龄化人口的快速增长,了解认知能力下降及其原因,如压力,在不同群体中表现不同,是至关重要的。本章着眼于压力如何影响老年人的认知能力下降,特别强调医疗专业人员在其工作领域所面临的困难以及如何减轻后果。认知能力下降的严重程度和过程因人而异,受生活方式、病史和工作压力等因素的影响。COVID-19大流行使医生本已很高的要求变得更加不稳定。贫困地区和退伍军人的压力更强调了工作压力对心理健康的负面影响。改善心理健康和减少倦怠的技术包括弹性训练、数字工具、支持性领导和基于正念的减压(MBSR)。通过将这些干预措施与组织变革相结合,可以实现加强工作与生活平衡和促进更健康的工作环境。衰老相关的认知损伤需要多模式的策略,包括有针对性的减压方法和组织调整。将心理健康作为医疗保健机构的首要任务,可促进工作人员的福祉,加强对患者的护理,并改善医疗保健结果。
{"title":"Stress in specific population: Cognitive decline in aging, occupational challenges, strategies for medical professionals.","authors":"Gargi Gautam, Nasrollah Moradikor","doi":"10.1016/bs.pbr.2025.01.017","DOIUrl":"https://doi.org/10.1016/bs.pbr.2025.01.017","url":null,"abstract":"<p><p>Understanding cognitive decline and its contributing causes, such as stress, which presents differently in different groups, is crucial given the aging population's rapid growth. This chapter looks at how stress affects older persons' cognitive decline, with a particular emphasis on the difficulties faced by medical professionals in their line of work and how to lessen the consequences. The severity and course of cognitive decline differ from person to person and are impacted by factors such as lifestyle, medical history, and stress at work. The COVID-19 pandemic has made medical practitioners' already high demands even more precarious. Stress in underprivileged areas and among veterans emphasizes the negative effects of work-related stress on mental health even more. Techniques that improve psychological well-being and lessen burnout include resilience training, digital tools, supportive leadership, and mindfulness-based stress reduction (MBSR). Enhancing work-life balance and promoting a healthier work environment can be achieved by combining these interventions with organizational changes. Aging-related cognitive impairment necessitates a multimodal strategy that includes targeted stress reduction methods and organizational adjustments. Setting mental health as a top priority in healthcare settings promotes the wellbeing of staff members, enhances patient care, and improves healthcare results.</p>","PeriodicalId":20598,"journal":{"name":"Progress in brain research","volume":"291 ","pages":"363-379"},"PeriodicalIF":0.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144030602","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Introduction to neurobiology and pharmacology of stress. 神经生物学和应激药理学导论。
4区 医学 Q3 Neuroscience Pub Date : 2025-01-01 Epub Date: 2025-02-03 DOI: 10.1016/bs.pbr.2025.01.006
Saeid Abbasi-Maleki, Babak Nakhaei, Samira Rostami Mehr, Nasrollah Moradikor

In order to improve individual and community health outcomes, stress research is crucial for developing our understanding of human biology, psychology, and social dynamics. It also informs therapeutic practices, public health campaigns, and educational activities. The chapter explores how neurotransmitters, including glutamate, GABA, adrenaline, norepinephrine, serotonin, dopamine, and adrenaline, mediate stress responses, impact mood and behavior, and play a part in a number of stress-related disorders. The relevance of focused research and therapy approaches aimed at reestablishing equilibrium within these systems is highlighted by the fact that dysregulation of these neurotransmitters can exacerbate health problems. Additionally, it is investigated how the amygdala, hippocampus, and prefrontal cortex interact to process emotions, build resilience, and determine an individual's susceptibility to stress. These interactions are regulated by both neuroplasticity and hereditary and epigenetic factors. The chapter discusses the pharmaceutical approach to stress management, which includes a variety of drugs such as beta-blockers, anxiolytics, and antidepressants that work by targeting different neurotransmitter systems to reduce anxiety and mood disorders. Even while these therapies work, they may have negative consequences and side effects that should be carefully considered in clinical settings. The chapter promotes a comprehensive approach to stress management that combines medication, lifestyle changes, psychotherapy, and stress-reduction methods. Healthcare workers can improve patient care and ultimately the health and quality of life for people with stress-related disorders by knowing the complexity of pharmaceutical therapies and how they affect the stress response.

为了改善个人和社区的健康结果,压力研究对于发展我们对人类生物学、心理学和社会动态的理解至关重要。它还为治疗实践、公共卫生运动和教育活动提供信息。本章探讨了包括谷氨酸、GABA、肾上腺素、去甲肾上腺素、血清素、多巴胺和肾上腺素在内的神经递质如何介导应激反应,影响情绪和行为,并在许多与压力相关的疾病中发挥作用。这些神经递质的失调会加剧健康问题,这一事实强调了旨在重建这些系统内平衡的重点研究和治疗方法的相关性。此外,它还研究了杏仁核、海马体和前额皮质如何相互作用来处理情绪,建立弹性,并确定个体对压力的易感性。这些相互作用受到神经可塑性和遗传及表观遗传因素的调节。本章讨论了压力管理的药物方法,其中包括各种药物,如-受体阻滞剂、抗焦虑药和抗抑郁药,这些药物通过针对不同的神经递质系统来减少焦虑和情绪障碍。即使这些疗法有效,它们也可能有负面后果和副作用,在临床环境中应该仔细考虑。本章提倡一种综合的压力管理方法,包括药物治疗、生活方式改变、心理治疗和减压方法。通过了解药物治疗的复杂性以及它们如何影响压力反应,医护人员可以改善患者的护理,并最终改善压力相关疾病患者的健康和生活质量。
{"title":"Introduction to neurobiology and pharmacology of stress.","authors":"Saeid Abbasi-Maleki, Babak Nakhaei, Samira Rostami Mehr, Nasrollah Moradikor","doi":"10.1016/bs.pbr.2025.01.006","DOIUrl":"https://doi.org/10.1016/bs.pbr.2025.01.006","url":null,"abstract":"<p><p>In order to improve individual and community health outcomes, stress research is crucial for developing our understanding of human biology, psychology, and social dynamics. It also informs therapeutic practices, public health campaigns, and educational activities. The chapter explores how neurotransmitters, including glutamate, GABA, adrenaline, norepinephrine, serotonin, dopamine, and adrenaline, mediate stress responses, impact mood and behavior, and play a part in a number of stress-related disorders. The relevance of focused research and therapy approaches aimed at reestablishing equilibrium within these systems is highlighted by the fact that dysregulation of these neurotransmitters can exacerbate health problems. Additionally, it is investigated how the amygdala, hippocampus, and prefrontal cortex interact to process emotions, build resilience, and determine an individual's susceptibility to stress. These interactions are regulated by both neuroplasticity and hereditary and epigenetic factors. The chapter discusses the pharmaceutical approach to stress management, which includes a variety of drugs such as beta-blockers, anxiolytics, and antidepressants that work by targeting different neurotransmitter systems to reduce anxiety and mood disorders. Even while these therapies work, they may have negative consequences and side effects that should be carefully considered in clinical settings. The chapter promotes a comprehensive approach to stress management that combines medication, lifestyle changes, psychotherapy, and stress-reduction methods. Healthcare workers can improve patient care and ultimately the health and quality of life for people with stress-related disorders by knowing the complexity of pharmaceutical therapies and how they affect the stress response.</p>","PeriodicalId":20598,"journal":{"name":"Progress in brain research","volume":"291 ","pages":"1-19"},"PeriodicalIF":0.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144042299","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Cardiovascular influence on cognitive decline: The heart's role in neurodegenerative disorders. 心血管对认知能力下降的影响:心脏在神经退行性疾病中的作用。
4区 医学 Q3 Neuroscience Pub Date : 2025-01-01 Epub Date: 2025-04-18 DOI: 10.1016/bs.pbr.2025.04.006
Gargi Gautam, Nasrollah Moradikor

Cognitive performance is greatly influenced by cardiovascular health, as vascular integrity and brain perfusion are directly related to diseases including Parkinson's disease, Alzheimer's disease, and vascular dementia. Examining the intricate relationship between the heart and brain, this chapter highlights how atrial fibrillation, diabetes, hypertension, and dyslipidemia affect neurovascular coupling (NVC). Chronic inflammation, oxidative stress, and endothelial dysfunction are some of the risk factors that lead to neurodegeneration. The cerebral microvasculature is further compromised by atherosclerosis and heart failure, which exacerbates neuronal damage and increases the risk of dementia. Supported by clinical and epidemiological data, the discussion delves into the mechanisms behind vascular dementia and the vascular contributions to Alzheimer's disease. Slowing cognitive deterioration requires early intervention through lipid management, blood pressure control, and anticoagulant medication. Additionally, developments in precision medicine and neurovascular-targeted therapies present encouraging paths toward management and prevention. Through the discussion of modifiable cardiovascular risks, this chapter emphasizes how vital vascular health is to maintaining cognitive function and slowing the progression of neurodegenerative diseases.

认知表现很大程度上受心血管健康的影响,因为血管完整性和脑灌注与帕金森病、阿尔茨海默病和血管性痴呆等疾病直接相关。本章探讨了心脏和大脑之间复杂的关系,重点介绍了心房颤动、糖尿病、高血压和血脂异常如何影响神经血管耦合(NVC)。慢性炎症、氧化应激和内皮功能障碍是导致神经变性的一些危险因素。动脉粥样硬化和心力衰竭进一步损害了大脑微血管,从而加剧了神经元损伤,增加了患痴呆的风险。在临床和流行病学数据的支持下,讨论深入探讨血管性痴呆背后的机制以及血管对阿尔茨海默病的贡献。减缓认知退化需要通过脂质管理、血压控制和抗凝药物进行早期干预。此外,精准医学和神经血管靶向治疗的发展为管理和预防提供了令人鼓舞的途径。通过对可改变的心血管风险的讨论,本章强调血管健康对于维持认知功能和减缓神经退行性疾病的进展是多么重要。
{"title":"Cardiovascular influence on cognitive decline: The heart's role in neurodegenerative disorders.","authors":"Gargi Gautam, Nasrollah Moradikor","doi":"10.1016/bs.pbr.2025.04.006","DOIUrl":"https://doi.org/10.1016/bs.pbr.2025.04.006","url":null,"abstract":"<p><p>Cognitive performance is greatly influenced by cardiovascular health, as vascular integrity and brain perfusion are directly related to diseases including Parkinson's disease, Alzheimer's disease, and vascular dementia. Examining the intricate relationship between the heart and brain, this chapter highlights how atrial fibrillation, diabetes, hypertension, and dyslipidemia affect neurovascular coupling (NVC). Chronic inflammation, oxidative stress, and endothelial dysfunction are some of the risk factors that lead to neurodegeneration. The cerebral microvasculature is further compromised by atherosclerosis and heart failure, which exacerbates neuronal damage and increases the risk of dementia. Supported by clinical and epidemiological data, the discussion delves into the mechanisms behind vascular dementia and the vascular contributions to Alzheimer's disease. Slowing cognitive deterioration requires early intervention through lipid management, blood pressure control, and anticoagulant medication. Additionally, developments in precision medicine and neurovascular-targeted therapies present encouraging paths toward management and prevention. Through the discussion of modifiable cardiovascular risks, this chapter emphasizes how vital vascular health is to maintaining cognitive function and slowing the progression of neurodegenerative diseases.</p>","PeriodicalId":20598,"journal":{"name":"Progress in brain research","volume":"294 ","pages":"33-46"},"PeriodicalIF":0.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144668207","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Healthy happiness: The effect of happiness in promoting brain health. 健康快乐:快乐在促进大脑健康方面的作用。
4区 医学 Q3 Neuroscience Pub Date : 2025-01-01 Epub Date: 2025-03-25 DOI: 10.1016/bs.pbr.2025.03.004
Matthew-Zane L Broderick, Qadir Khan, Nasrollah Moradikor

The Merriam-Webster dictionary defines happiness as "a state of well-being and contentment". Psychologically, happiness can be defined as a state of current well-being and positivity resulting from recent stimuli, as well as long-term life satisfaction and peace of mind. This refers to two aspects of happiness discussed in psychology, first coined by Aristotle, pleasurable happiness known as "hedonia" and the long-term happiness of living a good life called "eudaimonia". These early concepts break down the vaguer and complex idea of happiness into long-term pleasure and positivity. This is where it starts to get more challenging when we try to define happiness in neurobiological terms. Many regions, pathways and circuits in the brain work in tandem to create the conscious and recognized state of happiness we cognitively perceive as humans. However, happiness is a feeling that transcends species boundaries. It is a positive state of emotions that can be expressed in different ways, including contentedness, peace, pleasure, excitement, relief, joy, and more. To do this, happiness must be broken down into constituent parts, localized to specific neuroanatomical regions, and associated with specific projects and structures to really build the anatomical architecture of happiness. Understanding how emotion is quantified and experimentally studied allows the field of neuroscience to build a comprehensive "neurobiological happiness model". Here in this chapter, we discuss historical and novel findings into this emotion; we discuss its implication as an evolutionary advantage in the adaptive response, how laughter is associated with happiness, and how a state of positive well-being plays a role in promoting positive brain health.

韦氏词典将幸福定义为“一种幸福和满足的状态”。在心理学上,幸福可以被定义为一种由最近的刺激引起的当前的幸福和积极的状态,以及长期的生活满意度和内心的平静。这指的是心理学中讨论的幸福的两个方面,首先由亚里士多德提出,愉悦的幸福被称为“享乐主义”,而长期幸福的美好生活被称为“快乐主义”。这些早期的概念将模糊而复杂的幸福概念分解为长期的快乐和积极。当我们试图用神经生物学的术语来定义幸福时,这就开始变得更具挑战性了。大脑中的许多区域、路径和回路协同工作,创造了我们作为人类认知到的有意识和公认的幸福状态。然而,幸福是一种超越物种界限的感觉。它是一种积极的情绪状态,可以用不同的方式表达,包括满足、和平、愉悦、兴奋、放松、喜悦等等。要做到这一点,幸福必须被分解成组成部分,定位于特定的神经解剖区域,并与特定的项目和结构相关联,以真正构建幸福的解剖结构。理解情绪是如何被量化和实验研究的,可以让神经科学领域建立一个全面的“神经生物学幸福模型”。在本章中,我们将讨论有关这种情感的历史和新发现;我们讨论了它作为适应性反应的进化优势的含义,笑是如何与幸福联系在一起的,以及积极的幸福状态如何在促进积极的大脑健康中发挥作用。
{"title":"Healthy happiness: The effect of happiness in promoting brain health.","authors":"Matthew-Zane L Broderick, Qadir Khan, Nasrollah Moradikor","doi":"10.1016/bs.pbr.2025.03.004","DOIUrl":"https://doi.org/10.1016/bs.pbr.2025.03.004","url":null,"abstract":"<p><p>The Merriam-Webster dictionary defines happiness as \"a state of well-being and contentment\". Psychologically, happiness can be defined as a state of current well-being and positivity resulting from recent stimuli, as well as long-term life satisfaction and peace of mind. This refers to two aspects of happiness discussed in psychology, first coined by Aristotle, pleasurable happiness known as \"hedonia\" and the long-term happiness of living a good life called \"eudaimonia\". These early concepts break down the vaguer and complex idea of happiness into long-term pleasure and positivity. This is where it starts to get more challenging when we try to define happiness in neurobiological terms. Many regions, pathways and circuits in the brain work in tandem to create the conscious and recognized state of happiness we cognitively perceive as humans. However, happiness is a feeling that transcends species boundaries. It is a positive state of emotions that can be expressed in different ways, including contentedness, peace, pleasure, excitement, relief, joy, and more. To do this, happiness must be broken down into constituent parts, localized to specific neuroanatomical regions, and associated with specific projects and structures to really build the anatomical architecture of happiness. Understanding how emotion is quantified and experimentally studied allows the field of neuroscience to build a comprehensive \"neurobiological happiness model\". Here in this chapter, we discuss historical and novel findings into this emotion; we discuss its implication as an evolutionary advantage in the adaptive response, how laughter is associated with happiness, and how a state of positive well-being plays a role in promoting positive brain health.</p>","PeriodicalId":20598,"journal":{"name":"Progress in brain research","volume":"293 ","pages":"127-148"},"PeriodicalIF":0.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144181214","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Influence of feelings on the blood-brain barrier (BBB) and drug delivery. 感觉对血脑屏障(BBB)和药物传递的影响。
4区 医学 Q3 Neuroscience Pub Date : 2025-01-01 Epub Date: 2025-03-31 DOI: 10.1016/bs.pbr.2025.03.006
Omnia Ocab, Hager Adel Saad, Rowan Mashal, Karim Osama, Mahmoud Marzouk, Nabila Hamdi

The blood-brain barrier (BBB) is a critical regulator of cerebral homeostasis, displaying high dynamicity of influx and efflux of substances to and from the central nervous system (CNS). In an exploration of the neurochemical pathways through which positive and negative emotions can influence the physiological characteristics of the BBB, this chapter delves into the multifaceted relationship between emotional states and BBB integrity and permeability. Negative emotions exemplified by stress, chronic anxiety, and depression have shown harmful effects on the BBB, suggesting a state of hyperpermeability that compromises the otherwise conferred protection. Few reports in literature examined the exact molecular mechanisms by which negative emotions exhibit signs of damaged and leaky BBB. Although research deciphering those mechanisms is limited, there is consensus that the disruption of tight junction (TJ) protein integrity and expression, along with neuroinflammatory processes, oxidative Stress, and excitotoxicity, plays a role in the induction of BBB damage. Conversely, positive emotions have been shown to exert protective effects, potentially reversing the increased permeability of the BBB. Compared to the research focused on the neural correlates of negative emotions, the neuroscience literature on positive emotions and well-being is still in its infancy. A deeper understanding of the mechanisms by which positive emotions modulate the BBB remains necessary. Additionally, we discuss the therapeutic implications of these findings, considering how emotional well-being can be leveraged in developing treatments for neurological disorders. By integrating neuroscience, psychology, and pharmacology insights, this chapter aims to comprehensively understand the dynamic interplay between emotions and the BBB and its potential to inform novel therapeutic strategies.

血脑屏障(BBB)是大脑内稳态的重要调节因子,在物质流入和流出中枢神经系统(CNS)方面表现出高度的动态性。在积极和消极情绪影响血脑屏障生理特征的神经化学途径的探索中,本章深入探讨了情绪状态与血脑屏障完整性和通透性之间的多方面关系。压力、慢性焦虑和抑郁等负面情绪对血脑屏障产生了有害的影响,表明血脑屏障处于一种过度渗透的状态,损害了原本赋予的保护作用。文献中很少有研究负面情绪表现出血脑屏障受损和渗漏迹象的确切分子机制的报告。尽管对这些机制的研究有限,但人们一致认为,紧密连接(TJ)蛋白完整性和表达的破坏,以及神经炎症过程、氧化应激和兴奋毒性,在诱导血脑屏障损伤中起作用。相反,积极的情绪已被证明具有保护作用,可能逆转血脑屏障通透性的增加。与关注消极情绪的神经相关研究相比,关于积极情绪和幸福感的神经科学文献仍处于起步阶段。对积极情绪调节血脑屏障的机制有更深入的了解仍然是必要的。此外,我们讨论了这些发现的治疗意义,考虑到情绪健康如何在开发神经系统疾病的治疗中发挥作用。通过整合神经科学、心理学和药理学的见解,本章旨在全面了解情绪和血脑屏障之间的动态相互作用及其为新的治疗策略提供信息的潜力。
{"title":"Influence of feelings on the blood-brain barrier (BBB) and drug delivery.","authors":"Omnia Ocab, Hager Adel Saad, Rowan Mashal, Karim Osama, Mahmoud Marzouk, Nabila Hamdi","doi":"10.1016/bs.pbr.2025.03.006","DOIUrl":"https://doi.org/10.1016/bs.pbr.2025.03.006","url":null,"abstract":"<p><p>The blood-brain barrier (BBB) is a critical regulator of cerebral homeostasis, displaying high dynamicity of influx and efflux of substances to and from the central nervous system (CNS). In an exploration of the neurochemical pathways through which positive and negative emotions can influence the physiological characteristics of the BBB, this chapter delves into the multifaceted relationship between emotional states and BBB integrity and permeability. Negative emotions exemplified by stress, chronic anxiety, and depression have shown harmful effects on the BBB, suggesting a state of hyperpermeability that compromises the otherwise conferred protection. Few reports in literature examined the exact molecular mechanisms by which negative emotions exhibit signs of damaged and leaky BBB. Although research deciphering those mechanisms is limited, there is consensus that the disruption of tight junction (TJ) protein integrity and expression, along with neuroinflammatory processes, oxidative Stress, and excitotoxicity, plays a role in the induction of BBB damage. Conversely, positive emotions have been shown to exert protective effects, potentially reversing the increased permeability of the BBB. Compared to the research focused on the neural correlates of negative emotions, the neuroscience literature on positive emotions and well-being is still in its infancy. A deeper understanding of the mechanisms by which positive emotions modulate the BBB remains necessary. Additionally, we discuss the therapeutic implications of these findings, considering how emotional well-being can be leveraged in developing treatments for neurological disorders. By integrating neuroscience, psychology, and pharmacology insights, this chapter aims to comprehensively understand the dynamic interplay between emotions and the BBB and its potential to inform novel therapeutic strategies.</p>","PeriodicalId":20598,"journal":{"name":"Progress in brain research","volume":"293 ","pages":"203-242"},"PeriodicalIF":0.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144181220","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Impact of positive feelings and negative feelings on brain cell regeneration and degeneration. 积极感受和消极感受对脑细胞再生和退化的影响。
4区 医学 Q3 Neuroscience Pub Date : 2025-01-01 Epub Date: 2025-02-28 DOI: 10.1016/bs.pbr.2025.02.009
Al-Hassan Soliman Wadan, Mohamed Abdelsattar Ahmed, Dana Saeed Abd Elmonem El Gemaie, Nasrollah Moradikor

Brain cells intentionally break their DNA as a crucial step in memory formation and learning. This process allows for the expression of specific genes that are essential for these cognitive functions. While our cells are generally adept at repairing this self-inflicted DNA damage, the efficiency of this repair mechanism can decline with age or due to certain genetic factors. The deliberate DNA breakage in neurons enables the activation of rapid response genes, which in turn trigger broader transcriptional programs supporting various behaviors, including learning and memory. This process is a normal part of cellular function and gene expression. However, neurological disorders, certain syndromes, and the aging process can impair this DNA repair ability. When cells struggle to mend the intentional DNA breaks, it can lead to cellular weakening and eventual degeneration. The subsequent discussion will explore how positive and negative emotions influence the processes of brain cell regeneration and degradation.

脑细胞故意破坏它们的DNA,作为记忆形成和学习的关键步骤。这个过程允许表达对这些认知功能至关重要的特定基因。虽然我们的细胞通常擅长修复这种自我造成的DNA损伤,但这种修复机制的效率会随着年龄的增长或某些遗传因素而下降。神经元中故意的DNA断裂可以激活快速反应基因,从而触发更广泛的转录程序,支持各种行为,包括学习和记忆。这个过程是细胞功能和基因表达的正常部分。然而,神经系统疾病、某些综合征和衰老过程会损害这种DNA修复能力。当细胞努力修复故意的DNA断裂时,它会导致细胞衰弱并最终退化。接下来的讨论将探讨积极和消极情绪如何影响脑细胞再生和退化的过程。
{"title":"Impact of positive feelings and negative feelings on brain cell regeneration and degeneration.","authors":"Al-Hassan Soliman Wadan, Mohamed Abdelsattar Ahmed, Dana Saeed Abd Elmonem El Gemaie, Nasrollah Moradikor","doi":"10.1016/bs.pbr.2025.02.009","DOIUrl":"https://doi.org/10.1016/bs.pbr.2025.02.009","url":null,"abstract":"<p><p>Brain cells intentionally break their DNA as a crucial step in memory formation and learning. This process allows for the expression of specific genes that are essential for these cognitive functions. While our cells are generally adept at repairing this self-inflicted DNA damage, the efficiency of this repair mechanism can decline with age or due to certain genetic factors. The deliberate DNA breakage in neurons enables the activation of rapid response genes, which in turn trigger broader transcriptional programs supporting various behaviors, including learning and memory. This process is a normal part of cellular function and gene expression. However, neurological disorders, certain syndromes, and the aging process can impair this DNA repair ability. When cells struggle to mend the intentional DNA breaks, it can lead to cellular weakening and eventual degeneration. The subsequent discussion will explore how positive and negative emotions influence the processes of brain cell regeneration and degradation.</p>","PeriodicalId":20598,"journal":{"name":"Progress in brain research","volume":"293 ","pages":"1-15"},"PeriodicalIF":0.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144182197","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Hormetic pathways in environmental enrichment in animal models and humans. 动物模型和人类环境富集中的激光途径。
4区 医学 Q3 Neuroscience Pub Date : 2025-01-01 Epub Date: 2025-05-20 DOI: 10.1016/bs.pbr.2025.05.007
Maylin Hanampa-Maquera, Rafael Cândido Lourenço, Alexis Bailey, Rosana Camarini

Environmental Enrichment (EE), which provides enhanced sensory, cognitive, motor, and social stimulation, has emerged as a powerful paradigm for investigating neuroplasticity and stress resilience. This chapter explores how EE functions through hormetic mechanisms-where multiple mild stimuli trigger adaptive responses that promote beneficial outcomes. Since Hebb's pioneering work on neuronal ensembles, research has demonstrated that EE enhances neurogenesis, synaptic plasticity, and neurotrophic factor expression (BDNF and NGF) while modulating inflammatory processes, epigenetic pathways, and metabolic function. These adaptive responses operate according to biphasic dose-response patterns characteristic of hormesis, where moderate stimulation produces benefits that may diminish or become detrimental with excessive exposure. In animal models, EE has shown remarkable efficacy in mitigating cognitive decline, reducing anxiety-like behaviors, attenuating addiction vulnerability, and protecting against neurodegenerative diseases. The modulation of the hypothalamic-pituitary-adrenal (HPA) axis and the shifts in microglial phenotype observed with EE illustrate its role as a hormetic stimulus, as it can act as a mild stressor that promotes adaptive neuroplasticity, enhancing the organism's ability to cope with future stressors. In humans, analogous enrichments through physical exercise, cognitive challenges, social engagement, and music facilitate neuroplasticity, protect against cognitive decline, and promote stress resilience. The hormetic framework also explains why enrichment must be tailored to individual thresholds-excessive stimulation can overwhelm adaptive capacities, transforming beneficial eustress into harmful distress. By understanding EE as a hormetic intervention, this chapter bridges basic neuroscience with translational applications that may enhance resilience against neuropsychiatric disorders typically prevalent in aging.

环境富集(Environmental Enrichment, EE)提供了增强的感觉、认知、运动和社会刺激,已成为研究神经可塑性和应激恢复力的有力范例。本章探讨情感表达是如何通过刺激机制发挥作用的——多重轻微刺激触发促进有益结果的适应性反应。自Hebb在神经元集合方面的开创性工作以来,研究表明,EE增强神经发生、突触可塑性和神经营养因子表达(BDNF和NGF),同时调节炎症过程、表观遗传途径和代谢功能。这些适应性反应是根据激效的双相剂量-反应模式运作的,适度刺激产生的益处可能会随着过度暴露而减弱或变得有害。在动物模型中,情感表达在减轻认知能力下降、减少焦虑样行为、减轻成瘾脆弱性和预防神经退行性疾病方面显示出显著的功效。用EE观察到的下丘脑-垂体-肾上腺(HPA)轴的调节和小胶质细胞表型的变化说明了其作为一种激效刺激的作用,因为它可以作为一种轻度应激源,促进适应性神经可塑性,增强生物体应对未来应激源的能力。在人类中,通过体育锻炼、认知挑战、社会参与和音乐进行类似的丰富可以促进神经可塑性,防止认知能力下降,并促进压力恢复能力。激效框架也解释了为什么必须根据个体阈值进行强化——过度的刺激会压倒适应能力,将有益的压力转化为有害的痛苦。通过将情感表达理解为一种刺激物干预,本章将基础神经科学与翻译应用联系起来,这些应用可能增强对衰老中普遍存在的神经精神疾病的恢复能力。
{"title":"Hormetic pathways in environmental enrichment in animal models and humans.","authors":"Maylin Hanampa-Maquera, Rafael Cândido Lourenço, Alexis Bailey, Rosana Camarini","doi":"10.1016/bs.pbr.2025.05.007","DOIUrl":"https://doi.org/10.1016/bs.pbr.2025.05.007","url":null,"abstract":"<p><p>Environmental Enrichment (EE), which provides enhanced sensory, cognitive, motor, and social stimulation, has emerged as a powerful paradigm for investigating neuroplasticity and stress resilience. This chapter explores how EE functions through hormetic mechanisms-where multiple mild stimuli trigger adaptive responses that promote beneficial outcomes. Since Hebb's pioneering work on neuronal ensembles, research has demonstrated that EE enhances neurogenesis, synaptic plasticity, and neurotrophic factor expression (BDNF and NGF) while modulating inflammatory processes, epigenetic pathways, and metabolic function. These adaptive responses operate according to biphasic dose-response patterns characteristic of hormesis, where moderate stimulation produces benefits that may diminish or become detrimental with excessive exposure. In animal models, EE has shown remarkable efficacy in mitigating cognitive decline, reducing anxiety-like behaviors, attenuating addiction vulnerability, and protecting against neurodegenerative diseases. The modulation of the hypothalamic-pituitary-adrenal (HPA) axis and the shifts in microglial phenotype observed with EE illustrate its role as a hormetic stimulus, as it can act as a mild stressor that promotes adaptive neuroplasticity, enhancing the organism's ability to cope with future stressors. In humans, analogous enrichments through physical exercise, cognitive challenges, social engagement, and music facilitate neuroplasticity, protect against cognitive decline, and promote stress resilience. The hormetic framework also explains why enrichment must be tailored to individual thresholds-excessive stimulation can overwhelm adaptive capacities, transforming beneficial eustress into harmful distress. By understanding EE as a hormetic intervention, this chapter bridges basic neuroscience with translational applications that may enhance resilience against neuropsychiatric disorders typically prevalent in aging.</p>","PeriodicalId":20598,"journal":{"name":"Progress in brain research","volume":"295 ","pages":"189-227"},"PeriodicalIF":0.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144795179","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Biomarkers: From early detection to treatment personalization. 生物标志物:从早期检测到个性化治疗。
4区 医学 Q3 Neuroscience Pub Date : 2025-01-01 Epub Date: 2025-08-25 DOI: 10.1016/bs.pbr.2025.08.008
Saman Fatima, Sakshi Tiwari, Bilal Siddiqi, Syed Naved Quadri, M Z Abdin

Neurodegenerative disorders (NDs), such as Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS), present increasing issues associated with the unavoidable aging of the world's population. These challenges are further highlighted by the socioeconomic consequences of these diseases. The identification and use of biomarkers for prompt diagnosis, careful observation, and efficient treatment approaches is essential to overcoming these obstacles. The primary methods for diagnosing neurodegenerative illnesses are invasive procedures like lumbar punctures to measure CSF fluid or functional brain imaging methods. Biomarkers for underlying proteinopathy in blood serum and cerebral fluid have been the focus of recent biological research, particularly in vivo. With their ability to provide novel pathways for early detection, illness progression tracking, and individualized treatment plans, biomarkers have become essential instruments in precision medicine. The classification of biomarkers including fluid, digital imaging, and molecular biomarkers is examined in this chapter, with an emphasis on their function in neurodegenerative diseases. In neurodegenerative illnesses and the aging brain, tau, amyloid-β, α-synuclein, and TDP-43 are commonly seen to be deposited together rather than separately. These may be disregarded, and it might be challenging to determine their clinicopathological significance. An overview of illness pathophysiology, diagnostic implications, and the most recent molecular and ultrastructural categories for neurodegenerative disorders are given in this chapter. Addressing these issues through interdisciplinary research and technological advancements will be crucial for the future of biomarker-driven precision medicine. This chapter provides an in-depth overview of the evolving landscape of biomarkers and their transformative impact on the early detection and personalized treatment of neurodegenerative diseases.

神经退行性疾病(NDs),如阿尔茨海默病(AD)、帕金森病(PD)和肌萎缩侧索硬化症(ALS),随着世界人口不可避免的老龄化,呈现出越来越多的问题。这些疾病的社会经济后果进一步突出了这些挑战。识别和使用生物标志物进行及时诊断、仔细观察和有效的治疗方法对于克服这些障碍至关重要。诊断神经退行性疾病的主要方法是侵入性手术,如腰椎穿刺来测量脑脊液或功能性脑成像方法。血清和脑液中潜在蛋白病的生物标志物已成为最近生物学研究的焦点,特别是在体内。生物标志物能够为早期检测、疾病进展跟踪和个性化治疗计划提供新的途径,已成为精准医学的重要工具。生物标志物的分类包括流体、数字成像和分子生物标志物,本章将重点介绍它们在神经退行性疾病中的功能。在神经退行性疾病和老化的大脑中,tau、淀粉样蛋白-β、α-突触核蛋白和TDP-43通常聚集在一起,而不是单独沉积。这些可能被忽视,它可能是具有挑战性的确定其临床病理意义。本章概述了神经退行性疾病的病理生理学、诊断意义以及最新的分子和超微结构分类。通过跨学科研究和技术进步来解决这些问题对于生物标志物驱动的精准医学的未来至关重要。本章深入概述了生物标志物的发展前景及其对神经退行性疾病的早期检测和个性化治疗的变革性影响。
{"title":"Biomarkers: From early detection to treatment personalization.","authors":"Saman Fatima, Sakshi Tiwari, Bilal Siddiqi, Syed Naved Quadri, M Z Abdin","doi":"10.1016/bs.pbr.2025.08.008","DOIUrl":"https://doi.org/10.1016/bs.pbr.2025.08.008","url":null,"abstract":"<p><p>Neurodegenerative disorders (NDs), such as Alzheimer's disease (AD), Parkinson's disease (PD), and amyotrophic lateral sclerosis (ALS), present increasing issues associated with the unavoidable aging of the world's population. These challenges are further highlighted by the socioeconomic consequences of these diseases. The identification and use of biomarkers for prompt diagnosis, careful observation, and efficient treatment approaches is essential to overcoming these obstacles. The primary methods for diagnosing neurodegenerative illnesses are invasive procedures like lumbar punctures to measure CSF fluid or functional brain imaging methods. Biomarkers for underlying proteinopathy in blood serum and cerebral fluid have been the focus of recent biological research, particularly in vivo. With their ability to provide novel pathways for early detection, illness progression tracking, and individualized treatment plans, biomarkers have become essential instruments in precision medicine. The classification of biomarkers including fluid, digital imaging, and molecular biomarkers is examined in this chapter, with an emphasis on their function in neurodegenerative diseases. In neurodegenerative illnesses and the aging brain, tau, amyloid-β, α-synuclein, and TDP-43 are commonly seen to be deposited together rather than separately. These may be disregarded, and it might be challenging to determine their clinicopathological significance. An overview of illness pathophysiology, diagnostic implications, and the most recent molecular and ultrastructural categories for neurodegenerative disorders are given in this chapter. Addressing these issues through interdisciplinary research and technological advancements will be crucial for the future of biomarker-driven precision medicine. This chapter provides an in-depth overview of the evolving landscape of biomarkers and their transformative impact on the early detection and personalized treatment of neurodegenerative diseases.</p>","PeriodicalId":20598,"journal":{"name":"Progress in brain research","volume":"297 ","pages":"131-153"},"PeriodicalIF":0.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145637995","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Future horizons: Innovation, aging, and equity. 未来展望:创新、老龄化和公平。
4区 医学 Q3 Neuroscience Pub Date : 2025-01-01 Epub Date: 2025-11-17 DOI: 10.1016/bs.pbr.2025.08.010
Manoj Kumar Jana, Vishnu Swarup, Suryaprakash Tripathy, Neeraj Mohan Gupta, Sai Sutheertha Chatla, Deepesh Joshi, Subhadeep Mandal, Anushka Giri, Sayantam Jana, Mahesh Narayan, Sirsendu Bikash Maiti, Sudip Das
<p><p>Precision medicine is on the verge of transforming the treatment of neurodegenerative diseases (NDDs) like Alzheimer's disease (AD) and Parkinson's disease (PD), in response to the intricate interactions of genetic, epigenetic, environmental, and lifestyle factors underlying disease heterogeneity. As the world's aging populations grow, with dementia cases expected to double by 2040 and the costs amounting to over €130 billion a year in Europe alone, there is an urgent need for novel strategies to stem the socioeconomic costs of NDDs. Conventional "one-drug-fits-all" strategies that depend on late-stage symptom treatment are progressively insufficient for disorders that are marked by heterogeneous molecular pathways and unpredictable clinical courses. Recent improvements in artificial intelligence (AI), multi-omics integration, and biomarker research now allow patients to be stratified into subpopulations following their genetic risk profiles, neuroimaging signatures, and fluid biomarkers (e.g., amyloid-beta, tau, α-synuclein), enabling early diagnosis and focused treatments. For example, artificial intelligence platforms such as the IHI-PROMINENT project are creating forecasting algorithms to chart disease progression and tailor treatment outcomes, and gene therapy and antisense oligonucleotides (ASOs) address precise mutations in familial AD and PD. These advances are supported by pharmacogenomics, which individualizes drug regimens according to metabolic profiles to reduce side effects and maximize efficacy. Still, translating these advances into practice has major barriers to overcome, such as large-scale biomarker validation, multi-omics standardization, and incorporating real-world evidence from digital health technologies. Aging populations only add complexity to this environment, as comorbidities like diabetes and cardiovascular diseases interact with neurodegenerative pathways, requiring system-based, holistic approaches to care. Equity is still a key challenge: differences in access to sophisticated diagnostics (e.g., PET scans, CSF examination) and expensive therapies (e.g., monoclonal antibodies, CAR-T cell therapy) threaten to worsen global health disparities. In retaliation, initiatives such as the JPND research paradigm advance remote clinical trials and telemedicine platforms for the diverse community in decentralized settings, and policies target reducing financial disincentives through risk-sharing strategies and public-private partnerships. Precision medicine in the treatment of NDDs depends on an integrated network among academia, clinics, and industry, by taking advantage of communal biobanks and AI-enabled big data analysis, for refining the drug development process and validating new targets, e.g., neuroinflammatory signaling and gut-brain axis dysfunction. Innovations, like CRISPR-mediated editing and ambient neuroimaging, have innate or potential power to personalize treatment by identifying early-stage and even pre-
精准医学即将改变神经退行性疾病(ndd)的治疗,如阿尔茨海默病(AD)和帕金森病(PD),以应对潜在疾病异质性的遗传、表观遗传、环境和生活方式因素之间复杂的相互作用。随着世界老龄化人口的增长,预计到2040年痴呆症病例将翻一番,仅在欧洲每年的成本就超过1300亿欧元,迫切需要新的战略来遏制ndd的社会经济成本。依赖于晚期症状治疗的传统“一刀切”策略,对于以异质分子途径和不可预测的临床过程为特征的疾病来说,逐渐不够用。人工智能(AI)、多组学整合和生物标志物研究的最新进展,现在可以根据患者的遗传风险谱、神经成像特征和液体生物标志物(如淀粉样蛋白- β、tau、α-突触核蛋白)将患者分层为亚群,从而实现早期诊断和重点治疗。例如,人工智能平台(如IHI-PROMINENT项目)正在创建预测算法来绘制疾病进展和定制治疗结果,基因疗法和反义寡核苷酸(ASOs)可以解决家族性AD和PD的精确突变。这些进步得到了药物基因组学的支持,药物基因组学根据代谢特征个性化药物方案,以减少副作用并最大限度地提高疗效。然而,将这些进步转化为实践还需要克服一些主要障碍,例如大规模生物标志物验证、多组学标准化以及整合来自数字卫生技术的真实证据。人口老龄化只会增加这一环境的复杂性,因为糖尿病和心血管疾病等合并症与神经退行性途径相互作用,需要基于系统的整体护理方法。公平仍然是一项关键挑战:在获得复杂诊断(如PET扫描、脑脊液检查)和昂贵疗法(如单克隆抗体、CAR-T细胞疗法)方面的差异有可能加剧全球健康差距。作为报复,JPND研究范式等举措推进了分散环境下多样化社区的远程临床试验和远程医疗平台,政策目标是通过风险分担战略和公私伙伴关系减少财政障碍。ndd治疗中的精准医疗依赖于学术界、诊所和产业界之间的综合网络,利用公共生物库和人工智能支持的大数据分析,完善药物开发过程并验证新的靶点,例如神经炎症信号和肠-脑轴功能障碍。像crispr介导的编辑和环境神经成像这样的创新,通过识别早期甚至症状前的患者,并根据遗传风险调整个人的生活方式,具有先天或潜在的个性化治疗能力。然而,围绕数据隐私、算法偏见和终身持续治疗干预的知情同意的伦理考虑应该指导而不是滞后这种转变。随着对预防而不是延迟护理的推动,精准医学代表了卫生保健领域的革命性范式转变,并有可能将ndd从毁灭性的致命诊断转变为易于管理的慢性疾病,并使大众能够公平地获得创新。成功需要在转化研究、跨学科培训和全球监管协调方面的持续投资,以将精准医学的承诺转化为数百万神经退行性疾病患者生活质量的切实改善。
{"title":"Future horizons: Innovation, aging, and equity.","authors":"Manoj Kumar Jana, Vishnu Swarup, Suryaprakash Tripathy, Neeraj Mohan Gupta, Sai Sutheertha Chatla, Deepesh Joshi, Subhadeep Mandal, Anushka Giri, Sayantam Jana, Mahesh Narayan, Sirsendu Bikash Maiti, Sudip Das","doi":"10.1016/bs.pbr.2025.08.010","DOIUrl":"https://doi.org/10.1016/bs.pbr.2025.08.010","url":null,"abstract":"&lt;p&gt;&lt;p&gt;Precision medicine is on the verge of transforming the treatment of neurodegenerative diseases (NDDs) like Alzheimer's disease (AD) and Parkinson's disease (PD), in response to the intricate interactions of genetic, epigenetic, environmental, and lifestyle factors underlying disease heterogeneity. As the world's aging populations grow, with dementia cases expected to double by 2040 and the costs amounting to over €130 billion a year in Europe alone, there is an urgent need for novel strategies to stem the socioeconomic costs of NDDs. Conventional \"one-drug-fits-all\" strategies that depend on late-stage symptom treatment are progressively insufficient for disorders that are marked by heterogeneous molecular pathways and unpredictable clinical courses. Recent improvements in artificial intelligence (AI), multi-omics integration, and biomarker research now allow patients to be stratified into subpopulations following their genetic risk profiles, neuroimaging signatures, and fluid biomarkers (e.g., amyloid-beta, tau, α-synuclein), enabling early diagnosis and focused treatments. For example, artificial intelligence platforms such as the IHI-PROMINENT project are creating forecasting algorithms to chart disease progression and tailor treatment outcomes, and gene therapy and antisense oligonucleotides (ASOs) address precise mutations in familial AD and PD. These advances are supported by pharmacogenomics, which individualizes drug regimens according to metabolic profiles to reduce side effects and maximize efficacy. Still, translating these advances into practice has major barriers to overcome, such as large-scale biomarker validation, multi-omics standardization, and incorporating real-world evidence from digital health technologies. Aging populations only add complexity to this environment, as comorbidities like diabetes and cardiovascular diseases interact with neurodegenerative pathways, requiring system-based, holistic approaches to care. Equity is still a key challenge: differences in access to sophisticated diagnostics (e.g., PET scans, CSF examination) and expensive therapies (e.g., monoclonal antibodies, CAR-T cell therapy) threaten to worsen global health disparities. In retaliation, initiatives such as the JPND research paradigm advance remote clinical trials and telemedicine platforms for the diverse community in decentralized settings, and policies target reducing financial disincentives through risk-sharing strategies and public-private partnerships. Precision medicine in the treatment of NDDs depends on an integrated network among academia, clinics, and industry, by taking advantage of communal biobanks and AI-enabled big data analysis, for refining the drug development process and validating new targets, e.g., neuroinflammatory signaling and gut-brain axis dysfunction. Innovations, like CRISPR-mediated editing and ambient neuroimaging, have innate or potential power to personalize treatment by identifying early-stage and even pre-","PeriodicalId":20598,"journal":{"name":"Progress in brain research","volume":"297 ","pages":"427-468"},"PeriodicalIF":0.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145638060","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Stress and gender differences in brain development. 大脑发育中的压力和性别差异。
4区 医学 Q3 Neuroscience Pub Date : 2025-01-01 Epub Date: 2025-04-02 DOI: 10.1016/bs.pbr.2025.01.013
Abraham Olufemi Asuku, Priscilla Omobonke Adebayo, Gbonjubola Oyinlola Ogungbangbe

This chapter investigates the ways in which male and female brains are differently affected by stress during early development, which in turn affects how susceptible each group is to stress-related illnesses. When examining the structure and function of the brain, gender differences and stress must be taken into account. Male and female brain development differs in response to the prenatal testis's secretion of androgen. It appears that when it comes to responding to stress, encoding memories, feeling emotions, solving specific issues, and making decisions, men and women use distinct areas of the brain. Findings revealed that stress led to specific changes in brain structure and function, with gender-specific differences observed. The prefrontal cortex, the hippocampus, and the amygdala are among the brain regions connected to the stress response. The stress response has been linked to the presentation of numerous mental and psychosomatic conditions. The way men and women respond to stress varies on a biological and psychological level. To gain more insight into the gender differences seen throughout brain development, these disparities must also be investigated. This chapter implies that gender-specific vulnerabilities should be addressed and healthy brain development should be promoted by stress-related interventions.

本章调查了男性和女性大脑在早期发育过程中受到压力影响的不同方式,这反过来又影响了每个群体对压力相关疾病的易感性。在检查大脑的结构和功能时,必须考虑到性别差异和压力。男性和女性大脑发育对产前睾丸分泌雄激素的反应不同。研究表明,在应对压力、编码记忆、感受情绪、解决具体问题和做出决定时,男性和女性使用的大脑区域是不同的。研究结果显示,压力会导致大脑结构和功能的特定变化,并存在性别差异。前额皮质、海马体和杏仁核是大脑中与压力反应有关的区域。应激反应与许多精神和心身疾病的表现有关。男性和女性应对压力的方式在生理和心理层面上有所不同。为了更深入地了解大脑发育过程中的性别差异,还必须对这些差异进行调查。本章暗示,应解决性别特定的脆弱性问题,并应通过与压力有关的干预措施促进健康的大脑发育。
{"title":"Stress and gender differences in brain development.","authors":"Abraham Olufemi Asuku, Priscilla Omobonke Adebayo, Gbonjubola Oyinlola Ogungbangbe","doi":"10.1016/bs.pbr.2025.01.013","DOIUrl":"https://doi.org/10.1016/bs.pbr.2025.01.013","url":null,"abstract":"<p><p>This chapter investigates the ways in which male and female brains are differently affected by stress during early development, which in turn affects how susceptible each group is to stress-related illnesses. When examining the structure and function of the brain, gender differences and stress must be taken into account. Male and female brain development differs in response to the prenatal testis's secretion of androgen. It appears that when it comes to responding to stress, encoding memories, feeling emotions, solving specific issues, and making decisions, men and women use distinct areas of the brain. Findings revealed that stress led to specific changes in brain structure and function, with gender-specific differences observed. The prefrontal cortex, the hippocampus, and the amygdala are among the brain regions connected to the stress response. The stress response has been linked to the presentation of numerous mental and psychosomatic conditions. The way men and women respond to stress varies on a biological and psychological level. To gain more insight into the gender differences seen throughout brain development, these disparities must also be investigated. This chapter implies that gender-specific vulnerabilities should be addressed and healthy brain development should be promoted by stress-related interventions.</p>","PeriodicalId":20598,"journal":{"name":"Progress in brain research","volume":"291 ","pages":"319-337"},"PeriodicalIF":0.0,"publicationDate":"2025-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143980675","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Progress in brain research
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1