在老化過程中,人體會產生一種慢性、低度、無菌的發炎狀態,學術上稱為「發炎老化(inflammaging)」。這種長期存在的微發炎狀態與多種年齡相關疾病(如糖尿病、心血管疾病、神經退化疾病等)息息相關,且被認為是這些疾病發展的潛在驅動力。
代謝發炎是另一種與營養過剩有關的慢性發炎形式,特別與代謝症候群與肥胖有關。研究顯示,代謝發炎與發炎老化共享類似的生理機制,兩者皆涉及免疫系統對特定刺激(如過量營養素、脂肪酸、微生物分子等)的反應。
這些刺激會透過一小部分具「退化性辨識能力」的免疫受體來啟動先天免疫反應,形成長期發炎狀態。
腸道菌群是連結營養、免疫與老化的重要橋樑。它不僅會釋放促發炎物質,還與身體的晝夜節律調節和多個器官系統進行訊息交流。腸道菌相的失衡不僅會促進發炎老化與代謝發炎,也與加速老化及慢性病惡化有關。
文章也提倡使用新一代的生物標記技術,包括:
DNA 甲基化:評估基因表達與老化程度。
醣體學(Glycomics):分析免疫相關醣鏈變化。
代謝體學(Metabolomics)與脂質體學(Lipidomics):探索代謝健康與脂質代謝狀態。
這些技術可用來評估「生理年齡」與「實際年齡」之間的差異,對於預測疾病風險與制定個人化預防策略極具潛力。
Review
Nat Rev Endocrinol
. 2018 Oct;14(10):576-590. doi: 10.1038/s41574-018-0059-4.
Inflammaging: a new immune-metabolic viewpoint for age-related diseases
Claudio Franceschi 1, Paolo Garagnani 2 3 4 5, Paolo Parini 3, Cristina Giuliani 6 7, Aurelia Santoro 2 8
Affiliations Expand
PMID: 30046148
Abstract
Ageing and age-related diseases share some basic mechanistic pillars that largely converge on inflammation. During ageing, chronic, sterile, low-grade inflammation - called inflammaging - develops, which contributes to the pathogenesis of age-related diseases. From an evolutionary perspective, a variety of stimuli sustain inflammaging, including pathogens (non-self), endogenous cell debris and misplaced molecules (self) and nutrients and gut microbiota (quasi-self). A limited number of receptors, whose degeneracy allows them to recognize many signals and to activate the innate immune responses, sense these stimuli. In this situation, metaflammation (the metabolic inflammation accompanying metabolic diseases) is thought to be the form of chronic inflammation that is driven by nutrient excess or overnutrition; metaflammation is characterized by the same mechanisms underpinning inflammaging. The gut microbiota has a central role in both metaflammation and inflammaging owing to its ability to release inflammatory products, contribute to circadian rhythms and crosstalk with other organs and systems. We argue that chronic diseases are not only the result of ageing and inflammaging; these diseases also accelerate the ageing process and can be considered a manifestation of accelerated ageing. Finally, we propose the use of new biomarkers (DNA methylation, glycomics, metabolomics and lipidomics) that are capable of assessing biological versus chronological age in metabolic diseases.
發炎老化是指隨著年齡增長,體內慢性低度發炎逐漸加劇的一種生理狀態。這種狀態常伴隨血液中發炎標記物濃度升高,與多種慢性病及老年虛弱狀況密切相關。雖然看似自然老化的一部分,但實際上可能是導致健康惡化的重要誘因。
研究指出,多種因素可能共同促成發炎老化的發生,包括:
基因易感性:某些遺傳背景可能使人更易出現慢性發炎。
中央型肥胖:內臟脂肪與發炎反應密切相關。
腸道通透性與菌群失衡:腸漏與腸道菌組改變可能引發免疫反應。
細胞衰老與NLRP3發炎體活化:老化細胞釋放發炎因子,促使免疫系統長期活化。
線粒體功能障礙與氧化壓力:能量代謝異常導致自由基生成,促進發炎。
免疫系統失調與慢性感染:長期感染會持續激活免疫系統。
越來越多證據顯示,發炎老化不僅是心血管疾病的風險因子,還可能是其成因之一。臨床試驗支持發炎與動脈粥樣硬化、心衰竭等心血管疾病之間的因果關聯。此外,發炎老化與其他常見慢性病(如糖尿病、慢性腎病、失智症)亦有關聯,但在這些情況中,是否抑制發炎能改善病情仍存在爭議。
發炎老化也與老年人常見的「虛弱症候群」和多重慢病密切相關,這些狀態不僅影響生活品質,也會干擾對治療的反應。研究者推測,發炎可能透過抑制生長因子、促進分解代謝、干擾體內平衡調節等機制,加速功能退化。未來的臨床試驗應聚焦於:早期調控發炎是否能預防或延緩虛弱與心血管病的發生。
Review
Nat Rev Cardiol
. 2018 Sep;15(9):505-522. doi: 10.1038/s41569-018-0064-2.
Inflammageing: chronic inflammation in ageing, cardiovascular disease, and frailty
Luigi Ferrucci 1, Elisa Fabbri 2
Affiliations Expand
PMID: 30065258
PMCID: PMC6146930
Abstract
Most older individuals develop inflammageing, a condition characterized by elevated levels of blood inflammatory markers that carries high susceptibility to chronic morbidity, disability, frailty, and premature death. Potential mechanisms of inflammageing include genetic susceptibility, central obesity, increased gut permeability, changes to microbiota composition, cellular senescence, NLRP3 inflammasome activation, oxidative stress caused by dysfunctional mitochondria, immune cell dysregulation, and chronic infections. Inflammageing is a risk factor for cardiovascular diseases (CVDs), and clinical trials suggest that this association is causal. Inflammageing is also a risk factor for chronic kidney disease, diabetes mellitus, cancer, depression, dementia, and sarcopenia, but whether modulating inflammation beneficially affects the clinical course of non-CVD health problems is controversial. This uncertainty is an important issue to address because older patients with CVD are often affected by multimorbidity and frailty - which affect clinical manifestations, prognosis, and response to treatment - and are associated with inflammation by mechanisms similar to those in CVD. The hypothesis that inflammation affects CVD, multimorbidity, and frailty by inhibiting growth factors, increasing catabolism, and interfering with homeostatic signalling is supported by mechanistic studies but requires confirmation in humans. Whether early modulation of inflammageing prevents or delays the onset of cardiovascular frailty should be tested in clinical trials.
嗜中性球是人體最前線的免疫細胞之一,主要透過吞噬作用來清除入侵病原體。然而,科學家發現嗜中性球還有另一套防禦機制:在受到活化時,它們會釋放出染色質與抗菌蛋白,形成一種網狀結構,稱為「細胞外捕網(Neutrophil Extracellular Traps, NETs)」。
這些NETs 能主動捕捉革蘭氏陽性與陰性細菌,並在網狀結構中釋放高濃度的抗菌蛋白,有效:
分解毒力因子:削弱病原體的致病能力。
限制感染擴散:固定病原體於局部,防止其在體內擴散。
提升局部免疫濃度:集中抗菌分子,提高殺菌效率。
這讓NETs 成為急性感染中一種快速且有效的免疫策略。
NETs 的存在不僅是實驗室觀察到的現象,研究發現它們廣泛存在於實驗性痢疾與人類闌尾炎這兩種急性發炎疾病中。這代表 NETs 是身體對於細菌感染的自然反應之一,且具有重要的臨床意涵。
了解 NETs 的生理角色,有助於我們深入掌握免疫系統如何調節與對抗細菌感染,並可能為未來發炎性疾病的治療提供新方向。例如,若NETs 過度活化,可能對組織造成傷害,因此掌握其平衡機制對於免疫疾病管理至關重要。
Science
. 2004 Mar 5;303(5663):1532-5. doi: 10.1126/science.1092385.
Neutrophil extracellular traps kill bacteria
Volker Brinkmann 1, Ulrike Reichard, Christian Goosmann, Beatrix Fauler, Yvonne Uhlemann, David S Weiss, Yvette Weinrauch, Arturo Zychlinsky
Affiliations Expand
PMID: 15001782
Free article
Abstract
Neutrophils engulf and kill bacteria when their antimicrobial granules fuse with the phagosome. Here, we describe that, upon activation, neutrophils release granule proteins and chromatin that together form extracellular fibers that bind Gram-positive and -negative bacteria. These neutrophil extracellular traps (NETs) degrade virulence factors and kill bacteria. NETs are abundant in vivo in experimental dysentery and spontaneous human appendicitis, two examples of acute inflammation. NETs appear to be a form of innate response that binds microorganisms, prevents them from spreading, and ensures a high local concentration of antimicrobial agents to degrade virulence factors and kill bacteria.
阿茲海默症長期被認為是神經退化導致的疾病,但越來越多研究指出,發炎反應與免疫系統活化也是其核心病理之一。嗜中性球,作為先天免疫系統的重要成員,正逐漸被證實與阿茲海默症的進展有密切關聯。
實驗顯示,在阿茲海默症動物模型中,當體內累積的Aβ42胜肽出現時,會促使嗜中性球表面的LFA-1整合素進入高親和狀態,使嗜中性球迅速附著於血管內皮並穿越進入腦組織。進入中樞神經系統後,嗜中性球釋放出:
細胞外捕網(NETs)
發炎性細胞激素 IL-17
這些物質會加劇神經發炎並損傷神經元,進一步導致認知功能下降。
研究發現,暫時性耗盡嗜中性球或封鎖LFA-1路徑的小鼠模型,不僅展現較少的腦部發炎與類澱粉蛋白沉積,還在已出現認知障礙的階段出現記憶改善現象。更值得注意的是:
缺乏LFA-1的小鼠完全避免了認知退化
膠質細胞增生(gliosis)顯著減少
這指出,LFA-1可能是一個可行的治療標靶。
在人類阿茲海默症患者的腦組織中,同樣觀察到嗜中性球穿越血腦屏障、進入腦部,並與NETs共同存在於發炎區域。這強化了嗜中性球與阿茲海默症發病機制之間的因果關係。
結論:本研究開啟了利用免疫調節方式治療阿茲海默症的可能性。針對LFA-1或嗜中性球活性的藥物,或可為未來記憶退化的延緩提供新方向。
Nat Med
. 2015 Aug;21(8):880-6. doi: 10.1038/nm.3913. Epub 2015 Jul 27.
Neutrophils promote Alzheimer's disease-like pathology and cognitive decline via LFA-1 integrin
Elena Zenaro 1, Enrica Pietronigro 1, Vittorina Della Bianca 1, Gennj Piacentino 1, Laura Marongiu 1, Simona Budui 1, Ermanna Turano 2, Barbara Rossi 1, Stefano Angiari 1, Silvia Dusi 1, Alessio Montresor 3, Tommaso Carlucci 1, Sara Nanì 1, Gabriele Tosadori 3, Lucia Calciano 4, Daniele Catalucci 5, Giorgio Berton 1, Bruno Bonetti 2, Gabriela Constantin 3
Affiliations Expand
PMID: 26214837
DOI: 10.1038/nm.3913
Abstract
Inflammation is a pathological hallmark of Alzheimer's disease, and innate immune cells have been shown to contribute to disease pathogenesis. In two transgenic models of Alzheimer's disease (5xFAD and 3xTg-AD mice), neutrophils extravasated and were present in areas with amyloid-β (Aβ) deposits, where they released neutrophil extracellular traps (NETs) and IL-17. Aβ42 peptide triggered the LFA-1 integrin high-affinity state and rapid neutrophil adhesion to integrin ligands. In vivo, LFA-1 integrin controlled neutrophil extravasation into the CNS and intraparenchymal motility. In transgenic Alzheimer's disease models, neutrophil depletion or inhibition of neutrophil trafficking via LFA-1 blockade reduced Alzheimer's disease-like neuropathology and improved memory in mice already showing cognitive dysfunction. Temporary depletion of neutrophils for 1 month at early stages of disease led to sustained improvements in memory. Transgenic Alzheimer's disease model mice lacking LFA-1 were protected from cognitive decline and had reduced gliosis. In humans with Alzheimer's disease, neutrophils adhered to and spread inside brain venules and were present in the parenchyma, along with NETs. Our results demonstrate that neutrophils contribute to Alzheimer's disease pathogenesis and cognitive impairment and suggest that the inhibition of neutrophil trafficking may be beneficial in Alzheimer's disease.
慢性腦部低灌流是指大腦長期處於血流供應不足的狀態,無法滿足神經元正常運作所需的氧氣與營養。這種持續性的血流減少與多種神經退化性疾病密切相關,特別是在血管性認知障礙(VCI)中的作用日益受到重視。研究指出,CCH是引發大腦病理變化的主要推手,尤其會影響認知功能與記憶力的區域。
當大腦長時間缺乏足夠血液供應時,會引起一系列病理變化,包括:
白質病變:慢性缺血會導致大腦白質區域的結構受損,影響神經訊息的傳遞效率。
神經元細胞死亡:血氧與葡萄糖供應減少會導致神經元功能障礙並最終凋亡。
發炎反應加劇:低灌流狀態會誘發慢性發炎,進一步損害神經結構。
這些變化不僅影響記憶力、注意力與執行功能,還會加速認知退化,導致從輕度認知障礙發展為失智症。
儘管CCH對大腦健康影響重大,但透過早期偵測與適當干預,有可能延緩VCI的發展。目前研究探索的方向包括:
改善腦血流的藥物治療:如降血壓與抗血栓藥物有助於維持腦部血液循環。
促進神經修復的營養補充與神經保護劑。
生活方式干預:規律運動、健康飲食與管理慢性疾病(如高血壓、糖尿病)均有助於降低CCH風險。
早期影像檢測與認知評估:有助於即時發現低灌流現象並介入治療。
將CCH作為血管性認知障礙的早期標誌物,代表我們有機會將治療策略從症狀控制轉為預防性干預。這一轉變有望為未來開發改變病程的療法鋪路,使得人們在出現明顯認知障礙前就能及早介入,延緩甚至預防失智症的發生。
Acta Neuropathol Commun. 2023 Jun 12;11:93. doi: 10.1186/s40478-023-01590-1
Chronic cerebral hypoperfusion: a critical feature in unravelling the etiology of vascular cognitive impairment
Vismitha Rajeev 1,2, Yuek Ling Chai 1,2,#, Luting Poh 1,2,#, Sharmelee Selvaraji 1,2,3,#, David Y Fann 1, Dong-Gyu Jo 4, T Michael De Silva 5, Grant R Drummond 5, Christopher G Sobey 5, Thiruma V Arumugam 4,5, Christopher P Chen 1,2,6, Mitchell K P Lai 1,2,6,✉
Author information
Article notes
Copyright and License information
PMCID: PMC10259064 PMID: 37309012
Abstract
Vascular cognitive impairment (VCI) describes a wide spectrum of cognitive deficits related to cerebrovascular diseases. Although the loss of blood flow to cortical regions critically involved in cognitive processes must feature as the main driver of VCI, the underlying mechanisms and interactions with related disease processes remain to be fully elucidated. Recent clinical studies of cerebral blood flow measurements have supported the role of chronic cerebral hypoperfusion (CCH) as a major driver of the vascular pathology and clinical manifestations of VCI. Here we review the pathophysiological mechanisms as well as neuropathological changes of CCH. Potential interventional strategies for VCI are also reviewed. A deeper understanding of how CCH can lead to accumulation of VCI-associated pathology could potentially pave the way for early detection and development of disease-modifying therapies, thus allowing preventive interventions instead of symptomatic treatments.
Keywords: Vascular dementia, Neuronal cell death, Chronic cerebral hypoperfusion, White matter lesions
血管性認知障礙(VCI)涵蓋從輕度認知缺損到完全失智的多種表現,其共同特徵是由腦血管病變所引起。VCI常與阿茲海默症(AD)同時出現,形成混合型失智症,這類混合型病變現已成為年齡相關認知障礙的主要原因之一。
腦血管不僅是大腦氧氣與營養的供應系統,更扮演著訊號傳遞的角色,維持神經元與膠質細胞的健康。當血管受損,這些細緻的神經血管互動也隨之崩解,成為失智發展的基礎。
大腦半球白質是訊息傳導的高速通道,但由於其血液供應系統細小且脆弱,當血流供應不足或小血管病變發生時,白質極易發生:
白質病變(leukoaraiosis)
微小梗塞與微出血
慢性發炎反應
這些變化會中斷大腦各區域的連結,影響記憶、執行功能與處理速度等高階認知功能,進而導致認知障礙與失智。
血管異常不僅自身會導致認知功能下降,還會加速阿茲海默症的進展。具體表現包括:
血腦屏障破壞促使類澱粉蛋白沉積;
慢性缺血導致神經元能量代謝失衡;
微血管炎症促進Tau蛋白異常磷酸化。
因此,血管與神經退化性路徑彼此交織,加速病情惡化。
研究強調,預防失智的關鍵時機是中年期,而非老年期。控制以下血管危險因子尤為重要:
高血壓
糖尿病
高膽固醇
吸菸與缺乏運動
透過早期的健康管理,能有效降低血管損傷機率,進而延緩或預防晚年認知衰退。
Neuron. Author manuscript; available in PMC: 2014 Nov 20.
Published in final edited form as: Neuron. 2013 Nov 20;80(4):10.1016/j.neuron.2013.10.008. doi: 10.1016/j.neuron.2013.10.008
Author information
Copyright and License information
PMCID: PMC3842016 NIHMSID: NIHMS531326 PMID: 24267647
The publisher's version of this article is available at Neuron
Vascular cognitive impairment defines alterations in cognition, ranging from subtle deficits to full-blown dementia, attributable to cerebrovascular causes. Often coexisting with Alzheimer’s disease, mixed vascular and neurodegenerative dementia has emerged as the leading cause of age-related cognitive impairment. Central to the disease mechanism is the crucial role that cerebral blood vessels play in brain health, not only for the delivery of oxygen and nutrients, but also for the trophic signaling that links inextricably the well being of neurons and glia to that of cerebrovascular cells. This review will examine how vascular damage disrupts these vital homeostatic interactions, focusing on the hemispheric white matter, a region at heightened risk for vascular damage, and on the interplay between vascular factors and Alzheimer’s disease. Finally, preventative and therapeutic prospects will be examined, highlighting the importance of midlife vascular risk factor control in the prevention of late-life dementia.
阿茲海默症(Alzheimer’s Disease, AD)傳統上被視為以類澱粉蛋白沉積與Tau蛋白變異為主的神經退化性疾病。然而,最新研究強調:AD並非單一病因造成,而是涉及多重因素的異質性疾病。其中,血管功能障礙正迅速成為被關注的核心病理之一。
血管異常,包括腦血流不足與血腦屏障破損,可能在症狀出現前多年就已開始發展,並與神經元損傷密切相關,甚至可能促進類澱粉與Tau的病變擴散。
多項研究指出,以下血管問題在AD早期即已存在:
血腦屏障(BBB)破壞:使有害物質進入大腦,導致慢性發炎與神經毒性。
腦血流減少:降低氧氣與營養供應,加速神經元退化。
小血管病變:與腦白質病變及微出血密切相關,這些是超過一半失智症患者共同的影像特徵。
這些血管異常與傳統的神經退化機制密切交織,共同推動認知功能惡化。
目前已有多種MRI技術與影像序列可應用於臨床與研究,例如:
動態對比增強MRI:評估血腦屏障的滲漏程度。
動脈自旋標記(ASL)MRI:無需注射顯影劑即可量測腦血流。
高解析度T2與SWI序列:可偵測微小血管病變與微出血。
這些影像技術簡單易行,能早期發現與AD相關的血管異常,具有高潛力作為預測與診斷工具。
目前AD的研究與診斷主要聚焦於類澱粉蛋白與Tau蛋白標誌,然而整合血管生物標誌能為我們提供更全面的疾病理解。這種跨領域結合將有助於:
發現早期變化並提前介入;
精準分型,提升個別化治療的準確性;
促進新藥研發方向轉向血管保護與修復。
Alzheimers Dement
. 2019 Jan;15(1):158-167. doi: 10.1016/j.jalz.2018.07.222.
Vascular dysfunction-The disregarded partner of Alzheimer's disease
Melanie D Sweeney 1, Axel Montagne 1, Abhay P Sagare 1, Daniel A Nation 2, Lon S Schneider 3, Helena C Chui 4, Michael G Harrington 5, Judy Pa 6, Meng Law 7, Danny J J Wang 6, Russell E Jacobs 1, Fergus N Doubal 8, Joel Ramirez 9, Sandra E Black 10, Maiken Nedergaard 11, Helene Benveniste 12, Martin Dichgans 13, Costantino Iadecola 14, Seth Love 15, Philip M Bath 16, Hugh S Markus 17, Rustam Al-Shahi Salman 8, Stuart M Allan 18, Terence J Quinn 19, Rajesh N Kalaria 20, David J Werring 21, Roxana O Carare 22, Rhian M Touyz 23, Steve C R Williams 24, Michael A Moskowitz 25, Zvonimir S Katusic 26, Sarah E Lutz 27, Orly Lazarov 27, Richard D Minshall 28, Jalees Rehman 29, Thomas P Davis 30, Cheryl L Wellington 31, Hector M González 32, Chun Yuan 33, Samuel N Lockhart 34, Timothy M Hughes 34, Christopher L H Chen 35, Perminder Sachdev 36, John T O'Brien 37, Ingmar Skoog 38, Leonardo Pantoni 39, Deborah R Gustafson 40, Geert Jan Biessels 41, Anders Wallin 42, Eric E Smith 43, Vincent Mok 44, Adrian Wong 45, Peter Passmore 46, Frederick Barkof 47, Majon Muller 48, Monique M B Breteler 49, Gustavo C Román 50, Edith Hamel 51, Sudha Seshadri 52, Rebecca F Gottesman 53, Mark A van Buchem 54, Zoe Arvanitakis 55, Julie A Schneider 55, Lester R Drewes 56, Vladimir Hachinski 57, Caleb E Finch 58, Arthur W Toga 59, Joanna M Wardlaw 8, Berislav V Zlokovic 60
Affiliations Expand
PMID: 30642436
PMCID: PMC6338083
Erratum in
[No authors listed]
Alzheimers Dement. 2022 Mar;18(3):522. doi: 10.1002/alz.12483. Epub 2022 Feb 2.
PMID: 35112491 No abstract available.
Abstract
Increasing evidence recognizes Alzheimer's disease (AD) as a multifactorial and heterogeneous disease with multiple contributors to its pathophysiology, including vascular dysfunction. The recently updated AD Research Framework put forth by the National Institute on Aging-Alzheimer's Association describes a biomarker-based pathologic definition of AD focused on amyloid, tau, and neuronal injury. In response to this article, here we first discussed evidence that vascular dysfunction is an important early event in AD pathophysiology. Next, we examined various imaging sequences that could be easily implemented to evaluate different types of vascular dysfunction associated with, and/or contributing to, AD pathophysiology, including changes in blood-brain barrier integrity and cerebral blood flow. Vascular imaging biomarkers of small vessel disease of the brain, which is responsible for >50% of dementia worldwide, including AD, are already established, well characterized, and easy to recognize. We suggest that these vascular biomarkers should be incorporated into the AD Research Framework to gain a better understanding of AD pathophysiology and aid in treatment efforts.
Keywords: Alzheimer's disease; Biomarkers; Blood-brain barrier; Cerebral blood flow; MRI; Vascular.
Copyright © 2018 the Alzheimer's Association. Published by Elsevier Inc. All rights reserved.
Figures