TWI839714B - Use of the composition for preparing a medicament for treating traumatic brain injury - Google Patents

Use of the composition for preparing a medicament for treating traumatic brain injury Download PDF

Info

Publication number
TWI839714B
TWI839714B TW111115290A TW111115290A TWI839714B TW I839714 B TWI839714 B TW I839714B TW 111115290 A TW111115290 A TW 111115290A TW 111115290 A TW111115290 A TW 111115290A TW I839714 B TWI839714 B TW I839714B
Authority
TW
Taiwan
Prior art keywords
ccl5
mice
injury
cells
brain
Prior art date
Application number
TW111115290A
Other languages
Chinese (zh)
Other versions
TW202342502A (en
Inventor
周思怡
何文孝
顏嘉宏
蔣永孝
巴利 霍夫曼
張文昌
Original Assignee
臺北醫學大學
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 臺北醫學大學 filed Critical 臺北醫學大學
Priority to TW111115290A priority Critical patent/TWI839714B/en
Publication of TW202342502A publication Critical patent/TW202342502A/en
Application granted granted Critical
Publication of TWI839714B publication Critical patent/TWI839714B/en

Links

Landscapes

  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

本發明提出一種組成物用於製備治療創傷性腦損傷之藥物的用途,該組成物包括CCL5,用以調節大腦海馬迴的過氧化物(Reactive oxygen species,ROS)含量。The present invention provides a composition for preparing a drug for treating traumatic brain injury. The composition includes CCL5 and is used to regulate the content of reactive oxygen species (ROS) in the hippocampus of the brain.

Description

組成物用於製備治療創傷性腦損傷之藥物的用途Use of the composition for preparing a drug for treating traumatic brain injury

本發明係有關於一種組成物用於製備治療疾病之藥物的用途,特別是指一種組成物用於製備治療創傷性腦損傷之藥物的用途。The present invention relates to the use of a composition for preparing a drug for treating a disease, and in particular to the use of a composition for preparing a drug for treating traumatic brain injury.

創傷性腦損傷(Traumatic Brain Injury,TBI)是指頭部受到一次或多次的外力撞擊,如車禍、跌倒、運動傷害或激烈的搖晃後造成的腦功能損害。在一些病理學上的研究指出,每年有超過數百萬人因為創傷性腦損傷而求診,這些傷害可以透過簡易外傷分數(Abbreviated Injury Score,AIS)或是昏迷指數 (Glasgow Coma Scale,GCS)來評估腦損傷的嚴重程度。前者是透過電腦斷層(Computed Tomography,CT)的影像或核磁共振(Magnetic Resonance Imaging,MRI)的影像做分析;後者則是透過病人的眼睛、說話以及運動反應作為分析依據。Traumatic brain injury (TBI) refers to brain function damage caused by one or more external impacts to the head, such as car accidents, falls, sports injuries or violent shaking. Some pathological studies have shown that more than millions of people seek medical treatment for traumatic brain injuries every year. The severity of these injuries can be assessed by the Abbreviated Injury Score (AIS) or the Glasgow Coma Scale (GCS). The former is analyzed through images of computed tomography (CT) or magnetic resonance imaging (MRI); the latter is analyzed based on the patient's eyes, speech and motor reactions.

以GCS為例:13-15分為輕度TBI、9-12分為中度TBI、3-8分為重度TBI。而在台灣,2007-2008年期間就有超過90000個TBI的個案發生,當中有超過50%的人是輕度TBI。在最近的一些研究也發現輕度TBI後會增加很多神經退化性疾病的發病風險,例如:阿茲海默症(Alzheimer Disease,AD)、帕金森氏症(Parkinson's Disease,PD)等等,因此輕微TBI是一個非常需要重視的臨床腦部疾病。Take GCS as an example: 13-15 points are mild TBI, 9-12 points are moderate TBI, and 3-8 points are severe TBI. In Taiwan, there were more than 90,000 TBI cases in 2007-2008, and more than 50% of them were mild TBI. Some recent studies have also found that mild TBI increases the risk of many neurodegenerative diseases, such as Alzheimer's Disease (AD), Parkinson's Disease (PD), etc. Therefore, mild TBI is a clinical brain disease that needs to be taken seriously.

TBI 後會對大腦造成兩種傷害,分別是初級傷害以及二級傷害。初級傷害是指在腦受傷的過程中對腦組織造成的直接傷害,例如:因為外力而導致神經細胞與軸突撕裂、血腦屏障(Brain blood barrier (BBB) breakdown)的破壞、突觸損傷以及腦部血管出血;二級傷害則是指因為傷害後造成了生理以及生化層面上的變化而隨之而來的傷害。例如:因為TBI破壞了BBB造成腦出血,腦出血會造成組織缺氧(hypoxia)近而導致細胞內氧化壓力(Oxidative stress)上升造成傷害,或是TBI造成的腦壓的改變並使星狀膠質細胞腫大(astrocyte swelling),令星狀膠質細胞無法吸收組織內過多的谷氨酸(glutamate),對神經細胞造成興奮性毒害(excitotoxic damage),活化NADPH oxidase 產生大量過氧化物(Reactive oxygen species,ROS)造成氧化傷害。神經細胞內的細胞質ROS上升會對細胞造成直接的傷害,這些傷害主要是ROS氧化細胞內的蛋白質、核酸以及脂質導致這些分子失去正常功能。TBI causes two types of damage to the brain: primary damage and secondary damage. Primary damage refers to direct damage to brain tissue during the process of brain injury, such as tearing of nerve cells and axons, breakdown of the blood-brain barrier (BBB), synaptic injury, and bleeding of brain blood vessels caused by external forces; secondary damage refers to damage caused by changes in the physiological and biochemical levels after the injury. For example, TBI damages the BBB and causes cerebral hemorrhage, which causes tissue hypoxia and increases oxidative stress in cells, causing damage. Alternatively, TBI causes changes in brain pressure and astrocyte swelling, which prevents astrocytes from absorbing excess glutamate in the tissue, causing excitotoxic damage to nerve cells and activating NADPH oxidase to produce a large amount of reactive oxygen species (ROS), causing oxidative damage. Increased cytoplasmic ROS in nerve cells can cause direct damage to cells, which is mainly caused by ROS oxidizing proteins, nucleic acids, and lipids in cells, causing these molecules to lose their normal functions.

細胞內氧化壓力的來源有三個,分別是:細胞質 (cytoplasmic)、粒線體(mitochondria)以及過氧化體(peroxisome)。細胞質中的ROS在訊息傳遞以及疾病生理學中扮演很重要的角色,而細胞質中主要的ROS生產者NADPH oxidase (NOX) 家族,尤其是NOX2。 NOX2是超氧陰離子(superoxide,O 2)主要生產者,NOX2會把NADPH的電子傳遞到氧(O 2)以形成O 2。NOX2會參與於不同的代謝以及疾病的進程中,例如: 發炎反應中嗜中性球的活化、磷酸果糖激酶(phosphofructokinase)會與NOX2結合並刺激發炎反應。NOX的活性會隨著TBI後的不同時間點而改變,在TBI的初期在神經細胞內的NOX活性會上升,而TBI的後期小膠質細胞的NOX活性會上升。 There are three sources of intracellular oxidative stress: cytoplasmic, mitochondria, and peroxisome. ROS in the cytoplasm play an important role in signal transmission and disease physiology, and the main ROS producer in the cytoplasm is the NADPH oxidase (NOX) family, especially NOX2. NOX2 is the main producer of superoxide (O 2 ), and NOX2 transfers the electrons of NADPH to oxygen (O 2 ) to form O 2 . NOX2 is involved in different metabolisms and disease processes, such as: activation of neutrophils in inflammatory responses, and phosphofructokinase binds to NOX2 and stimulates inflammatory responses. The activity of NOX changes at different time points after TBI. In the early stage of TBI, the activity of NOX in nerve cells increases, while in the late stage of TBI, the activity of NOX in microglia increases.

除了O 2,細胞內另外兩個常見的ROS為Hydrogen peroxide (H 2O 2)以及Hydroxyl radical ( .OH)。H 2O 2的主要來源是由於O 2被超氧化物歧化酶(superoxide dismutase,SOD)分解後所產生,雖然H 2O 2能造成的氧化傷害比O 2來得低,但它半衰期比O 2來得長而且能輕易穿越細胞膜,因此也能對細胞造成氧化傷害。除此之外,若細胞內累積過多的H 2O 2,會增加Fenton reaction產生最具氧化能力的 .OH。Fenton reaction是細胞中的二價鐵離子(Fe 2+)透個與H 2O 2作電子交換以形成三價鐵(Fe 3+)、 .OH以及OH -。. In addition to O 2 , the other two common ROS in cells are Hydrogen peroxide (H 2 O 2 ) and Hydroxyl radical ( . OH). The main source of H 2 O 2 is the decomposition of O 2 by superoxide dismutase (SOD). Although H 2 O 2 can cause less oxidative damage than O 2 , its half-life is longer than O 2 and it can easily pass through the cell membrane, so it can also cause oxidative damage to cells. In addition, if too much H 2 O 2 accumulates in the cell, it will increase the Fenton reaction to produce the most oxidative . OH. The Fenton reaction is a process in which divalent iron ions (Fe 2+ ) in cells exchange electrons with H 2 O 2 to form trivalent iron (Fe 3+ ), OH , and OH - .

為了避免神經細胞因氧化壓力而死亡,神經細胞會在TBI後增加抗氧化物的表現量。TBI後主要會產生兩種ROS,分別是O 2以及 H 2O 2; 而主要負責分解這兩種ROS的抗氧化物是超氧化物歧化酶(superoxide dismutase,SOD)、過氧化氫酶(catalase,CAT)和穀胱甘肽過氧化物酶(glutathione peroxidase,GPX),這三種酶能夠一步步把O 2以及 H 2O 2最終轉化成沒有毒性的H ­2O以及O 2。SOD是重要的解毒酶亦是三個抗氧化物裹面第一個接觸的ROS的,可催化O 2轉化成兩個分子—H 2O 2以及O 2。CAT會利用鐵(iron)或是錳(manganese)離子作為輔助因子(cofactor)並催化H 2O 2還原成H 2O以及O 2,但在大腦中CAT對氧化壓力的靈敏度並不高,因此CAT在大腦的活性相對較低。GPX是一個很重要的酶把H 2O 2分解成H 2O同時也能把過氧化的脂質(lipid peroxides)分解成大腦相對應的醇類,而GPX的活性是需要取決於穀胱甘肽(glutathione,GSH)相關的代謝路徑。 In order to prevent nerve cells from dying due to oxidative stress, nerve cells will increase the expression of antioxidants after TBI. Two main types of ROS are produced after TBI, namely O 2 and H 2 O 2 ; the antioxidants mainly responsible for decomposing these two ROS are superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPX). These three enzymes can gradually convert O 2 and H 2 O 2 into non-toxic H 2 O and O 2 . SOD is an important detoxification enzyme and the first ROS to be contacted by the three antioxidants. It can catalyze the conversion of O 2 into two molecules - H 2 O 2 and O 2 . CAT uses iron or manganese ions as cofactors and catalyzes the reduction of H2O2 into H2O and O2 , but CAT is not very sensitive to oxidative stress in the brain, so CAT activity in the brain is relatively low. GPX is a very important enzyme that breaks down H2O2 into H2O and can also break down lipid peroxides into corresponding alcohols in the brain. The activity of GPX depends on the metabolic pathway related to glutathione (GSH).

GSH的代謝路徑包括還原態(reduced form)的GSH、氧化態GSH(Glutathione disulfide,GSSG)以及兩個協同的酶分別是穀胱甘肽還原酶(glutathione reductase,GR)與GPX以維持氧化還原的平衡。 NADPH是GSH電子傳遞中最重要的電子提供者。在神經元中,通過PPP(pentose phosphate pathway, PPP)氧化葡萄糖,以維持足夠水平的NADPH用於抗氧化活性。在正常的細胞中,GSH與GSSG比例大慨是90%,但當細胞出現老化、慢性發炎以及氧化傷害時,GSH與GSSG的比例可能會降至50% 甚至是低於50%。The metabolic pathway of GSH includes reduced form of GSH, oxidized form of GSH (Glutathione disulfide, GSSG) and two synergistic enzymes, glutathione reductase (GR) and GPX, to maintain the balance of redox. NADPH is the most important electron donor in GSH electron transfer. In neurons, glucose is oxidized through PPP (pentose phosphate pathway, PPP) to maintain sufficient levels of NADPH for antioxidant activity. In normal cells, the ratio of GSH to GSSG is about 90%, but when cells age, suffer from chronic inflammation and oxidative damage, the ratio of GSH to GSSG may drop to 50% or even lower than 50%.

因此,本發明是研究TBI後於腦中產生的ROS調節機制,以提供預防和治療此類疾病的有效藥物或療法。Therefore, the present invention is to study the regulatory mechanism of ROS generated in the brain after TBI in order to provide effective drugs or therapies for the prevention and treatment of such diseases.

為了達成上述目的,本發明提供一種組成物用於製備治療創傷性腦損傷之藥物的用途,該組成物包括CCL5,用以調節大腦海馬迴受傷後所產生的過氧化物(Reactive oxygen species,ROS)含量。To achieve the above object, the present invention provides a composition for preparing a drug for treating traumatic brain injury, wherein the composition includes CCL5 for regulating the level of reactive oxygen species (ROS) produced after injury to the hippocampus.

在本發明之一實施例中,其中該創傷性腦損傷尤指輕微創傷性腦損傷(mild TBI)。In one embodiment of the present invention, the traumatic brain injury is particularly mild traumatic brain injury (mild TBI).

在本發明之一實施例中,其中該組成物用於活化穀胱甘肽過氧化物酶1(Glutathione peroxidase 1, GPX1)。In one embodiment of the present invention, the composition is used to activate glutathione peroxidase 1 (GPX1).

在本發明之一實施例中,其中該組成物用於調節海馬迴的慢性發炎。In one embodiment of the present invention, the composition is used to regulate chronic inflammation of the hippocampus.

在本發明之一實施例中,其中該組成物於腦損害發生後3天內投予。In one embodiment of the present invention, the composition is administered within 3 days after the brain damage occurs.

在本發明之一實施例中,其中該組成物於腦損害發生後1小時內投予。In one embodiment of the present invention, the composition is administered within 1 hour after the brain damage occurs.

在本發明之一實施例中,其中該組成物可減緩創傷性腦損傷的神經元細胞氧化壓力,降低受氧化壓力傷害的細胞數。In one embodiment of the present invention, the composition can alleviate the oxidative stress of neurons in traumatic brain injury and reduce the number of cells damaged by oxidative stress.

有關本發明的詳細說明和技術內容,配合圖式說明如下,然而所附圖式僅提供參考與說明用,以幫助理解本發明,非用以限制本發明之範圍。The detailed description and technical contents of the present invention are described below with reference to the accompanying drawings. However, the attached drawings are only provided for reference and explanation to help understand the present invention and are not intended to limit the scope of the present invention.

首先,說明本發明的實驗材料與方法:First, the experimental materials and methods of the present invention are described:

一、輕度 TBI 的動物實驗:1. Animal Experiments on Mild TBI:

從國家實驗動物中心購入八週齡雄性C57BL/6Jnarl小鼠(以下稱WT)、從國外傑克遜實驗室(The Jackson Laboratory)購入CCL5基因剃除小鼠(Stock No: 005090,以下稱CCL5-KO或CCL5 -/-),並於國家動物中心繁殖培養。實驗採用八週齡雄性CCL5基因剃除小鼠。小鼠以每籠 3-5隻為一組飼養,在 12:12 小時的光/暗循環中自由獲取食物和水,室溫設定為 25°C,研究中的所有行為測試也在 25°C 下進行。 Eight-week-old male C57BL/6Jnarl mice (hereinafter referred to as WT) were purchased from the National Animal Center, and CCL5-KO mice (Stock No: 005090, hereinafter referred to as CCL5-KO or CCL5 -/- ) were purchased from The Jackson Laboratory and bred and cultured at the National Animal Center. Eight-week-old male CCL5-KO mice were used in the experiment. Mice were housed in groups of 3-5 per cage, with free access to food and water in a 12:12 h light/dark cycle, and the room temperature was set at 25°C. All behavioral tests in the study were also performed at 25°C.

2-3 個月大的 WT 或CCL5-KO用 2.5% 異氟醚(isoflurane , Panion & BF biotech Inc.)以 1.5–2.0 L/min 的空氣流速持續2 分鐘 15 秒以麻醉小鼠;隨後,將小鼠胸部朝下放在泡沫海綿上(尺寸:18.5 cm × 7 cm × 8 cm),用以在重物落下(weight drop)裝置下方支撐頭部和身體。重物落下裝置由一個空心圓柱形管(內徑 1.2 公分,高 100公分)組成,垂直放置在小鼠頭部中心上方約 2公分處。將 30 克重物(直徑 1公分,高 5.2公分)從空心圓柱形管中釋放落下並撞擊小鼠以誘發輕度 TBI。在小鼠受到衝擊後到小鼠能將身體翻正反射這段時間為小鼠 “失去意識時間”。2-3 month old WT or CCL5-KO mice were anesthetized with 2.5% isoflurane (isoflurane, Panion & BF biotech Inc.) at an air flow rate of 1.5–2.0 L/min for 2 min 15 s; then, the mice were placed chest down on a foam sponge (size: 18.5 cm × 7 cm × 8 cm) to support the head and body under a weight drop device. The weight drop device consisted of a hollow cylindrical tube (1.2 cm inner diameter, 100 cm height) and was placed vertically approximately 2 cm above the center of the mouse's head. A 30 g weight (1 cm diameter, 5.2 cm height) was released from the hollow cylindrical tube and hit the mouse to induce mild TBI. The time from when the mouse was hit to when it was able to right itself reflexively was the mouse's "loss of consciousness time".

用 NAC(A7250,Sigma-Aldrich)或重組小鼠 CCL5/RANTES 蛋白(478-MR-025,R&D系統)處理 CCL5-KO 小鼠。NAC溶解在生理鹽水中,給藥劑量為5mg/kg或20mg/kg。輕度 TBI 後一小時,每天通過腹腔注射治療 CCL5-KO 小鼠,直到 4dpi (Days post injury,受傷後天數)進行行為測試。在受傷後的2天(2 dpi),進行記憶回溯測試後注射NAC,以避免NAC 對短期記憶回朔測試有所影響。CCL5 重組蛋白以 PBS 稀釋,並給予 CCL5-KO 小鼠300 μg/kg 劑量。為了追蹤 CCL5 的位置,CCL5 重組蛋白使用 Alexa Fluor™ 594 微量蛋白標記試劑套組(A30008,Invitrogen)進行標記。mTBI 後 24 小時,犧牲小鼠,並用anti-CCL5 抗體(1:100,sc-365826,Santa Cruz biotechnology,Dallas,USA)和donkey anti-rabbit-488(1:400,Invitrogen, A32790)進行免疫染色。CCL5-KO mice were treated with NAC (A7250, Sigma-Aldrich) or recombinant mouse CCL5/RANTES protein (478-MR-025, R&D Systems). NAC was dissolved in saline and administered at a dose of 5 mg/kg or 20 mg/kg. CCL5-KO mice were treated by intraperitoneal injection every day one hour after mild TBI until behavioral testing at 4 dpi (days post injury). NAC was injected after the memory retrieval test 2 days after injury (2 dpi) to avoid the effect of NAC on the short-term memory retrieval test. CCL5 recombinant protein was diluted in PBS and administered to CCL5-KO mice at a dose of 300 μg/kg. To track the location of CCL5, recombinant CCL5 protein was labeled using Alexa Fluor™ 594 Micro-Volume Protein Labeling Kit (A30008, Invitrogen). 24 h after mTBI, mice were sacrificed and immunostained with anti-CCL5 antibody (1:100, sc-365826, Santa Cruz biotechnology, Dallas, USA) and donkey anti-rabbit-488 (1:400, Invitrogen, A32790).

二、行為測試:2. Behavioral test:

神經功能包括幾個行為測試 -改良版神經缺損評分(mNSS):mNSS 用以表現輕度 TBI後 WT 和 CCL5-KO 小鼠的整體神經元功能,包括運動、感覺、反射和平衡。參數評分為 0 到 18(健康評分:0;最高缺陷評分:18)。記憶功能測試包括新奇事物認知測試(Novel Object Recognition Test,NOR)和巴德斯迷宮(Barnes maze)。所有測試均通過EthoVision ® XT軟體進行分析。 Neurological function included several behavioral tests - modified neurological deficit score (mNSS): mNSS was used to represent global neuronal function in WT and CCL5-KO mice after mild TBI, including movement, sensation, reflexes, and balance. Parameters were scored from 0 to 18 (healthy score: 0; maximum deficit score: 18). Memory function tests included the Novel Object Recognition Test (NOR) and the Barnes maze. All tests were analyzed using EthoVision ® XT software.

(一) 新奇事物認知測試1. Novelty Perception Test

新奇事物認知測試是利用小鼠會對新事物有興趣的特性而進行的物件認知行為測試,小鼠會比較傾向與新物件接觸,進而比較小鼠對新舊物件的探索時間,以分析小鼠記憶表現情形。小鼠被隨機分為四組,並在受傷後 4、7、14 和 28 天(dpi)做測試分析。第1天讓小鼠在封閉的箱子(57 cm × 57 cm箱子)中適應10分鐘,第2天再將2個相同的物體以相等距離放置在箱子中,並讓小鼠探索10分鐘。24 小時後,其中一個物體被替換為一個大小相同但形狀和顏色不同的物體,再讓小鼠探索10分鐘,並記錄。實驗利用EthoVision® XT 軟體記錄和分析小鼠行走軌跡。記憶能力被定義為辨別指數百分比(DI%):(新物件探索時間 - 熟悉物件探索時間)/總探索時間 x 100%。不同組中的物體組合相同,以避免物體形狀混淆。在試驗之間用乙醇清潔該裝置以避免氣味影響識別。The novel object recognition test is an object recognition behavior test that uses the characteristic of mice to be interested in new objects. Mice will tend to contact new objects, and then compare the exploration time of mice on new and old objects to analyze the memory performance of mice. Mice were randomly divided into four groups and tested and analyzed at 4, 7, 14 and 28 days after injury (dpi). On the first day, mice were allowed to adapt to a closed box (57 cm × 57 cm box) for 10 minutes. On the second day, two identical objects were placed in the box at equal distances and mice were allowed to explore for 10 minutes. After 24 hours, one of the objects was replaced with an object of the same size but different shape and color, and the mice were allowed to explore for another 10 minutes and recorded. The experiment used EthoVision® XT software to record and analyze the walking trajectory of mice. Memory performance was defined as the discrimination index percentage (DI%): (novel object exploration time - familiar object exploration time) / total exploration time x 100%. The object combinations were the same in different groups to avoid confusion of object shapes. The apparatus was cleaned with ethanol between trials to avoid odor affecting recognition.

(二) 巴德斯迷宮(II) Baders Labyrinth

迷宮由一個白色圓形平台(直徑 92 cm)組成,在 3、6、9 和 12 點鐘位置有等間距的孔。測試前將小鼠在黑暗盒子中熟悉 30 分鐘。第1天,小鼠適應逃生箱5分鐘,迷宮適應6分鐘。在接下來的 4 天裡,將老鼠放在迷宮的中心,讓牠們自由探索 6 分鐘。當他們到達逃生箱或探索超過 6 分鐘時,即停止測試。記憶回溯測試也如前所述進行。在巴德斯迷宮訓練的最後一天後的第 2 天和第 7 天進行短期記憶和長期記憶回溯測試。使用上述軟體測量訓練和記憶回溯測試期間的滯留期、移動距離和目標象限訪問百分比。在TBI前的巴德斯迷宮(-11 dpi 巴德斯迷宮)中,小鼠接受了訓練和記憶回溯測試,以確認牠們的學習和記憶能力。2dpi 記憶回溯測試(逃生箱的位置與 11 dpi 巴德斯迷宮的位置相同)表示 TBI 後 WT 和 CCL5-KO 小鼠的原始記憶受損。在受傷後的測試,會變更逃生箱的所在位置。The maze consisted of a white circular platform (92 cm in diameter) with equally spaced holes at the 3, 6, 9, and 12 o'clock positions. Mice were familiarized in a dark box for 30 min before testing. On day 1, mice were habituated to the escape box for 5 min and the maze for 6 min. On the following 4 days, mice were placed in the center of the maze and allowed to explore freely for 6 min. Testing was stopped when they reached the escape box or explored for more than 6 min. Memory recall tests were also performed as described previously. Short-term and long-term memory recall tests were performed on days 2 and 7 after the last day of training on the Bards maze. Retention period, distance moved, and percentage of target quadrant visits during training and memory recall tests were measured using the above software. Mice were trained and tested for memory recall in the Bards maze before TBI (-11 dpi Bards maze) to confirm their learning and memory abilities. The 2dpi memory recall test (the location of the escape box was the same as the 11 dpi Bards maze) indicated that primary memory was impaired in WT and CCL5-KO mice after TBI. In the post-injury test, the location of the escape box was changed.

在行為分析後不超過 24 小時內犧牲動物進行以下細胞和分子分析。因此,分子分析結果可與行為結果連結。Animals were sacrificed for the following cellular and molecular analyses no more than 24 hours after the behavioral analysis. Thus, the molecular analysis results could be linked to the behavioral results.

三、FJC染色3. FJC staining

利用 Zoletil 50 (66F4, Virbac) 和 Rompum 溶液 (PP1523, Bayer) 麻醉小鼠,然後用 4% PFA 灌注;將穫取的大腦以低溫恆溫器切片(厚度為25 μm)並進行 Fluoro-Jade C (FJC) 染色(TR-100-FJ,Biosensis,USA)。將腦組織固定在塗有明膠的載玻片上,並在 57°C 下加熱 30 分鐘。組織用連續稀釋的乙醇和水,以水化組織,然後用 0.06% KMnO 4避光靜置10分鐘。之後,將組織與含有 1 μg/ml DAPI與 0.0001% FJC 的溶液,避光靜置10分鐘進行呈色。組織用二甲苯(Xylene)脫水。使用螢光顯微鏡(STP6000,LEICA)獲取圖像,並用 Imager J 軟件分析 FJC 陽性細胞。 Mice were anesthetized with Zoletil 50 (66F4, Virbac) and Rompum solution (PP1523, Bayer) and then perfused with 4% PFA; the harvested brains were cryostat-sectioned (25 μm thick) and stained with Fluoro-Jade C (FJC) (TR-100-FJ, Biosensis, USA). Brain tissues were mounted on gelatin-coated slides and heated at 57°C for 30 min. The tissues were hydrated with serially diluted ethanol and water and then incubated with 0.06% KMnO 4 for 10 min in the dark. Afterwards, the tissues were incubated with a solution containing 1 μg/ml DAPI and 0.0001% FJC for 10 min in the dark for staining. Tissues were dehydrated with xylene. Images were acquired using a fluorescence microscope (STP6000, LEICA), and FJC-positive cells were analyzed using Imager J software.

四、免疫化學染色:IV. Immunochemical staining:

在傷後 1 天和 7 天進行 Hypoxyprobe 染色,以測量 TBI 後海馬迴的氧化壓力(Oxidative stress, OS) 損傷。在灌注之前,給動物施用60mg/kg腹腔注射的Hypoxyprobe溶液(溶解在0.9% NaCl中的鹽酸哌莫硝唑pimonidazole hydrochloride)。60 分鐘後,用 0.1 M 磷酸鹽緩衝液配置的4% 多聚甲醛(paraformaldehyde ,PFA)灌注小鼠。獲取的腦組織冷凍切片 (厚度為25 μm) 並進行免疫組織化學處理。把組織放在 0.1 M 磷酸鹽緩衝液中含有 1% 碳酸氫鈉(sodium bicarbonate)30 分鐘。在含有3% BSA 的PBSc/m 溶液處理作用後,將組織與一級抗體在 4°C 下靜置過夜。使用NeuN(Novus-NBP1-77686X,1:200 稀釋)、Iba-1(GeneTex、GTX101495, 1:1000 稀釋)和 GFAP(GeneTex, GTX108711, 1: 1000 稀釋度)的兔抗體測量神經元、微小膠細胞和星形膠細胞的數量。Anti-pimonidazole-FITC(1:100,Hypoxyprobe™ Plus 試劑套組,Biosensis)用於顯示 Hypoxyprobe訊號。Anti-NeuN 或 GFAP用以共同染色以標示有氧化傷害的神經元和星形膠質細胞。洗去一抗後,將組織與donkey anti-rabbit-568(1:400,Invitrogen,A10042)或donkey anti-rabbit-488(1:400,Invitrogen,A32790)在室溫下作用反應40 分鐘。通過螢光顯微鏡獲取圖像並使用 Image J 進行分析。對照組由省略一抗組成,並且由不知情的觀察者進行計數。Hypoxyprobe staining was performed 1 and 7 days after injury to measure oxidative stress (OS) damage in the hippocampus after TBI. Before perfusion, animals were given 60 mg/kg of Hypoxyprobe solution (pimonidazole hydrochloride dissolved in 0.9% NaCl) intraperitoneally. After 60 minutes, mice were perfused with 4% paraformaldehyde (PFA) in 0.1 M phosphate buffer. The obtained brain tissue was frozen and sectioned (thickness 25 μm) and processed for immunohistochemistry. The tissue was placed in 0.1 M phosphate buffer containing 1% sodium bicarbonate for 30 minutes. After treatment with 3% BSA in PBSc/m, tissues were incubated with primary antibodies overnight at 4°C. Rabbit antibodies against NeuN (Novus-NBP1-77686X, 1:200 dilution), Iba-1 (GeneTex, GTX101495, 1:1000 dilution), and GFAP (GeneTex, GTX108711, 1:1000 dilution) were used to measure the number of neurons, microglia, and astrocytes. Anti-pimonidazole-FITC (1:100, Hypoxyprobe™ Plus Kit, Biosensis) was used to visualize Hypoxyprobe signals. Anti-NeuN or GFAP was used for co-staining to mark neurons and astrocytes with oxidative damage. After washing away the primary antibody, the tissues were incubated with donkey anti-rabbit-568 (1:400, Invitrogen, A10042) or donkey anti-rabbit-488 (1:400, Invitrogen, A32790) for 40 min at room temperature. Images were acquired by fluorescence microscopy and analyzed using Image J. The control group consisted of omitting the primary antibody, and counting was performed by a blinded observer.

五、NADPH氧化酶活性測定:5. NADPH oxidase activity assay:

在 4dpi、7 dpi、14 dpi 和 28 dpi 研究組織內ROS基礎值與 NADPH 氧化酶活性。在體積為 200 μl Krebs 緩衝液(pH為7.4,118.4 mM氯化鈉, 25mM的NaHCO 3,11.7 mM葡萄糖,4.75 mM氯化鉀,1.2mM的MgSO 4,2.5mM的CaCl 2.2H 2O,1.2 mM KH 2PO 4),加入50 µM 光澤精(lucigenin)(Sigma-Aldrich, 2315971)反應,測量50 秒內所產生的ROS作為組織ROS基礎值。此後通過添加 50 µM NADPH 來測定 NADPH 氧化酶活性。每秒計數(CPS)由 Triathler Multilabel Test(425-004, HIDEX, Turku, Finland)測量,並用組織的濕重(µg)平均。NADPH 氧化酶活性以350 秒反應期間的ROS量曲線下面積進行分析。TBI 組的數據與對照組(sham group)比較,以測量 NADPH 氧化酶活性的倍數變化。 The basal ROS value and NADPH oxidase activity in tissues were studied at 4 dpi, 7 dpi, 14 dpi and 28 dpi. 50 µM lucigenin (Sigma-Aldrich, 2315971) was added to 200 μl Krebs buffer (pH 7.4, 118.4 mM NaCl, 25 mM NaHCO 3 , 11.7 mM glucose, 4.75 mM KCl, 1.2 mM MgSO 4 , 2.5 mM CaCl 2 .2H 2 O, 1.2 mM KH 2 PO 4 ) and the ROS generated within 50 seconds was measured as the basal ROS value in tissues. NADPH oxidase activity was then measured by adding 50 µM NADPH. Counts per second (CPS) were measured by Triathler Multilabel Test (425-004, HIDEX, Turku, Finland) and averaged with the wet weight of tissue (µg). NADPH oxidase activity was analyzed as the area under the curve of ROS during the 350-second reaction period. Data from the TBI group were compared with the sham group to measure the fold change in NADPH oxidase activity.

六、蛋白質西方墨點分析:VI. Protein Western Blot Analysis:

組織與細胞利用含有蛋白酶-磷酸酶抑製劑(78441,Thermo Scientific™,USA)的RIPA 緩衝液(#20-188,Millipore,USA)裂解。並以10% Tris-HCl蛋白凝膠進行蛋白質電泳;每個樣本均加入30 μg 總蛋白。凝膠通過 Trans-Blot ®Cell 系統 (Bio-Rad)分析並轉漬到 PVDF 膜上。轉漬好的蛋白膜用阻斷緩衝液(5% skim milk溶於含 0.05% Tween 20 的 Tris 緩衝鹽水,TBST)在室溫下作用 1 小時,然後與不同的一抗(包括anti-actin(Millipore,MAB1501)、GPX-1 (GeneTex, GTX116040)、SOD-1 (GeneTex, GTX100659) 和 SOD-2 (GeneTex, GTX116093) 在 4 °C 下反應一個晚上。將膜用 TBST 洗滌 3 次後在室溫下與 HRP結合的二抗(Goat anti-Mouse  IgG-HRP,115-035-003;Goat anti-Rabbit  IgG-HRP,111-035-003,來自 Jackson ImmunoResearch)作用 1 小時。蛋白質訊號利用 Clarity Western ECL Substrate 試劑(Bio-Rad, 1705061)顯現;蛋白表現量由 Image J 軟體進行量化。 Tissues and cells were lysed using RIPA buffer (#20-188, Millipore, USA) containing protease-phosphatase inhibitors (78441, Thermo Scientific™, USA). Protein electrophoresis was performed using 10% Tris-HCl protein gel; 30 μg of total protein was added to each sample. Gels were analyzed using the Trans-Blot ® Cell system (Bio-Rad) and transferred to PVDF membranes. The stained membrane was incubated with blocking buffer (5% skim milk in Tris-buffered saline containing 0.05% Tween 20, TBST) at room temperature for 1 hour, and then incubated with different primary antibodies (including anti-actin (Millipore, MAB1501), GPX-1 (GeneTex, GTX116040), SOD-1 (GeneTex, GTX100659) and SOD-2 (GeneTex, GTX116093) at 4 °C overnight. The membrane was washed three times with TBST and then incubated with HRP-conjugated secondary antibodies (Goat anti-Mouse IgG-HRP, 115-035-003; Goat anti-Rabbit IgG-HRP, 111-035-003, from Jackson ImmunoResearch) at room temperature for 1 hour. Protein signals were visualized using Clarity Western ECL Substrate reagent (Bio-Rad, 1705061), and protein expression was quantified using Image J software.

七、RNA分離與即時定量PCR分析:VII. RNA Isolation and Real-time Quantitative PCR Analysis:

通過 TRIZOL(15596018, Invitrogen)萃取組織 RNA,並使用 High-Capacity cDNA Reverse Transcription Kit (4368813, Applied Biosystems™)逆轉錄成 cDNA。使用 iTaq Universal SYBR Green Supermix(Bio-Rad)和 StepOnePlus™ Real-Time PCR System(4376600, Applied Biosystems™, USA)進行定量 PCR。引子序列為:Tissue RNA was extracted by TRIZOL (15596018, Invitrogen) and reverse transcribed into cDNA using High-Capacity cDNA Reverse Transcription Kit (4368813, Applied Biosystems™). Quantitative PCR was performed using iTaq Universal SYBR Green Supermix (Bio-Rad) and StepOnePlus™ Real-Time PCR System (4376600, Applied Biosystems™, USA). The primer sequences were:

GPX1:Forward—5'-CGTTTGAGTCCCAACATCTC-3'; reverse—5′-CGTTCATCTCGGTGTAGTCC-3′,產物大小:199 bp。CCL5:Forward—5'-TGCTGCTTTGCCTACCTC-3'; reverse—5′-CTTGAACCCACTTCTTCTCT-3′,產物大小:151 bp。IL-1β:Forward—5'-GCACTACAGGCTCCGAGATGAAC-3'; reverse—5′-TTGTCGTTGCTTGGTTCTCCTTGT-3′,產物大小:147 bp。IL-10:Forward—5'-GCTCTTACTGACTGGCATGAG-3';reverse—5'-CGCAGCTCTAGGAGCATGTG-3',產物大小:105 bp。TNF-α:Forward—5'-GGAACTGGCAGAAGAGGCACTC-3';reverse—5'-GCAGGAATGAGAAGAGGCTGAGAC-3',產物大小:89 bp。小鼠GADPH:Forward—5'-GTGTTCCTACCCCCAATGTGT-3';reverse—5'-AGAGTGGGAGTTGCTGTTGAAG-3',產物大小:176 bp。人類GADPH:Forward—5'- CACAAGAGGAAGAGAGAGA-3';reverse—5'- CACAGGGTACTTTATTGATG -3',產物大小:164 bp(用於 SHSY5Y 人母細胞瘤細胞系)。mRNA 的表達用 GADPH 標準化,並使用 2 -ΔCT(TBI)/2-ΔCT(sham)的百分比計算。 GPX1: Forward—5'-CGTTTGAGTCCCAACATCTC-3';reverse—5'-CGTTCATCTCGGTGTAGTCC-3', product size: 199 bp. CCL5: Forward—5'-TGCTGCTTTGCCTACCTC-3';reverse—5'-CTTGAACCCACTTCTTCTCT-3', product size: 151 bp. IL-1β: Forward—5'-GCACTACAGGCTCCGAGATGAAC-3';reverse—5'-TTGTCGTTGCTTGGTTCTCCTTGT-3', product size: 147 bp. IL-10: Forward—5'-GCTCTTACTGACTGGCATGAG-3';reverse—5'-CGCAGCTCTAGGAGCATGTG-3', product size: 105 bp. TNF-α: Forward—5'-GGAACTGGCAGAAGAGGCACTC-3';reverse—5'-GCAGGAATGAGAAGAGGCTGAGAC-3', product size: 89 bp. Mouse GADPH: Forward—5'-GTGTTCCTACCCCCAATGTGT-3';reverse—5'-AGAGTGGGAGTTGCTGTTGAAG-3', product size: 176 bp. Human GADPH: Forward—5'- CACAAGAGGAAGAGAGAGA-3';reverse—5'- CACAGGGTACTTTATTGATG -3', product size: 164 bp (for SHSY5Y human blastoma cell line). mRNA expression was normalized with GADPH and calculated using the percentage of 2 - ΔCT(TBI)/2-ΔCT(sham).

八、GSH/GSSG 活性測定8. GSH/GSSG activity assay

穀胱甘肽(Glutathione)濃度通過穀胱甘肽比色測定試劑組(K261,BioVision Inc,美國)測量。海馬迴組織在冰上用穀胱甘肽測定緩衝液(4μl/組織重量(mg))均質化。將 20 μl 樣品與 160 μl 反應混合物或還原反應混合物混合以分別檢測總 GSH 和還原態的 GSH 產量。GSH總產量,使用 iMark 微孔盤吸光度讀數器(Bio-Rad,USA)以415 nm波長測量樣品 30 分鐘。對於還原態的 GSH,在 415 nm 處測量樣品並使用標準穀胱甘肽標準曲線進行運算。Glutathione concentration was measured by Glutathione Colorimetric Assay Kit (K261, BioVision Inc, USA). Hippocampal tissues were homogenized with Glutathione Assay Buffer (4 μl/tissue weight (mg)) on ice. 20 μl of sample was mixed with 160 μl of reaction mixture or reduction reaction mixture to detect total GSH and reduced GSH production, respectively. For total GSH production, samples were measured at 415 nm for 30 min using an iMark microplate absorbance reader (Bio-Rad, USA). For reduced GSH, samples were measured at 415 nm and calculated using a standard glutathione standard curve.

九、細胞培養、轉染、細胞存活性測定:IX. Cell culture, transfection, and cell viability assay:

初代神經元由懷孕第 16.5-17 天 (E16.5-17)的C57BL/6 和 CCL5 -/-小鼠胚胎培養。胚胎腦組織用含有2 mg/ml 木瓜蛋白酶(Worthington,LS003119)和 0.05% Trypsin -EDTA(Gibco,25200-072)的Dulbecco's Modified Eagle 培養基緩衝液(DMEM,12800-017,Gibco)中作用分解14 分鐘,並使用含有 10% v/v 熱失活性的胎牛血清 (FBS, 10437-028, Gibco)、1% v/v 抗生素-抗真菌劑 (15240-062, Gibco)和 2 mM L-谷氨酰胺(25030,Gibco)的神經基礎培養液(neurobasal medium, 21103-049, Gibco)培養。待細胞附著 2 小時後,將培養液更換為“完全培養液”(成份為神經基礎培養液,並含有 1% v/v N-2 補充劑(17502048,Gibco),2% v/v B-27 補充劑(17504044,Gibco),1% v/ v 抗生素-抗真菌劑和 2 mM L-谷氨酰胺)。之後,每 3 天更換一半培養液。在培養 第4天時先給予細胞10、50、100 和 250 pg/ml濃度的CCL5/RANTES重組蛋白。30 分鐘後,再給與細胞25、50、100、250、500 μM濃度的過氧化氫(PanReac AppliChem, 131077) 24 小時。在細胞株研究中,實驗利用 Lipofectamine™ 2000 轉染試劑 (11668019, Invitrogen) 將CCL5質體和EGFP質體 (#6084-1, Addgene, MA, USA) 送入SHSY5Y細胞中表現。之後,將轉染的細胞移到 6 孔/24 孔盤中,並用上述劑量的過氧化氫和 CCL5處理,用於蛋白質收集和 MTT 測定。 Primary neurons were cultured from gestational day 16.5-17 (E16.5-17) C57BL/6 and CCL5 -/- mouse embryos. Embryonic brain tissue was lysed in Dulbecco's Modified Eagle's Buffered Medium (DMEM, 12800-017, Gibco) containing 2 mg/ml papain (Worthington, LS003119) and 0.05% Trypsin-EDTA (Gibco, 25200-072) for 14 min and cultured in neurobasal medium (21103-049, Gibco) containing 10% v/v heat-inactivated fetal bovine serum (FBS, 10437-028, Gibco), 1% v/v antibiotic-antimycotic (15240-062, Gibco) and 2 mM L-glutamine (25030, Gibco). After 2 hours of cell attachment, the culture medium was replaced with "complete medium" (neurobasal medium containing 1% v/v N-2 supplement (17502048, Gibco), 2% v/v B-27 supplement (17504044, Gibco), 1% v/v antibiotic-antimycotic and 2 mM L-glutamine). After that, half of the medium was replaced every 3 days. On the 4th day of culture, the cells were first given 10, 50, 100 and 250 pg/ml of CCL5/RANTES recombinant protein. After 30 minutes, the cells were treated with 25, 50, 100, 250, 500 μM hydrogen peroxide (PanReac AppliChem, 131077) for 24 hours. In the cell line study, CCL5 plasmid and EGFP plasmid (#6084-1, Addgene, MA, USA) were expressed in SHSY5Y cells using Lipofectamine™ 2000 transfection reagent (11668019, Invitrogen). Afterwards, the transfected cells were transferred to 6-well/24-well plates and treated with the above doses of hydrogen peroxide and CCL5 for protein collection and MTT assay.

細胞存活率分析部分,將細胞與 MTT 溶液(10 μg/ml,M6494. Invitrogen™)一起培養1小時。隨後,加入 200 μl DMSO 以裂解細胞並以 550 nm波長讀取訊號。For cell viability analysis, cells were incubated with MTT solution (10 μg/ml, M6494. Invitrogen™) for 1 hour. Subsequently, 200 μl of DMSO was added to lyse the cells and the signal was read at 550 nm.

十、DCFDA 標記和免疫細胞化學染色:10. DCFDA labeling and immunocytochemical staining:

將初級神經元以密度為 5 × 10 5種到6 孔盤中。第4天加入100 pg/ml CCL5重組蛋白,等待30分鐘後再分別加入0、250、500μM H 2O 2。24小時後,移除培養基並用PBS清洗,在每一個well加入2ml的PBS與20μl DCFDA solution (10mM),然後讓DCFDA solution與細胞於培養箱內作用1小時。1小時後,移除含有DCFDA溶液的PBS除並以4%PFA固定10分鐘。隨後用含有 1 μg/ml DAPI 的緩衝液作用 30 分鐘。將細胞與具有神經元細胞骨架標記物 Tuj-1 (1:2000, MAB5564, Millipore, USA) 的一抗在 4°C 下作用一個晚上。二抗使用Anti-mouse 568 (1:400, Invitrogen, USA)。實驗結果利用螢光顯微鏡觀察並使用 Image J 軟體進行分析。 Primary neurons were seeded into 6-well plates at a density of 5 × 10 5 . On the 4th day, 100 pg/ml CCL5 recombinant protein was added, and 0, 250, and 500 μM H 2 O 2 were added after 30 minutes. After 24 hours, the medium was removed and washed with PBS. 2 ml of PBS and 20 μl of DCFDA solution (10 mM) were added to each well, and then the DCFDA solution and cells were allowed to act in the incubator for 1 hour. After 1 hour, the PBS containing the DCFDA solution was removed and fixed with 4% PFA for 10 minutes. Then, the cells were treated with a buffer containing 1 μg/ml DAPI for 30 minutes. The cells were incubated with primary antibody against the neuronal cytoskeleton marker Tuj-1 (1:2000, MAB5564, Millipore, USA) at 4°C overnight. The secondary antibody used was Anti-mouse 568 (1:400, Invitrogen, USA). The results were observed under a fluorescent microscope and analyzed using Image J software.

十一、CCL5 ELISA測定:11. CCL5 ELISA assay:

實驗利用小鼠 CCL5/RANTES DuoSet ELISA 試劑組(DY478,R&D System)測量 WT 和 CCL5-KO 海馬迴中的 CCL5/RANTES 含量。利用PBS將capture antibody稀釋至2μg/ml,並加入100μl稀釋的capture antibody到96孔盤之中並在室溫靜置過夜。移除capture antibody,然後加入含有1% BSA 的PBS靜置2 小時以阻斷非專一性作用,然後用含有 0.05% Tween 20的 PBS (洗滌緩衝液)洗滌。加入100 μl 組織樣品或標準品到96孔微孔盤中,在室溫下作用2 小時,然後用洗滌緩衝液洗滌。接下來,將樣品與生物素結合的檢測抗體(detection antibody)作用2 小時,然後與Streptavidin-HRP作用30分鐘以產生訊號。微孔盤內的樣品再用分光光譜儀讀取 450 nm 波長數值。利用不同濃度CCL5標準品的吸光值繪製出標準曲線,得到標準曲線的方程式後把樣本的吸光值代入,計算海馬迴組織中CCL5的含量。The mouse CCL5/RANTES DuoSet ELISA kit (DY478, R&D System) was used to measure CCL5/RANTES levels in the hippocampus of WT and CCL5-KO. The capture antibody was diluted to 2 μg/ml in PBS, and 100 μl of the diluted capture antibody was added to a 96-well plate and incubated overnight at room temperature. The capture antibody was removed, and PBS containing 1% BSA was added for 2 hours to block non-specific interactions, followed by washing with PBS containing 0.05% Tween 20 (wash buffer). 100 μl of tissue samples or standards were added to a 96-well microtiter plate, incubated for 2 hours at room temperature, and then washed with wash buffer. Next, the sample was incubated with a biotin-conjugated detection antibody for 2 hours, and then with Streptavidin-HRP for 30 minutes to generate a signal. The sample in the microplate was then read at a wavelength of 450 nm using a spectrophotometer. A standard curve was drawn using the absorbance values of CCL5 standards of different concentrations. After obtaining the equation of the standard curve, the absorbance value of the sample was substituted into the equation to calculate the CCL5 content in the hippocampus tissue.

十二、量化和統計分析:12. Quantification and statistical analysis:

使用 GraphPad Prizm 8.0 (GraphPad Software, Dan Diego, CA, USA) 進行統計分析。非配對 t檢驗用於檢測兩組之間的差異,One-way ANOVA分析用於檢測同一組內的差異,信賴區間為 95%,Two-way ANOVA分析用於多因子檢驗。Bonferroni 校正用於任何系列測量。 p值<0.05視為有顯著差異。所有結果均以平均值±SEM表示。 Statistical analysis was performed using GraphPad Prizm 8.0 (GraphPad Software, Dan Diego, CA, USA). Unpaired t- test was used to detect differences between two groups, one-way ANOVA analysis was used to detect differences within the same group with a confidence interval of 95%, and two-way ANOVA analysis was used for multifactorial testing. Bonferroni correction was used for any serial measurements. A p value < 0.05 was considered significant. All results are presented as mean ± SEM.

實驗結果說明:Experimental results:

一、輕度腦損傷後 CCL5 缺乏小鼠的記憶恢復延遲1. Delayed memory recovery in CCL5-deficient mice after mild brain injury

WT和 CCL5-KO小鼠在 2 個月大時使用重物落下模型誘發小鼠輕度腦損傷。在重物落下誘發腦損傷後,兩種類型的小鼠的失去意識時間(翻正反射)較只有麻醉的對照組均為增加,但兩組動物之間沒有差異(第 1A圖的A)。改良版神經損傷程度評分(mNSS)評估受傷一天後神經損傷程度。WT 和 CCL5-KO 小鼠的 mNSS 評分數值約為 3,顯示為輕度損傷(第 1A圖的B)。WT and CCL5-KO mice were induced with mild brain injury using a heavy-fall model at 2 months of age. After heavy-fall-induced brain injury, the duration of unconsciousness (righting reflex) of both types of mice was increased compared with the anesthetized control group, but there was no difference between the two groups of animals (Figure 1A, A). The modified neurological severity score (mNSS) assessed the degree of neurological injury one day after injury. The mNSS score values of WT and CCL5-KO mice were approximately 3, indicating mild injury (Figure 1A, B).

利用新奇事物認知測試(Novel Object Recognition Test,NOR)和巴德斯迷宮(Barnes maze)分析小鼠的物件識別記憶和空間記憶(第1B圖的A)。創傷前,未受傷的 WT 和 CCL5-KO 組對新事物的偏好兩組間沒有差異。輕度腦損傷後兩種老鼠對新事物的偏好均有降低,WT組在受傷 7 天後開始恢復,但在 CCL5-KO 組在 受傷14 天後才開始恢復(第1B圖的B),兩組小鼠的運動能力均未受到腦傷所影響(第1B圖的C)。The object recognition and spatial memory of mice were analyzed using the Novel Object Recognition Test (NOR) and the Barnes maze (A in Figure 1B). Before the injury, there was no difference between the uninjured WT and CCL5-KO groups in their preference for novel objects. After mild brain injury, the preference for novel objects of both mice decreased. The WT group began to recover 7 days after the injury, but the CCL5-KO group began to recover only 14 days after the injury (B in Figure 1B). The motor ability of both groups of mice was not affected by brain injury (C in Figure 1B).

在創傷前,巴德斯迷宮中短期回溯記憶(SM,訓練後 2 天)和長期回溯記憶(LM,訓練後 7 天)的滯留期在WT和 CCL5-KO小鼠的對照組和腦損傷組(mTBI 組)皆相似, 兩個腦損傷組在受傷 2 天後回溯測試的滯留期都增加了(2 天回溯)(第1C圖的A和B)。創傷後(dpi)第4天(第1C圖的C)以及第 28 天(第1C圖的D)分別進行新的一輪巴德斯迷宮學習和記憶測試。WT小鼠迅速從腦傷引起的記憶受損中恢復;WT小鼠的滯留期在對照組和損傷組之間沒有差異。在WT小鼠中,對照組和腦損傷組之間的記憶回溯測試和步行路徑距離也沒有差異(第1D圖的A~D)。反之,創傷後 4 天的新一輪巴德斯迷宮中,CCL5-KO 腦損傷組滯留期比對照組更高(第1C圖的C)。在創傷後28天第二輪新的巴德斯迷宮 中 CCL5-KO 腦損傷組滯留期甚至更高(第1C圖的D)。尤以創傷後第二次訓練第四天的滯留期較第一次訓練更高。在 CCL5-KO TBI 組中,短期記憶回溯的滯留期和行走距離有所增加(第1D圖的E~H)。Before injury, the retention of short-term recall memory (SM, 2 days after training) and long-term recall memory (LM, 7 days after training) in the Bards maze was similar in both control and brain injury (mTBI) groups of WT and CCL5-KO mice. Both brain injury groups showed increased retention in the recall test 2 days after injury (2-day recall) (A and B in Figure 1C). A new round of Bards maze learning and memory testing was performed on day 4 (C in Figure 1C) and day 28 (D in Figure 1C) post-injury (dpi). WT mice recovered rapidly from memory impairment caused by brain injury; the retention period of WT mice did not differ between the control and injury groups. In WT mice, there was also no difference in memory retrieval test and walking distance between the control and injury groups (A to D in Figure 1D). In contrast, in a new round of Bards maze 4 days after trauma, the retention period of the CCL5-KO injury group was higher than that of the control group (C in Figure 1C). The retention period of the CCL5-KO injury group was even higher in a second round of a new Bards maze 28 days after trauma (D in Figure 1C). In particular, the retention period on the fourth day of the second training after trauma was higher than that of the first training. In the CCL5-KO TBI group, the retention time and walking distance of short-term memory retrieval were increased (Figure 1D, E-H).

二、輕度創傷後,神經元受損的數量在CCL5缺乏小鼠的海馬迴中增加2. After mild trauma, the number of damaged neurons increased in the hippocampus of CCL5-deficient mice

為了解創傷後CCL5-KO記憶障礙的可能原因,本發明分析了創傷後28天的海馬迴神經元損傷和總神經元數量。FJC標記受損的神經元,在WT和CCL5-KO小鼠腦損傷組海馬迴中均增加;與 WT 腦損傷組相比,KO 腦損傷組中FJC標記的受損神經元更多(第2A圖的A、C)。此外, NeuN標記的海馬迴神經元數量在CCL5-KO小鼠腦損傷組中明顯減少(第2A圖的B、D),但在 WT 腦損傷組的海馬迴神經元數目並沒有減少現象。此外,mTBI 後1個月,WT小鼠的發炎相關細胞-星形膠細胞(astrocyte) 與微小膠細胞(microglia) 均沒有增加,對照組和腦損傷組海馬迴組織中的 GFAP或Iba1陽性細胞數相仿(第2B圖);然而,在mTBI後,在CCL5-KO小鼠海馬迴組織中發現了更多的星形膠細胞,微小膠細胞則無明顯變化(第2B圖的A、C和B、D)。另發現 4-7 dpi 後WT小鼠海馬迴中的 TNF-α、IL-1β 和 IL-10 增加,但 14 天後減少。相反,在 14-28 dpi 時,CCL5-KO小鼠中的 TNF-α、IL-1β 增加,但是IL-10則無如WT顯著增加的現象(第2B圖的E-G)。這些發現表示創傷後CCL5-KO小鼠的免疫反應與WT小鼠有著不同的調節,特別是對於 IL-10 的活化,在CCL5-KO小鼠中,在輕度腦外傷以及星形膠細胞增生增加後,更多的神經元死亡和/或受損。這些發現與行為表現數據一致。總之,在缺乏 CCL5 的小鼠中,海馬迴神經元從輕度創傷中的恢復較少。To understand the possible causes of CCL5-KO memory impairment after trauma, the present invention analyzed the neuronal damage and total neuron number in the hippocampus 28 days after trauma. FJC-labeled damaged neurons increased in the hippocampus of both WT and CCL5-KO mouse brain injury groups; compared with the WT brain injury group, there were more FJC-labeled damaged neurons in the KO brain injury group (A, C in Figure 2A). In addition, the number of hippocampal neurons labeled by NeuN was significantly reduced in the CCL5-KO mouse brain injury group (B, D in Figure 2A), but there was no decrease in the number of hippocampal neurons in the WT brain injury group. In addition, one month after mTBI, the inflammation-related cells-astrocytes and microglia in WT mice did not increase, and the number of GFAP or Iba1-positive cells in the hippocampus tissue of the control group and the brain injury group was similar (Figure 2B); however, after mTBI, more astrocytes were found in the hippocampus tissue of CCL5-KO mice, while microglia did not change significantly (A, C and B, D in Figure 2B). It was also found that TNF-α, IL-1β and IL-10 increased in the hippocampus of WT mice after 4-7 dpi, but decreased after 14 days. In contrast, TNF-α and IL-1β were increased in CCL5-KO mice at 14-28 dpi, but IL-10 was not significantly increased compared to WT (Fig. 2B, E-G). These findings suggest that the immune response after trauma in CCL5-KO mice is regulated differently than in WT mice, particularly with respect to IL-10 activation, and that more neurons die and/or are damaged after mild brain trauma and increased astrocytosis in CCL5-KO mice. These findings are consistent with the behavioral data. In summary, hippocampal neurons recover less from mild trauma in mice lacking CCL5.

三、CCL5含量與創傷後早期神經元氧化壓力相關3. CCL5 content is associated with early neuronal oxidative stress after trauma

為了進一步了解CCL5與腦外傷後神經元恢復之間的關係,透過ELISA分析組織內CCL5的含量。海馬迴組織中CCL5的含量在 4 dpi後增加,並在WT小鼠 14 dpi後逐漸恢復(第3A圖的A);這與 WT 小鼠記憶恢復的時間線相關。周邊血液血清中的CCL5表現量則與腦內變化無明顯相關(第3A圖的B)。氧化壓力被認為是腦外傷後早期神經元損傷的主要原因。受到氧化壓力的細胞可被hypoxyproble標記(第3B圖的A, B圖中綠色所標示),同步與神經元標記-NeuN(第3B圖的紅色標示、第3C圖的A的紅色標示)或星形膠細胞標記-GFAP(紅色標示,第3C圖的B)以了解受到氧化傷害的細胞類型。Hypoxyprobe標記的細胞大多是NeuN陽性,但不是GFAP陽性的細胞。在WT和CCL5-KO小鼠中,對照組、1 dpi 和 7 dpi 小鼠的海馬迴神經元總數沒有顯著差異(第3B圖的A–C, NeuN +)。在WT小鼠中,Hypoxyprobe 標記的細胞在 1 dpi 時增加,並在 7 dpi時恢復到與對照組相同(第3B圖的A, D-hypoxyprobe +), Hypoxyprobe 標記與NeuN +共標記的細胞變化與C類似(第3B圖的A、E-hypoxyprobe +;NeuN +)。相比之下,在CCL5-KO小鼠中,在 1 dpi 至 7 dpi Hypoxyprobe標記細胞數均較控制組為多(第3B圖的B, D- hypoxyprobe +),尤其是在與NeuN +共標記的神經元中更為顯著增加(第3B圖的B、E- hypoxyprobe +;NeuN +)。這表明在CCL5-KO小鼠創傷後可以看到更多神經元處於氧化壓力,因為WT小鼠在1 dpi 時增加的氧化壓力在7 dpi後降低至正常,但在 KO小鼠中則沒有,因此,CCL5可能在創傷後神經元氧化壓力平衡有著關鍵作用。 To further understand the relationship between CCL5 and neuronal recovery after brain trauma, the content of CCL5 in tissues was analyzed by ELISA. The content of CCL5 in hippocampal tissue increased after 4 dpi and gradually recovered after 14 dpi in WT mice (Figure 3A, A); this is related to the timeline of memory recovery in WT mice. The expression of CCL5 in peripheral blood serum was not significantly correlated with changes in the brain (Figure 3A, B). Oxidative stress is considered to be the main cause of early neuronal damage after brain trauma. Cells under oxidative stress can be labeled with hypoxyproble (green in Figure 3B, A, B) and synchronized with the neuronal marker NeuN (red in Figure 3B, red in Figure 3C, A) or astrocyte marker GFAP (red in Figure 3C, B) to understand the cell types that are damaged by oxidative damage. Hypoxyprobe-labeled cells are mostly NeuN-positive, but not GFAP-positive cells. In WT and CCL5-KO mice, there was no significant difference in the total number of hippocampal neurons between control, 1 dpi, and 7 dpi mice (Figure 3B, A–C, NeuN + ). In WT mice, the number of Hypoxyprobe-labeled cells increased at 1 dpi and recovered to the same level as the control group at 7 dpi (A, D-hypoxyprobe + in Figure 3B), and the changes in cells co-labeled with Hypoxyprobe and NeuN + were similar to those in C (A, E-hypoxyprobe + ; NeuN + in Figure 3B). In contrast, in CCL5-KO mice, the number of Hypoxyprobe-labeled cells was higher than that in the control group from 1 dpi to 7 dpi (B, D- hypoxyprobe + in Figure 3B), especially in neurons co-labeled with NeuN + (B, E- hypoxyprobe + ; NeuN + in Figure 3B). This suggests that more neurons under oxidative stress can be seen in CCL5-KO mice after trauma, because the increased oxidative stress in WT mice at 1 dpi decreased to normal after 7 dpi, but not in KO mice. Therefore, CCL5 may play a key role in the balance of oxidative stress in neurons after trauma.

四、創傷後CCK5-KO小鼠海馬迴ROS 生成增加和抗氧化劑活化能力受損IV. Increased ROS production and impaired antioxidant activation in the hippocampus of CCK5-KO mice after trauma

創傷誘導NADPH氧化酶活化並增加細胞ROS;NADPH氧化酶產生的O 2 -可以被SOD轉化為H 2O 2,然後被GPX系統清除(第4A圖的A)。因此,本發明使用光澤精(lucigenin)測定研究小鼠海馬迴組織中NADPH氧化酶所產生的ROS。海馬迴組織中NADPH 氧化酶生成的ROS在對照組和受傷後4天和7天WT小鼠之間沒有顯著差異,但在CCL5-KO小鼠中,NADPH 氧化酶生成的ROS在受傷後第4天和第7天顯著增加(第4A圖的B)。 Trauma induces NADPH oxidase activation and increases cellular ROS; O 2 - generated by NADPH oxidase can be converted to H 2 O 2 by SOD and then cleared by the GPX system (A in FIG. 4A). Therefore, the present invention uses lucigenin to measure ROS generated by NADPH oxidase in mouse hippocampal tissue. There was no significant difference in ROS generated by NADPH oxidase in hippocampal tissue between the control group and WT mice 4 and 7 days after injury, but in CCL5-KO mice, ROS generated by NADPH oxidase increased significantly on days 4 and 7 after injury (B in FIG. 4A).

創傷後第4天和第7天,WT 組織中抗氧化劑GPX1的蛋白質含量明顯升高,而SOD1和SOD2的蛋白質含量則沒有變化(第4B圖的A~D)。相比之下,這些抗氧化劑蛋白在CCL5-KO小鼠海馬迴損傷後並沒有增加。(第4B圖的E~G); 本發明發現在 4 和 7 dpi 後CCL5-KO海馬迴中的SOD1蛋白減少(第4B圖的A、E)。進一步研究GPX1在WT 和CCL5-KO之間的功能和活性,與 WT 相比,CCL5-KO海馬迴組織中的GSH生成率較低(第4C圖的A); CCL5-KO海馬迴組織中還原形態的GSH含量也較低(第4C圖的B)及CCL5-KO小鼠海馬迴中GPX1的 mRNA表現量也較低(第4C圖的C)。WT小鼠 mTBI後海馬迴GPX1 mRNA表現隨著天數增加;相反,GPX1在缺乏CCL5的小鼠中沒有被活化的情況(第4C圖的D)。這表示在正常情況下,CCL5透過增加抗氧化-GPX1來降低組織ROS含量;相反,在缺乏CCL5的小鼠中,海馬迴組織ROS則會累積增加。At 4 and 7 days after injury, the protein level of the antioxidant GPX1 in WT tissues was significantly increased, while the protein levels of SOD1 and SOD2 did not change (Figure 4B, A-D). In contrast, these antioxidant proteins did not increase after hippocampal injury in CCL5-KO mice. (Figure 4B, E-G); The present invention found that SOD1 protein was reduced in the CCL5-KO hippocampus after 4 and 7 dpi (Figure 4B, A, E). Further investigation of the function and activity of GPX1 between WT and CCL5-KO revealed that the rate of GSH production in CCL5-KO hippocampus tissue was lower than that in WT (Fig. 4C, A); the content of reduced GSH in CCL5-KO hippocampus tissue was also lower (Fig. 4C, B); and the mRNA expression of GPX1 in the hippocampus of CCL5-KO mice was also lower (Fig. 4C, C). The expression of GPX1 mRNA in the hippocampus of WT mice increased with the number of days after mTBI; in contrast, GPX1 was not activated in mice lacking CCL5 (Fig. 4C, D). This indicates that under normal circumstances, CCL5 reduces tissue ROS levels by increasing the antioxidant GPX1; conversely, in mice lacking CCL5, ROS accumulation in hippocampal tissue increases.

五、細胞內和細胞外表現CCL5均可保護神經元免受ROS誘導的細胞死亡5. Both intracellular and extracellular CCL5 can protect neurons from ROS-induced cell death

本發明進一步測試CCL5在體外培養系統中的保護作用。WT初代神經元細胞有表現CCL5或將重組CCL5蛋白添加到CCL5-KO初代神經元培養液中時,可減少 H 2O 2產生ROS所導致的細胞死亡(第5A圖)。利用DCFDA 標記因 ROS所導致CCL5-KO 初代神經元細胞的凋亡,DCFDA 標記的死亡細胞數隨著處理H 2O 2的濃度增加,添加CCL5重組蛋白至培養液中則減少了DCFDA 標記的死亡細胞數。高劑量(500 μM H 2O 2)會殺死大部分神經元,添加CCL5重組蛋白並不能挽救細胞的死亡(第5B圖)。另外藉由轉殖CCL5基因到SHSY5Y細胞,以大量表達 CCL5(第5C圖的C)也可以增加細胞在H 2O 2處理後的存活率(第5C圖的A)。當用H 2O 2處理細胞時,GPX1蛋白含量增加;有趣的是,額外表達CCL5會增加GPX1蛋白的表現量,而H 2O 2處理後GPX1含量仍進一步增加(第5C圖的B)。直接給予細胞重組CCL5蛋白(10、100 pg/ml)到CCL5-KO初代神經元培養液中,也會增加GPX1蛋白的表現量(第5C圖的D)。 The present invention further tests the protective effect of CCL5 in an in vitro culture system. When WT primary neurons express CCL5 or recombinant CCL5 protein is added to the culture medium of CCL5-KO primary neurons, cell death caused by ROS generated by H 2 O 2 can be reduced (Figure 5A). DCFDA was used to mark the apoptosis of CCL5-KO primary neurons caused by ROS. The number of DCFDA-labeled dead cells increased with the concentration of H 2 O 2 treatment, and the addition of CCL5 recombinant protein to the culture medium reduced the number of DCFDA-labeled dead cells. High doses (500 μM H 2 O 2 ) will kill most neurons, and the addition of CCL5 recombinant protein cannot rescue cell death (Figure 5B). In addition, by transferring the CCL5 gene into SHSY5Y cells, expressing CCL5 in large quantities (Figure 5C, C) can also increase the survival rate of cells after H2O2 treatment (Figure 5C, A). When cells are treated with H2O2 , GPX1 protein content increases; interestingly, forehead expression of CCL5 increases GPX1 protein expression, and GPX1 content further increases after H2O2 treatment (Figure 5C , B). Direct administration of recombinant CCL5 protein (10, 100 pg/ml) to CCL5-KO primary neuron culture medium also increases GPX1 protein expression (Figure 5C, D).

六、N-乙酰半胱氨酸( N-acetyl cysteine, NAC),一種GPX前驅物,可減少CCL5-KO小鼠在輕度腦外傷後的組織內的氧化壓力並防止記憶喪失 6. N -acetyl cysteine (NAC), a GPX pro-promoter, reduces oxidative stress in tissues and prevents memory loss in CCL5-KO mice after mild brain injury

為了進一步明確CCL5保護神經元的機制,在重物落下誘發的腦外傷後,以腹腔注射法,給予CCL5-KO小鼠GPX1的前驅物NAC 5 mg/kg或20 mg/kg。對照組則是腦傷後給予小鼠注射生理食鹽水(第6A圖)。首先測試創傷前對記憶的影響。創傷前不同組的滯留期時間長度相似,但在 2天回溯記憶測試中,創傷後給予生理食鹽水的CCL5-KO小鼠(KO-TBI 組)滯留期時間增加。相比之下,在創傷後接受NAC治療的小鼠有較短的滯留期時間。此外,NAC 20 mg/kg 組的滯留期時間與對照組相似(第6B圖的A)。與生理食鹽水處理的TBI組相比,5 mg/kg 和 20 mg/kg NAC 處理後顯著改善了 4 dpi新學習和記憶訓練(第6B圖的B); 20 mg/kg NAC 也顯著改善了短期回朔記憶力(第6B圖的C)。海馬迴組織內ROS含量在5 mg/kg 和20 mg/kg NAC 處理組也因而降低(第6B圖的D)。創傷後NAC處理的小鼠海馬迴中受到氧化壓力的神經元(hypoxyprobe +和 NeuN +細胞)數量也減少了(第6C圖的A、B)。此外,NAC 處理後小鼠海馬迴中的GPX1 mRNA表現量升高(第6C圖的C)。 To further clarify the mechanism by which CCL5 protects neurons, CCL5-KO mice were given 5 mg/kg or 20 mg/kg of NAC, a pro-promoter of GPX1, by intraperitoneal injection after heavy falling TBI. Mice in the control group were injected with saline after TBI (Figure 6A). The effect of pre-injury on memory was first tested. The retention time of different groups before trauma was similar, but in the 2-day retrospective memory test, the retention time of CCL5-KO mice (KO-TBI group) given saline after trauma increased. In contrast, mice treated with NAC after trauma had a shorter retention time. In addition, the retention time of the NAC 20 mg/kg group was similar to that of the control group (Figure 6B, A). Compared with the saline-treated TBI group, 5 mg/kg and 20 mg/kg NAC treatment significantly improved new learning and memory training at 4 dpi (Figure 6B, B); 20 mg/kg NAC also significantly improved short-term recall memory (Figure 6B, C). The ROS content in the hippocampal tissue was also reduced in the 5 mg/kg and 20 mg/kg NAC-treated groups (Figure 6B, D). The number of neurons (hypoxyprobe + and NeuN + cells) under oxidative stress in the hippocampus of mice treated with NAC after trauma was also reduced (Figure 6C, A, B). In addition, the expression of GPX1 mRNA in the hippocampus of mice was increased after NAC treatment (Figure 6C, C).

七、CCL5增加GPX1 mRNA 轉錄,減少組織ROS,並防止CCL5-KO小鼠在輕度腦外傷後記憶喪失VII. CCL5 increases GPX1 mRNA transcription, reduces tissue ROS, and prevents memory loss in CCL5-KO mice after mild brain injury

使用CCL5重組蛋白進行了體內治療實驗。在重物落下誘發的腦外傷後,利用鼻腔內(in)途徑給予小鼠CCL5重組蛋白,重組蛋白可經由 rostral migratory stream進入小鼠腦室。CCL5重組蛋白先與 Alexa-594染料共軛標定,以追蹤重組蛋白從鼻腔內(in)給予後到腦內的分佈(第7A圖的B),給予小鼠PBS用來作對照組(第7A圖的A)。研究設計顯示如第7B圖,類似於NAC研究。創傷前不同組的滯留期時間相似。在 2 天回溯記憶測試中,PBS處理的腦損傷 KO小鼠(KO-TBI + PBS 組)滯留期時間因腦傷增加,接受 CCL5 重組蛋白的小鼠則表現出較短的滯留期,並與創傷前相似(第7C圖的A)。與 PBS處理的腦傷組相比,CCL5 重組蛋白治療組的新學習和記憶訓練表現明顯更好(第7C圖的B); 短期記憶回溯測試也得到了改善(第7C圖的C)。CCL5重組蛋白治療顯著降低海馬迴組織內 ROS的含量(第7C圖的D); 創傷後CCL5重組蛋白治療的小鼠海馬迴中受到氧化壓力的神經元(hypoxyprobe +和 NeuN +細胞)數量也減少了(第7D圖的A、B)。重要的是,給予CCL5也顯著增加了創傷後小鼠海馬迴中 GPX1 的 mRNA表現量(第7D圖的C)。 In vivo treatment experiments were performed using recombinant CCL5 protein. After heavy falling brain trauma, mice were given CCL5 recombinant protein via the intranasal (in) route, and the recombinant protein was able to enter the mouse ventricle via the rostral migratory stream. CCL5 recombinant protein was first co-labeled with Alexa-594 dye to track the distribution of the recombinant protein into the brain after intranasal (in) administration (B in Figure 7A), and mice were given PBS as a control group (A in Figure 7A). The study design is shown in Figure 7B, similar to the NAC study. The retention time before trauma was similar in different groups. In the 2-day retrieval memory test, the retention time of PBS-treated brain-injured KO mice (KO-TBI + PBS group) increased due to brain injury, while mice receiving CCL5 recombinant protein showed a shorter retention period and was similar to that before injury (Figure 7C, A). Compared with the PBS-treated brain-injured group, the CCL5 recombinant protein-treated group performed significantly better in new learning and memory training (Figure 7C, B); the short-term memory retrieval test was also improved (Figure 7C, C). Treatment with recombinant CCL5 significantly reduced the level of ROS in hippocampal tissue (D in Figure 7C); the number of neurons (hypoxyprobe + and NeuN + cells) exposed to oxidative stress in the hippocampus of mice treated with recombinant CCL5 after trauma was also reduced (A and B in Figure 7D). Importantly, administration of CCL5 also significantly increased the mRNA expression of GPX1 in the hippocampus of mice after trauma (C in Figure 7D).

總之,本發明顯示了CCL5對抗氧化劑-GPX1的活化至關重要,它可以減少NADPH 氧化酶所產生的H 2O 2,並在mTBI後保護海馬迴神經元。缺乏CCL5的神經元產生更多的羥基自由基(hydroxyl radicals),可能藉由芬頓反應(Fenton reaction)破壞神經元。 In conclusion, the present invention shows that CCL5 is essential for the activation of the antioxidant GPX1, which can reduce H 2 O 2 produced by NADPH oxidase and protect hippocampal neurons after mTBI. Neurons lacking CCL5 produce more hydroxyl radicals, which may damage neurons through the Fenton reaction.

本發明確定CCL5在抗氧化劑GPX-1活化和保護神經元免受輕度腦外傷後的氧化傷害的新功能。CCL5的這種功能可進一步減少星形膠細胞增生、NADPH 氧化酶產生的 ROS、神經元病變死亡和輕度腦外傷後的顯著記憶障礙。The present invention identifies a novel function of CCL5 in activating the antioxidant GPX-1 and protecting neurons from oxidative damage after mild brain trauma. This function of CCL5 can further reduce astrocyte proliferation, ROS generated by NADPH oxidase, neuronal pathological death and significant memory impairment after mild brain trauma.

GPX酶家族是將H 2O 2轉化為無害的H 2O和O 2的關鍵。WT 海馬迴組織損傷後 4-7 天 ,GPX1蛋白和CCL5的含量均增加(第4B圖的D)。進一步發現SHSY5Y細胞中大量表達CCL5和給予額外的CCL5重組蛋白到初代神經元細胞,以及在將CCL5重組蛋白遞送到 CCL5-KO小鼠腦內後,均可增加GPX1蛋白的表現。在創傷後連續給予小鼠GPX前驅物NAC 3天,可減少神經元氧化壓力和損傷。在創傷後1小時內一次性給予小鼠CCL5重組蛋白到小鼠大腦中,也可減少海馬迴組織內的氧化壓力和神經元死亡。給予小鼠NAC 和CCL5都改善了海馬迴記憶認知功能。因此,本發明的數據強烈支持CCL5和GPX1在腦損傷後的抗氧化壓力和神經保護的作用,尤其關鍵時間為創傷後的1小時至3天。 The GPX enzyme family is key to converting H2O2 into harmless H2O and O2 . 4-7 days after WT hippocampal tissue injury, the levels of GPX1 protein and CCL5 increased (D in Figure 4B). It was further found that the high expression of CCL5 in SHSY5Y cells and the administration of additional CCL5 recombinant protein to primary neurons, as well as the delivery of CCL5 recombinant protein to the brain of CCL5-KO mice, increased the expression of GPX1 protein. Continuous administration of the GPX prodrug NAC to mice for 3 days after trauma reduced neuronal oxidative stress and damage. Giving mice CCL5 recombinant protein to the mouse brain once within 1 hour after trauma can also reduce oxidative stress and neuronal death in hippocampal tissue. Giving mice NAC and CCL5 both improved hippocampal memory and cognitive function. Therefore, the data of the present invention strongly support the role of CCL5 and GPX1 in anti-oxidative stress and neuroprotection after brain injury, especially the critical time is 1 hour to 3 days after trauma.

本發明發現在WT小鼠中腦損傷後的免疫趨化因子活化,特別是IL-10增加,但在腦損傷後的 CCL5-KO 小鼠中則沒有。這種免疫發炎反應在 7 天後減少,同時動物的行為表現也有所改善。儘管前 7 天 CCL5-KO 小鼠海馬迴的免疫發炎反應趨化物表現量較低,但卻有更多的氧化壓力傷害;尤其腦傷 7-14 天後,CCL5-KO小鼠的發炎增加。這些數據表明氧化壓力(oxidative stress, OS)和發炎反應之間平衡的重要性,尤其在腦傷後的1小時到前 3 天,這決定了之後神經元修復的方向。總之, CCL5對於腦損傷後的氧化壓力與免疫反應調節扮演重要的作用。The present invention found that immune trend factors were activated after brain injury in WT mice, especially IL-10 was increased, but not in CCL5-KO mice after brain injury. This immune inflammatory response decreased after 7 days, and the behavioral performance of the animals also improved. Although the expression of immune inflammatory response trends in the hippocampus of CCL5-KO mice was lower in the first 7 days, there was more oxidative stress damage; especially 7-14 days after brain injury, the inflammation of CCL5-KO mice increased. These data indicate the importance of the balance between oxidative stress (OS) and inflammatory response, especially from 1 hour to the first 3 days after brain injury, which determines the direction of subsequent neuronal repair. In conclusion, CCL5 plays an important role in regulating oxidative stress and immune response after brain injury.

因此,CCL5的調節和活化可能與腦損傷疾病進展和狀態相關。本發明研究發現CCL5-KO小鼠在輕度腦損傷1個月後,海馬迴組織有更多的星形膠細胞增生、氧化壓力產生和神經元損失。CCL5-KO小鼠培養的神經元對H 2O 2誘導的氧化壓力更加敏感和容易死亡,但當H 2O 2濃度高於500 μM時,CCL5的保護作用降低。 Therefore, the regulation and activation of CCL5 may be related to the progression and status of brain injury. This study found that CCL5-KO mice had more astrocyte proliferation, oxidative stress and neuronal loss in the hippocampus one month after mild brain injury. Neurons cultured in CCL5-KO mice were more sensitive to H2O2 - induced oxidative stress and prone to death, but when the H2O2 concentration was higher than 500 μM, the protective effect of CCL5 was reduced.

大多數腦損傷是輕微和重複性的,例如交通事故、美式足球、曲棍球和戰爭期間的爆炸。在本發明的輕度TBI模型中,CCL5 的保護劑量低於 500 pg/ml,海馬迴組織和血液中的 CCL5 含量幾乎沒有增加;嚴重的TBI可能會誘導更高含量的 CCL5,超過 1000 pg/ml 的釋放,這可能會活化不同的細胞訊號傳遞與功能。這也表明當損傷更嚴重時,CCL5在腦損傷中的作用和機制非常不同。這種二分法可能涉及不同的劑量相關的神經免疫效應。Most brain injuries are mild and repetitive, such as traffic accidents, American football, hockey, and bombings during war. In the mild TBI model of the present invention, the protective dose of CCL5 was less than 500 pg/ml, and the CCL5 content in hippocampal tissue and blood was almost not increased; severe TBI may induce the release of higher levels of CCL5, exceeding 1000 pg/ml, which may activate different cell signaling and functions. This also suggests that when the injury is more severe, the role and mechanism of CCL5 in brain injury are very different. This dichotomy may involve different dose-related neuroimmune effects.

了解如何治療輕度腦損傷至關重要且緊迫。根據本發明的研究,不同嚴重程度的損傷可能具有不同的神經元損傷機制,需要在臨床上進行不同的治療。然而,本發明發現創傷後的前 1 小時至3天是CCL5和NAC的關鍵-抗氧化激活和輕度腦損傷的治療。Understanding how to treat mild brain injury is crucial and urgent. According to the present invention, injuries of different severity may have different mechanisms of neuronal damage and require different treatments in the clinic. However, the present invention found that the first 1 hour to 3 days after trauma is the key for CCL5 and NAC-antioxidant activation and treatment of mild brain injury.

總之,CCL5在抗氧化GPX-1活化中具有獨特的功能,可減少輕度腦損傷後的細胞ROS和發炎反應誘導的星形細胞增生的現象,該功能可減少輕度腦外傷後進一步的神經元退化和記憶障礙。In conclusion, CCL5 has a unique function in antioxidant GPX-1 activation, which can reduce cellular ROS and inflammation-induced astrocytosis after mild brain injury, and this function can reduce further neuronal degeneration and memory impairment after mild brain injury.

以上所述僅為本發明之較佳可行實施例,非因此即侷限本發明之專利範圍,舉凡運用本發明說明書及圖式內容所為之等效結構變化,均理同包含於本發明之範圍內。The above description is only the preferred feasible embodiment of the present invention, and does not limit the patent scope of the present invention. All equivalent structural changes made by using the contents of the description and drawings of the present invention are also included in the scope of the present invention.

無。without.

第1A圖為本發明之WT和CCL5-KO小鼠在mTBI後的失去意識時間和mNSS評分。(A) WT和CCL5-KO對照組和重物落下導致的mTBI組的失去意識時間。(B)改良版神經缺損評分(mNSS)在受傷後1天進行。(*p<0.05;**p<0.01;***p<0.001。ns:無顯著差異。數據通過 t 檢驗分析並表示為平均值±S.E.M)。 第1B圖為本發明之WT和CCL5-KO小鼠在mTBI後的記憶障礙和恢復狀況。(A) 誘導輕度TBI和記憶認知實驗設計時間表—新奇事物認知測試(NOR)和巴德斯迷宮(BM)。 (B)WT和CCL5-KO小鼠在重物落下誘導輕度TBI後的對照組和mTBI組對新事物的偏好指數。在創傷後第4、7、14 和 28 天進行測試。(a:p < 0.05,aa:p < 0.01,aaa:p < 0.001,以t檢驗與對照組相比;b:p < 0.05,WT 28天與7天相比;*:p < 0.05,WT與CCL5-KO在同一時間點進行t檢驗)。(C) WT和 KO 小鼠的行走距離長度。(ns:沒有顯著差異),數據顯示為平均值±S.E.M(每組n = 9)。 巴德斯迷宮利用小鼠懼光的特性進行空間記憶行為測試,小鼠從迷宮中央釋放到找到逃脫箱所需時間定義為-滯留期。 第1C圖為本發明之WT和CCL5-KO小鼠在mTBI後的記憶障礙和恢復狀況。(A、B)小鼠在創傷前做一次巴德斯迷宮訓練測試,並在訓練後2天做短期記憶回朔測試(短期, SM)、與7 天後的長期記憶回朔測試(長期, LM)。可見各組小鼠之間並沒有差異。之後利用重物落誘發mTBI後,並於兩天後進行回朔記憶測試。可見mTBI組的滯留時間延長。(C、D)在創傷後4天和28天進行了新的學習記憶測試。WT小鼠mTBI組別在創傷後4天與28天的學習記憶測試與對照組沒有明顯差異。而CCL5-KO小鼠的mTBI組在兩次新的學習記憶測試的滯留期均明顯上升。數據通過雙向分析ANOVA並以平均值±S.E.M. 呈現 (D圖*, p = 0.0432,為與C圖中的CCL5-KO-TBI 4 dpi BM相比,通過雙向 ANOVA;D圖CCL5-KO-TBI 4 dpi與C圖中CCL5-KO-TBI 4 dpi相比,p = 0.0353通過t檢驗相比)(在每組中n = 5)。 第1D圖為巴德斯迷宮中WT和CCL5-KO小鼠在mTBI後空間記憶表現。(A、B、E、F)為創傷後11天在WT和CCL5-KO對照組和mTBI組的巴德斯迷宮中的代表性步行軌跡。 WT(C, D)和CCL5-KO(G, H)的對照組和mTBI組在11和16 dpi的滯留期和步行距離。(數據通過t檢驗分析並呈現平均值±SEM)。 第2A圖為創傷後28天海馬迴神經元損傷和神經元數量。 (A) WT和CCL5-KO海馬迴中FJC標記(綠色)的代表性圖像。(C)WT和CCL5 KO小鼠對照組和mTBI組中FJC染色陽性細胞的定量。在兩個mTBI組中均發現增加的FJC染色陽性神經元,CCL5-KO小鼠mTBI組比WT小鼠mTBI組更多受損神經元。(每組 n = 3~5)。(B)WT和CCL5-KO小鼠海馬迴中NeuN標記(紅色)神經元的代表性圖像。(D) 不同組別NeuN染色陽性細胞的定量。CCL5-KO mTBI組中的NeuN染色陽性神經元數顯著降低。(數據通過t檢驗分析並表示為平均值±SEM)(n = 3-5)。比例尺 = 100μm。 第2B圖為腦損傷後小鼠海馬迴的免疫細胞和免疫趨化物的活化。(A)在對照和mTBI的WT和CCL5-KO小鼠中,星形膠細胞由GFAP染色(綠色)標記。(C)量化每組3~4隻小鼠的GFAP標記細胞。(B)使用特異性標記物Iba1(紅色)對微小膠細胞進行免疫染色。(D)對每組2~3隻小鼠的Iba1染色陽性細胞進行定量。DAPI在A、B中以藍色標記細胞核。比例尺=100μm。腦損傷後WT和CCL5-KO小鼠海馬迴中發炎趨化因子TNF-α (E)、IL-1β (F)和IL-10的mRNA表現量。(*,p<0.05;**,p<0.01;***,p<0.001。通過t檢驗分析數據並表示為平均值±S.E.M)。 第3A圖為WT小鼠海馬迴組織和創傷後血清中CCL5的含量。透過ELISA分析小鼠mTBI後4、7、14和28天,與對照組小鼠海馬迴組織(A)和血清(B) 中CCL5的濃度。(數據通過對照組和創傷後組之間的t檢驗進行分析)。 第3B圖為創傷後1天和7天,在WT和CCL5-KO小鼠中,Hypoxyprobe標記的缺氧細胞與NeuN染色陽性神經元共同標定。 在1 dpi、7 dpi及對照組的WT和CCL5-KO小鼠中,缺氧細胞用Hypoxyprobe(綠色)標記,神經元用NeuN(紅色)標記。(A-B)WT和CCL5-KO小鼠海馬迴區域hypoxylprobe和NeuN標記的代表性圖像。(C-E)在創傷後第1天、第7天和對照組,在WT和CCL5-KO小鼠中對NeuN陽性細胞(NeuN+, C)、hypoxyprobe陽性細胞(hypoxyprobe+,D)以及Hypoxyprobe和NeuN均陽性的細胞(hypoxyprobe+NeuN+,E)進行定量。(數據通過t檢驗分析並表示為平均值±SEM)(每組n = 3)。比例尺= 100μm。 第3C圖為用NeuN或GFAP在小鼠海馬迴中與Hypoxyprobe進行雙重染色標記。(A-B)小鼠海馬迴中的缺氧細胞以Hypoxyprobe(綠色)標記,並與(A)中的NeuN(紅色)或(B)中的GFAP(紅色)共同染色標記。DAPI(藍色)標記細胞核。比例尺=100μm。 第4A圖為(A)腦外傷誘導細胞ROS生成與後續清除途徑(scavenger pathway)中的相關抗氧化劑。(B)在WT和 CCL5-KO小鼠中,與對照組相比,在4 dpi和7 dpi測量小鼠海馬迴組織中的NADPH氧化酶所產生的ROS。(數據通過t檢驗分析並表示為平均值±SEM);(每組n = 6)。 第4B圖為分析4 dpi和7 dpi和對照組小鼠海馬迴中抗氧化劑SOD1、SOD2和GPX1的蛋白質含量。(A)WT小鼠和CCL5-KO小鼠中SOD1、SOD2和GPX1蛋白轉印的代表性圖像。WT小鼠和CCL5-KO小鼠中SOD1(B、E),SOD2(C、F)和GPX1(D、G)蛋白表現的定量結果。(數據通過One-way ANOVA分析進行分析,並表示為平均值±S.E.M.)(每組 n = 5)。 第4C圖中的(A、B)為與WT小鼠相比,CCL5-KO小鼠的GSH活性和還原形式的GSH含量較低。(A)數據通過Two-way ANOVA分析進行分析,(B) 通過 t 檢驗分析,數據表示為平均值±S.E.M.。 (C)WT和CCL5-KO小鼠GPX1基因表現的定量PCR。(D)分析4 dpi、7 dpi及對照組中WT和CCL5-KO小鼠的GPX1基因表現的定量PCR。(數據通過 t 檢驗分析並表示為平均值±S.E.M.)。 第5A圖為CCL5對H 2O 2處理後細胞存活率的影響。(A、B)用CCL5(10 pg/ml、50 pg/ml、100 pg/ml和250 pg/ml)處理WT和CCL5-KO培養的初代皮層神經元細胞30分鐘,然後用H 2O 2處理(0、25、250和500μM)。細胞存活率為在24小時後通過MTT測定法檢測(數據通過雙向ANOVA分析並表示為平均值±S.E.M.)(n = 5)。 第5B圖為DCFDA染色(綠色)和神經元標記物Tuj-1(紅色)在CCL5-KO初代皮層神經元以CCL5和H 2O 2處理24小時後的代表性圖像。下方柱狀圖為在CCL5和H 2O 2處理後DCFDA標記的神經元細胞定量。(數據通過 t檢驗分析並表示為平均值±S.E.M.)(每組 n = 3)。比例尺= 100 μm。 第5C圖中的(A、B)為用EGFP或用CCL5質體轉染後,以0、25、100、250 μM H 2O 2處理24小時的SHSY5Y細胞。(A)通過MTT法測量EGFP和CCL5表達細胞的存活率。(數據通過t檢驗分析並表示為平均值±SEM,N = 3-4);(B) H 2O 2處理後,表達EGFP和CCL5的 SHSY5Y細胞中GPX1和Actin蛋白質轉印結果,GPX1蛋白的定量顯示在蛋白質轉印圖下方; (C)用EGFP或CCL5質體轉染後的CCL5基因表達含量。(D) CCL5重組蛋白處理後初代培養神經元中GPX1的蛋白含量。(數據表示為平均值±S.E.M.,N = 3-4)。 第6A圖為在誘導輕度TBI和巴德斯迷宮試驗後CCL5-KO小鼠腹腔給予NAC處理的實驗設計。 第6B圖為NAC處理改善mTBI後CCL5-KO小鼠的記憶障礙。(A)創傷前KO小鼠長期記憶回朔(LM)的滯留期,以及在重物落下創傷後2天的對照、mTBI和mTBI加不同劑量NAC處理(5毫克、20毫克)後的回朔記憶測試。(B)受傷後 4天4組小鼠的新巴德斯迷宮學習和記憶訓練進展。(C)4組之間短期回朔記憶測試(SM)的滯留期。(D) 4組小鼠海馬迴組織中NADPH oxidase所生成的ROS含量。 第6C圖為(A)CCL5-KO小鼠對照組、mTBI和mTBI加 NAC處理後hypoxyprobe標記海馬迴受到氧化壓力的神經細胞。(B)hypoxyprobe和NeuN 均陽性的細胞(hypoxyprobe+NeuN+)在不同組別中的定量。(C) 4組小鼠mTBI後12天GPX1 mRNA表現情況。 第7A圖為Alexa-594與CCL5抗體染色在小鼠海馬中的分佈狀況。(A)將接受PBS的小鼠視為對照組。(B)重組CCL5蛋白先以Alexa-594 標記並以鼻內遞送到CCL5-KO小鼠大腦中。 CCL5 特異性抗體標記CCL5(綠色)和CCL5共軛蛋白-Alexa-594(紅色)證實了通過鼻內遞送CCL5在小鼠海馬迴中的表現。細胞核由 DAPI(藍色)標記。比例尺=100μm。 第7B圖為輕度TBI後鼻腔給予CCL5-KO小鼠CCL5合成蛋白與後續巴德斯迷宮試驗的實驗設計。 第7C圖為(A)CCL5-KO小鼠在創傷前和創傷後2天的滯留期,分別在PBS處理和CCL5處理(300μg/kg)的小鼠中進行記憶回朔測試。(B) 2組小鼠於創傷後4天的新巴德斯迷宮學習和記憶訓練進展。(C)2組小鼠的短期回朔記憶測試(SM)的滯留期。(D)不同組別小鼠海馬迴組織中NADPH oxidase所生成的ROS含量。 第7D圖為(A)CCL5-KO小鼠對照、mTBI後以PBS或CCL5 處理後海馬迴的神經細胞內氧化壓力。(B)不同組中hypoxyprobe和NeuN陽性細胞(hypoxyprobe +NeuN +)的定量。(C)不同組別小鼠mTBI後4天,海馬迴中GPX1的mRNA表現量。 Figure 1A shows the duration of unconsciousness and mNSS score of WT and CCL5-KO mice after mTBI of the present invention. (A) The duration of unconsciousness of WT and CCL5-KO control groups and mTBI groups caused by heavy objects falling. (B) The modified neurological deficit score (mNSS) was performed 1 day after injury. (*p<0.05;**p<0.01;***p<0.001. ns: no significant difference. Data were analyzed by t test and expressed as mean ± SEM). Figure 1B shows the memory impairment and recovery of WT and CCL5-KO mice after mTBI of the present invention. (A) Timetable for inducing mild TBI and memory cognition experimental design - novel object recognition test (NOR) and Bards maze (BM). (B) Preference index for novel objects in control and mTBI groups of WT and CCL5-KO mice after mild TBI induced by heavy drop. Tests were performed on days 4, 7, 14, and 28 after injury. (a: p < 0.05, aa: p < 0.01, aaa: p < 0.001, compared with control group by t test; b: p < 0.05, WT 28 days compared with 7 days; *: p < 0.05, WT and CCL5-KO were tested by t test at the same time point). (C) Walking distance of WT and KO mice. (ns: not significantly different), data are shown as mean ± SEM (n = 9 per group). The Bards maze uses the light-phobia of mice to test spatial memory behavior. The time required for mice to be released from the center of the maze and find the escape box is defined as the retention period. Figure 1C shows the memory impairment and recovery of WT and CCL5-KO mice of the present invention after mTBI. (A, B) Mice were trained in the Bards maze once before the injury, and a short-term memory recall test (short-term, SM) was performed 2 days after the training, and a long-term memory recall test (long-term, LM) was performed 7 days later. It can be seen that there is no difference between the groups of mice. After that, mTBI was induced by the fall of a heavy object, and a recall memory test was performed two days later. It can be seen that the retention time of the mTBI group was prolonged. (C, D) New learning and memory tests were performed 4 and 28 days after injury. There was no significant difference between the WT mouse mTBI group and the control group in the learning and memory tests 4 and 28 days after injury. However, the retention period of the mTBI group of CCL5-KO mice in both new learning and memory tests was significantly increased. Data were analyzed by two-way ANOVA and presented as mean ± SEM (D panel *, p = 0.0432, compared with CCL5-KO-TBI 4 dpi BM in C panel, by two-way ANOVA; D panel CCL5-KO-TBI 4 dpi compared with CCL5-KO-TBI 4 dpi in C panel, p = 0.0353 by t test) (n = 5 in each group). Figure 1D shows spatial memory performance in Bards maze of WT and CCL5-KO mice after mTBI. (A, B, E, F) Representative walking tracks in Bards maze of WT and CCL5-KO control and mTBI groups 11 days after injury. Retention period and walking distance of WT (C, D) and CCL5-KO (G, H) control and mTBI groups at 11 and 16 dpi. (Data were analyzed by t-test and presented as mean ± SEM). Figure 2A shows neuronal damage and neuron number in hippocampus 28 days after injury. (A) Representative images of FJC labeling (green) in WT and CCL5-KO hippocampus. (C) Quantification of FJC-positive cells in control and mTBI groups of WT and CCL5 KO mice. Increased FJC-positive neurons were found in both mTBI groups, and more neurons were damaged in the CCL5-KO mouse mTBI group than in the WT mouse mTBI group. (n = 3-5 per group). (B) Representative images of NeuN-labeled (red) neurons in the hippocampus of WT and CCL5-KO mice. (D) Quantification of NeuN-positive cells in different groups. The number of NeuN-positive neurons was significantly reduced in the CCL5-KO mTBI group. (Data were analyzed by t-test and expressed as mean ± SEM) (n = 3-5). Scale bar = 100 μm. Figure 2B shows the activation of immune cells and immunoglobulins in the hippocampus of mice after brain injury. (A) Astrocytes were labeled by GFAP staining (green) in control and mTBI WT and CCL5-KO mice. (C) Quantification of GFAP-labeled cells in 3-4 mice per group. (B) Immunostaining of microglia using the specific marker Iba1 (red). (D) Quantification of Iba1-positive cells in 2-3 mice per group. DAPI marked the cell nuclei in blue in A and B. Scale bar = 100 μm. mRNA expression of inflammatory factors TNF-α (E), IL-1β (F), and IL-10 in the hippocampus of WT and CCL5-KO mice after brain injury. (*, p<0.05; **, p<0.01; ***, p<0.001. Data were analyzed by t-test and expressed as mean ± SEM). Figure 3A shows the content of CCL5 in the hippocampal tissue and serum after trauma in WT mice. The concentration of CCL5 in the hippocampal tissue (A) and serum (B) of mice in the control group at 4, 7, 14 and 28 days after mTBI in mice was analyzed by ELISA. (Data were analyzed by t-test between the control group and the post-traumatic group). Figure 3B shows that hypoxic cells labeled with Hypoxyprobe are co-labeled with NeuN-stained positive neurons in WT and CCL5-KO mice at 1 day and 7 days after trauma. Hypoxic cells were labeled with Hypoxyprobe (green) and neurons were labeled with NeuN (red) in WT and CCL5-KO mice at 1 dpi, 7 dpi, and control groups. (AB) Representative images of hypoxylprobe and NeuN labeling in the hippocampal region of WT and CCL5-KO mice. (CE) Quantification of NeuN-positive cells (NeuN+, C), hypoxyprobe-positive cells (hypoxyprobe+, D), and cells positive for both Hypoxyprobe and NeuN (hypoxyprobe+NeuN+, E) in WT and CCL5-KO mice at 1 dpi, 7 dpi, and control groups. (Data were analyzed by t-test and expressed as mean ± SEM) (n = 3 per group). Scale bar = 100 μm. Figure 3C shows double staining with NeuN or GFAP in mouse hippocampus with Hypoxyprobe. (AB) Hypoxic cells in mouse hippocampus were labeled with Hypoxyprobe (green) and co-stained with NeuN (red) in (A) or GFAP (red) in (B). DAPI (blue) marks the nucleus. Scale bar = 100 μm. Figure 4A shows (A) Brain trauma induces cellular ROS generation and related antioxidants in the scavenger pathway. (B) ROS generated by NADPH oxidase in mouse hippocampal tissues were measured at 4 dpi and 7 dpi in WT and CCL5-KO mice compared with control groups. (Data were analyzed by t-test and expressed as mean ± SEM); (n = 6 per group). Figure 4B shows the analysis of the protein content of antioxidants SOD1, SOD2, and GPX1 in the hippocampus of mice at 4 dpi and 7 dpi and control groups. (A) Representative images of protein transfer of SOD1, SOD2, and GPX1 in WT mice and CCL5-KO mice. Quantitative results of SOD1 (B, E), SOD2 (C, F), and GPX1 (D, G) protein expression in WT mice and CCL5-KO mice. (Data were analyzed by One-way ANOVA analysis and expressed as mean ± SEM) (n = 5 per group). Figure 4C (A, B) shows that GSH activity and reduced form of GSH content were lower in CCL5-KO mice compared with WT mice. (A) Data were analyzed by Two-way ANOVA analysis, (B) analyzed by t test, and data are expressed as mean ± SEM. (C) Quantitative PCR of GPX1 gene expression in WT and CCL5-KO mice. (D) Quantitative PCR of GPX1 gene expression in WT and CCL5-KO mice at 4 dpi, 7 dpi and control groups. (Data were analyzed by t test and expressed as mean ± SEM). Figure 5A shows the effect of CCL5 on cell survival after H 2 O 2 treatment. (A, B) WT and CCL5-KO cultured primary cortical neurons were treated with CCL5 (10 pg/ml, 50 pg/ml, 100 pg/ml and 250 pg/ml) for 30 minutes and then treated with H 2 O 2 (0, 25, 250 and 500 μM). Cell viability was detected by MTT assay after 24 hours (data were analyzed by two-way ANOVA and expressed as mean ± SEM) (n = 5). Figure 5B shows representative images of DCFDA staining (green) and neuronal marker Tuj-1 (red) in CCL5-KO primary cortical neurons treated with CCL5 and H 2 O 2 for 24 hours. The bar graph below is the quantification of DCFDA-labeled neurons after CCL5 and H 2 O 2 treatment. (Data were analyzed by t-test and expressed as mean ± SEM) (n = 3 per group). Scale bar = 100 μm. (A, B) in Figure 5C are SHSY5Y cells treated with 0, 25, 100, 250 μM H 2 O 2 for 24 hours after transfection with EGFP or CCL5 plasmid. (A) The survival rate of EGFP and CCL5 expressing cells was measured by MTT method. (Data were analyzed by t test and expressed as mean ± SEM, N = 3-4); (B) GPX1 and Actin protein transfer results in SHSY5Y cells expressing EGFP and CCL5 after H 2 O 2 treatment, and the quantification of GPX1 protein is shown below the protein transfer graph; (C) CCL5 gene expression level after transfection with EGFP or CCL5 plasmid. (D) GPX1 protein content in primary cultured neurons after CCL5 recombinant protein treatment. (Data are expressed as mean ± SEM, N = 3-4). Figure 6A shows the experimental design of intraperitoneal administration of NAC treatment to CCL5-KO mice after inducing mild TBI and Bards maze test. Figure 6B shows that NAC treatment improves memory impairment in CCL5-KO mice after mTBI. (A) Retention period of long-term memory recall (LM) in KO mice before injury, and recall memory test after control, mTBI, and mTBI plus different doses of NAC treatment (5 mg, 20 mg) 2 days after heavy object drop injury. (B) Progress of NeoBards maze learning and memory training in the four groups of mice 4 days after injury. (C) Retention period of short-term recall memory test (SM) among the four groups. (D) ROS content generated by NADPH oxidase in hippocampal tissue of the four groups of mice. Figure 6C shows (A) hypoxyprobe-labeled neurons in the hippocampus exposed to oxidative stress in CCL5-KO mice after control, mTBI, and mTBI plus NAC treatment. (B) Quantification of hypoxyprobe- and NeuN-positive cells (hypoxyprobe+NeuN+) in different groups. (C) Expression of GPX1 mRNA in the four groups of mice 12 days after mTBI. Figure 7A shows the distribution of Alexa-594 and CCL5 antibody staining in the mouse hippocampus. (A) Mice receiving PBS were considered as the control group. (B) Recombinant CCL5 protein was first labeled with Alexa-594 and delivered intranasally to the brain of CCL5-KO mice. CCL5-specific antibodies labeled CCL5 (green) and CCL5 conjugate-Alexa-594 (red) confirmed the expression of CCL5 in the mouse hippocampus by intranasal delivery. Cell nuclei were labeled by DAPI (blue). Scale bar = 100 μm. Fig. 7B shows the experimental design of intranasal administration of CCL5 synthetic protein to CCL5-KO mice after mild TBI and subsequent Bards maze test. Fig. 7C shows (A) Memory recall test in PBS-treated and CCL5-treated (300 μg/kg) mice before and 2 days after trauma. (B) Progress of learning and memory training in the new Bards maze of the two groups of mice 4 days after trauma. (C) Retention period of short-term recall memory test (SM) in two groups of mice. (D) ROS content generated by NADPH oxidase in hippocampal tissue of mice in different groups. Figure 7D shows (A) Oxidative stress in neurons of hippocampus of CCL5-KO mice after mTBI and PBS or CCL5 treatment. (B) Quantification of hypoxyprobe and NeuN positive cells (hypoxyprobe + NeuN + ) in different groups. (C) GPX1 mRNA expression in hippocampus of mice in different groups 4 days after mTBI.

無。without.

Claims (7)

一種組成物用於製備治療創傷性腦損傷之藥物的用途,該組成物包括CCL5,用以調節大腦海馬迴的過氧化物(Reactive oxygen species,ROS)含量。 A composition for preparing a drug for treating traumatic brain injury, the composition comprising CCL5, for regulating the level of reactive oxygen species (ROS) in the hippocampus of the brain. 如請求項1所述之用途,其中該創傷性腦損傷為輕微創傷性腦損傷(mild TBI)。 The use as described in claim 1, wherein the traumatic brain injury is mild traumatic brain injury (mild TBI). 如請求項2所述之用途,其中該組成物係用於活化穀胱甘肽過氧化物酶1(Glutathione peroxidase 1,GPX1)。 The use as described in claim 2, wherein the composition is used to activate glutathione peroxidase 1 (GPX1). 如請求項2所述之用途,其中該組成物係用於調節海馬迴的慢性發炎。 The use as described in claim 2, wherein the composition is used to regulate chronic inflammation of the hippocampus. 如請求項2所述之用途,其中該組成物係於腦損害發生後3天內投予。 The use as described in claim 2, wherein the composition is administered within 3 days after the occurrence of brain damage. 如請求項5所述之用途,其中該組成物係於腦損害發生後1小時內投予。 The use as described in claim 5, wherein the composition is administered within 1 hour after the occurrence of brain damage. 如請求項2所述之用途,其中該組成物係可減緩該創傷性腦損傷的神經元細胞氧化壓力,降低受該氧化壓力傷害的神經元細胞數。The use as described in claim 2, wherein the composition can alleviate the oxidative stress of the neurons in the traumatic brain injury and reduce the number of neurons damaged by the oxidative stress.
TW111115290A 2022-04-21 2022-04-21 Use of the composition for preparing a medicament for treating traumatic brain injury TWI839714B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
TW111115290A TWI839714B (en) 2022-04-21 2022-04-21 Use of the composition for preparing a medicament for treating traumatic brain injury

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
TW111115290A TWI839714B (en) 2022-04-21 2022-04-21 Use of the composition for preparing a medicament for treating traumatic brain injury

Publications (2)

Publication Number Publication Date
TW202342502A TW202342502A (en) 2023-11-01
TWI839714B true TWI839714B (en) 2024-04-21

Family

ID=89720534

Family Applications (1)

Application Number Title Priority Date Filing Date
TW111115290A TWI839714B (en) 2022-04-21 2022-04-21 Use of the composition for preparing a medicament for treating traumatic brain injury

Country Status (1)

Country Link
TW (1) TWI839714B (en)

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020006445A1 (en) * 2018-06-29 2020-01-02 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Biomarkers and treatment methods for traumatic brain injury associated impairments

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020006445A1 (en) * 2018-06-29 2020-01-02 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Biomarkers and treatment methods for traumatic brain injury associated impairments

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
期刊 Ho M.H., et al., "CCL5 via GPX1 activation protects hippocampal memory function after mild traumatic brain injury", Redox Biol. 46 2021; 102067. *

Also Published As

Publication number Publication date
TW202342502A (en) 2023-11-01

Similar Documents

Publication Publication Date Title
Torres-Cuevas et al. Brain oxidative damage in murine models of neonatal hypoxia/ischemia and reoxygenation
Zhang et al. Astaxanthin alleviates cerebral edema by modulating NKCC1 and AQP4 expression after traumatic brain injury in mice
Garcia et al. Developmental neurotoxicity elicited by prenatal or postnatal chlorpyrifos exposure: effects on neurospecific proteins indicate changing vulnerabilities.
Park et al. Human adipose tissue‐derived mesenchymal stem cells improve cognitive function and physical activity in ageing mice
Demars et al. Soluble amyloid precursor protein-α rescues age-linked decline in neural progenitor cell proliferation
Wang et al. Hydrogen Gas Attenuates Hypoxic‐Ischemic Brain Injury via Regulation of the MAPK/HO‐1/PGC‐1a Pathway in Neonatal Rats
Rossetti et al. Oxidation-reduction mechanisms in psychiatric disorders: A novel target for pharmacological intervention
Zhu et al. Neuroprotective effects of Astilbin on MPTP-induced Parkinson's disease mice: Glial reaction, α-synuclein expression and oxidative stress
Xu et al. Curcumin prevents brain damage and cognitive dysfunction during ischemic-reperfusion through the regulation of miR-7-5p
Guo et al. Cinnamic acid rescues behavioral deficits in a mouse model of traumatic brain injury by targeting miR-455-3p/HDAC2
Ma et al. Exogenous hydrogen sulfide ameliorates diabetes-associated cognitive decline by regulating the mitochondria-mediated apoptotic pathway and IL-23/IL-17 expression in db/db mice
Sinha et al. Amelioration of neurobehavioral and cognitive abilities of F1 progeny following dietary supplementation with Spirulina to protein malnourished mothers
Fu et al. Pelargonidin ameliorates MCAO-induced cerebral ischemia/reperfusion injury in rats by the action on the Nrf2/HO-1 pathway
Liu et al. (2 R, 3 S)-pinobanksin-3-cinnamate improves cognition and reduces oxidative stress in rats with vascular dementia
Yin et al. Dynamic balance of microglia and astrocytes involved in the remyelinating effect of ginkgolide B
Li et al. Curcumin suppress inflammatory response in traumatic brain injury via p38/MAPK signaling pathway
Liu et al. RvD1binding with FPR2 attenuates inflammation via Rac1/NOX2 pathway after neonatal hypoxic-ischemic injury in rats
Wang et al. An effective NADPH oxidase 2 inhibitor provides neuroprotection and improves functional outcomes in animal model of traumatic brain injury
Li et al. Retracted: Upregulated microRNA‐31 inhibits oxidative stress‐induced neuronal injury through the JAK/STAT3 pathway by binding to PKD1 in mice with ischemic stroke
US20220211724A1 (en) Composition comprising lipoic acid and vitamin d for the prevention and the treatment of neurodegenerative diseases and peripheral neuropathies
Cordaro et al. Physiological and biochemical changes in NRF2 pathway in aged animals subjected to brain injury
Deng et al. P2X7 receptor antagonism attenuates the intermittent hypoxia-induced spatial deficits in a murine model of sleep apnea via inhibiting neuroinflammation and oxidative stress
Elesawy et al. Postnatal baicalin ameliorates behavioral and neurochemical alterations in valproic acid-induced rodent model of autism: The possible implication of sirtuin-1/mitofusin-2/Bcl-2 pathway
Sheikh et al. The expression change of glial fibrillary acidic protein and tyrosine hydroxylase in substantia nigra of the Wistar rats exposed to chlorpyrifos: a novel environmental risk factor for Parkinson’s disease
Senzer Rationale for a phase III study of erythropoietin as a neurocognitive protectant in patients with lung cancer receiving prophylactic cranial irradiation