TW202340233A - Use of peptide in treating neurodegenerative disease or ameliorating cognitive function - Google Patents

Use of peptide in treating neurodegenerative disease or ameliorating cognitive function Download PDF

Info

Publication number
TW202340233A
TW202340233A TW112112599A TW112112599A TW202340233A TW 202340233 A TW202340233 A TW 202340233A TW 112112599 A TW112112599 A TW 112112599A TW 112112599 A TW112112599 A TW 112112599A TW 202340233 A TW202340233 A TW 202340233A
Authority
TW
Taiwan
Prior art keywords
amino acid
peptide
disease
acid sequence
seq
Prior art date
Application number
TW112112599A
Other languages
Chinese (zh)
Inventor
承鑫 劉
Original Assignee
大陸商首創生物技術有限公司
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 大陸商首創生物技術有限公司 filed Critical 大陸商首創生物技術有限公司
Publication of TW202340233A publication Critical patent/TW202340233A/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/08Peptides having 5 to 11 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/10Peptides having 12 to 20 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/08Linear peptides containing only normal peptide links having 12 to 20 amino acids

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Biomedical Technology (AREA)
  • Organic Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Neurosurgery (AREA)
  • Public Health (AREA)
  • Neurology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Psychology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The present invention relates to use of a peptide in treating a neurodegenerative disease or ameliorating a cognitive function. The present invention relates to the treatment of neurodegenerative diseases and cognitive disorders, and the amelioration and improvement of cognitive functions. In particular, the present invention relates to use of a peptide in preventing or treating a neurodegenerative disease or a cognitive disorder, preferably Alzheimer's disease and Parkinson's disease. The present invention further relates to use of a peptide in ameliorating or improving a cognitive function in a subject.

Description

肽用於治療神經退行性疾病或改善認知功能的用途Use of peptides to treat neurodegenerative diseases or improve cognitive function

本發明涉及神經退行性疾病和認知障礙症的治療;以及認知功能的改善和提高。具體而言,本發明涉及肽用於預防或治療神經退行性疾病和認知障礙症,優選阿爾茨海默病和帕金森病的用途。本發明還涉及肽用於改善或提高受試者認知功能的用途。 The present invention relates to the treatment of neurodegenerative diseases and cognitive impairment; and the improvement and improvement of cognitive function. In particular, the present invention relates to the use of peptides for the prevention or treatment of neurodegenerative diseases and cognitive disorders, preferably Alzheimer's disease and Parkinson's disease. The invention also relates to the use of peptides for improving or enhancing cognitive function in a subject.

神經退行性疾病或神經退化性疾病(Neurodegenerative Disorder或Neurodegenerative Disease)是神經元逐漸退化所直接導致的疾病。退行性過程可涉及神經元結構的進行性損失、神經元功能的進行性損失、或進行性神經元細胞死亡。這種進行性神經退行經常導致身體殘疾和精神惡化。許多神經退行性疾病是嚴重的進行性和不間斷疾病,並且治療方法很少。阿爾茨海默病(AD)和帕金森病(PD)是最主要的兩種神經退行性疾病。 認知障礙症(Neurocognitive Disorder)也稱為癡呆症(Dementia),是腦部疾病的其中一類,此症導致思考能力和記憶力長期而逐漸地退化,並使個人日常生活活動受到影響。最常見的癡呆症類型是阿爾茨海默病,阿爾茨海默病患者占所有癡呆症患者人數的50%到70%。癡呆症影響全球三千六百萬人口。大約10%的人口,會在有生之年中發病。癡呆症與年齡(老化)息息相關,約3%的人口在65到74歲之間得到癡呆症,另外19%的人口則在75到84歲之間,而將近一半的人口超過85歲得到癡呆症。 阿爾茨海默病(Alzheimer's Disease, AD)是一種發病進程緩慢、隨著時間不斷惡化的慢性神經退行性疾病。最常見的早期症狀為喪失短期記憶,當疾病逐漸進展,症狀可能逐漸出現,包括語言障礙、定向障礙、情緒不穩、喪失動機、無法自理和許多行為問題。當情況惡化時,患者往往會因此和家庭或社會脫節,並逐漸喪失身體機能,最終導致死亡。雖然疾程因人而異,但診斷後的平均餘命約為三到九年。在AD中,海馬體(hippocampus)是首先受到損傷的區域,表現症狀為記憶力衰退以及方向知覺的喪失(見Yangling Mu et al., Adult hippocampal neurogenesis and its role in Alzheimer's disease, Mol Neurodegener, 2011; 6: 85)。AD的進程與大腦中的β-澱粉樣蛋白(Aβ)斑塊沉積和Tau蛋白有關(見Clive Ballard et al., Alzheimer's disease, The Lancet. 2011 March, 377(9770): 1019–1031)。最近研究發現,神經炎症或小膠質細胞的啟動參與了阿爾茨海默病中tau蛋白纏結在新皮質中的擴散,進而導致阿爾茨海默病患者認知功能障礙的發生(見Tharick A. Pascoal et al., Microglial activation and tau propagate jointly across Braak stages, Nature Medicine, 27, pages1592–1599(2021 August))。小膠質細胞啟動是人類大腦免疫反應的一部分,是與阿爾茲海默病發展相關的關鍵因素。小膠質細胞的過度啟動不僅僅是一種炎症附帶現象,更是一種關鍵的上游機制,對AD的發展至關重要。研究還發現,神經炎症和某些炎症細胞因數參與AD的病理學(見Rishika Dhapola et al., Recent advances in molecular pathways and therapeutic implications targeting neuroinflammation for Alzheimer’s disease, Inflammopharmacology volume 29, pages1669–1681(2021 November))。因此,減少Aβ斑塊沉積、緩解神經炎症以及抑制小膠質細胞的啟動可視為治療或緩解AD的有效手段。 帕金森病(Parkinson's disease,PD)是一種影響中樞神經系統的慢性神經退行性疾病,主要影響運動神經系統。它的症狀通常隨時間緩慢出現,早期最明顯的症狀為顫抖、肢體僵硬、運動功能減退和步態異常,也可能有認知和行為問題。認知障礙症在病情嚴重的患者中相當常見。帕金森病的主要病理變化發生在中腦黑質腹側的緻密部,主要症狀大多數因為黑質緻密部的多巴胺性神經元退化(Jose A Obeso et al., Functional organization of the basal ganglia: therapeutic implications for Parkinson's disease, Movement Disorders, Volume 23, Issue S3 p. S548-S559, Sept. 2008)。司來吉蘭是一種單胺氧化酶(MAO)-B抑制劑(MAOI),2006年美國FDA批准司來吉蘭用於治療PD患者。 本領域中存在有效治療神經退行性疾病或者由神經退行性疾病引起的認知障礙症的藥物的需求。 WO2013/173941和CN104321337A公開了一種具有11至14個氨基酸殘基的鎮痛肽,經序列比對匹配兔α1-抗蛋白酶的片段。WO2016/165101公開了所述肽可以有效抑制HCV複製。此外,WO2016/165102和CN107847550A公開了該肽用於治療腦卒中(一種急性的腦血管疾病),驗證了所述肽能夠通過腦血屏障,並通過體外細胞實驗驗證了所述肽對PC12細胞免受谷氨酸和過氧化氫誘導的細胞毒性的保護作用。這些檔通過引用結合到本文中。WO2016/165102中測試的PC12細胞是一種衍生自顯示神經節細胞的某些特徵的大鼠腎上腺髓質的嗜鉻細胞瘤,其特別用於建立細胞缺氧損傷模型,從而類比急性腦血管疾病中的迅速缺氧和毒性狀態。然而,阿爾茨海默病和帕金森病屬於慢性疾病,它們並非由腦血管栓塞或破裂而導致的急性大腦缺氧和毒性引起。現有技術尚未提及所述肽用於治療AD或PD的可能性。 Neurodegenerative Disorder or Neurodegenerative Disease is a disease directly caused by the gradual degeneration of neurons. Degenerative processes may involve progressive loss of neuronal structure, progressive loss of neuronal function, or progressive neuronal cell death. This progressive neurodegeneration often leads to physical disability and mental deterioration. Many neurodegenerative diseases are severe, progressive and ongoing diseases with few treatments. Alzheimer's disease (AD) and Parkinson's disease (PD) are the two most important neurodegenerative diseases. Neurocognitive Disorder, also known as Dementia, is a type of brain disease that causes long-term and gradual deterioration of thinking ability and memory, and affects a person's daily life activities. The most common type of dementia is Alzheimer's disease, which accounts for 50% to 70% of all dementia patients. Dementia affects 36 million people worldwide. About 10% of the population will develop the disease during their lifetime. Dementia is closely related to age (aging). About 3% of the population develops dementia between the ages of 65 and 74, another 19% of the population develops between the ages of 75 and 84, and nearly half of the population over the age of 85 develops dementia. . Alzheimer's Disease (AD) is a chronic neurodegenerative disease that develops slowly and worsens over time. The most common early symptom is loss of short-term memory. As the disease progresses, symptoms may gradually appear, including language difficulties, disorientation, mood swings, loss of motivation, inability to care for oneself, and many behavioral problems. When the condition worsens, patients often become disconnected from family or society, gradually lose physical function, and eventually die. Although the course of the disease varies from person to person, the average life expectancy after diagnosis is about three to nine years. In AD, the hippocampus is the first area to be damaged, with symptoms such as memory decline and loss of directional awareness (see Yangling Mu et al., Adult hippocampal neurogenesis and its role in Alzheimer's disease, Mol Neurodegener, 2011; 6 : 85). The progression of AD is related to the deposition of beta-amyloid (Aβ) plaques and tau protein in the brain (see Clive Ballard et al., Alzheimer's disease, The Lancet. 2011 March, 377(9770): 1019–1031). Recent studies have found that neuroinflammation or microglial priming is involved in the spread of tau tangles in the neocortex in Alzheimer's disease, leading to the occurrence of cognitive dysfunction in Alzheimer's disease patients (see Tharick A. Pascoal et al., Microglial activation and tau propagate jointly across Braak stages, Nature Medicine, 27, pages1592–1599(2021 August)). Microglial priming is part of the immune response in the human brain and is a key factor associated with the development of Alzheimer's disease. Excessive activation of microglia is not only an epiphenomenon of inflammation, but also a key upstream mechanism that is crucial to the development of AD. Research has also found that neuroinflammation and certain inflammatory cytokines are involved in the pathology of AD (see Rishika Dhapola et al., Recent advances in molecular pathways and therapeutic implications targeting neuroinflammation for Alzheimer's disease, Inflammopharmacology volume 29, pages1669–1681 (2021 November) ). Therefore, reducing Aβ plaque deposition, alleviating neuroinflammation, and inhibiting the initiation of microglia can be regarded as effective means to treat or alleviate AD. Parkinson's disease (PD) is a chronic neurodegenerative disease affecting the central nervous system, mainly affecting the motor nervous system. Its symptoms usually appear slowly over time. The most obvious early symptoms are tremor, limb stiffness, reduced motor function and abnormal gait. Cognitive and behavioral problems may also be present. Dementia is quite common in severely ill patients. The main pathological changes of Parkinson's disease occur in the ventral substantia nigra pars compacta of the midbrain, and most of the main symptoms are due to the degeneration of dopamine neurons in the substantia nigra pars compacta (Jose A Obeso et al., Functional organization of the basal ganglia: therapeutic implications for Parkinson's disease, Movement Disorders, Volume 23, Issue S3 p. S548-S559, Sept. 2008). Selegiline is a monoamine oxidase (MAO)-B inhibitor (MAOI). In 2006, the US FDA approved selegiline for the treatment of PD patients. There is a need in the art for drugs that are effective in treating neurodegenerative diseases or cognitive impairment caused by neurodegenerative diseases. WO2013/173941 and CN104321337A disclose an analgesic peptide with 11 to 14 amino acid residues, which matches the fragment of rabbit α1-antiprotease through sequence alignment. WO2016/165101 discloses that the peptide can effectively inhibit HCV replication. In addition, WO2016/165102 and CN107847550A disclose that the peptide is used to treat stroke (an acute cerebrovascular disease), verify that the peptide can pass through the brain-blood barrier, and verify that the peptide is immune to PC12 cells through in vitro cell experiments. Protection from glutamate- and hydrogen peroxide-induced cytotoxicity. These documents are incorporated into this article by reference. The PC12 cells tested in WO2016/165102 are a type of pheochromocytoma derived from the rat adrenal medulla that exhibit certain characteristics of ganglion cells and are particularly useful in establishing a model of cellular hypoxic injury, thereby analogous to that seen in acute cerebrovascular disease. rapid hypoxic and toxic state. However, Alzheimer's disease and Parkinson's disease are chronic diseases that are not caused by acute cerebral hypoxia and toxicity caused by cerebral blood vessel embolism or rupture. The prior art does not mention the possibility of using said peptides to treat AD or PD.

本發明目的包括提供用於治療神經退行性疾病和認知障礙症的藥物,以及提供用於改善或提高受試者認知功能的活性成分或組合物。本發明目的還包括提供用於治療阿爾茨海默病和帕金森病的藥物。 本發明的技術問題之一通過提供本發明的肽以及包含所述肽的組合物來解決。 本發明涉及一種肽,其具有SEQ ID NO: 1或2的氨基酸序列或其變體或片段。“變體”和“片段”意指與本發明肽相比具有一定的氨基酸殘基變化並且基本上保留與所述肽的相同或類似的生物功能或活性的肽。氨基酸序列的變體可以是與所述氨基酸序列具有至少某個百分比同一性的氨基酸序列的變體,或者與所述氨基酸序列相比具有至少一個或多個氨基酸突變的氨基酸序列的變體。例如,所述變體可以與SEQ ID NO: 1或2的氨基酸序列具有至少70%的同一性。或者,所述所述變體可以在SEQ ID NO: 1或2的氨基酸序列中具有0至4個氨基酸突變。氨基酸序列的片段可以是與所述氨基酸序列相比在C-端和/N-端具有一個或多個缺失的氨基酸序列。所述缺失可以是1-5個,例如1、2、3、4或5個。氨基酸序列的片段的長度可以是8至20個氨基酸,例如10至15個氨基酸,例如10、11、12、13、14或15個氨基酸。 在一個方面中,本發明的肽選自: a) 包含與DEAQETAVSSH(SEQ ID NO: 1)具有至少70%同一性的氨基酸序列的肽; b) 包含在DEAQETAVSSH(SEQ ID NO: 1)中具有0至4個氨基酸突變的氨基酸序列的肽; c) 包含與DEAQETAVSSHEQD(SEQ ID NO: 2)具有至少70%同一性的氨基酸序列的肽;或 d) 包含在DEAQETAVSSHEQD(SEQ ID NO: 2)中具有0至4個氨基酸突變的氨基酸序列的肽。 在一個方面,本發明肽包含與SEQ ID NO: 1具有至少70%、至少75%、至少80%、至少85%、至少90%、至少95%、至少98%、至少99%或者100%同一性的氨基酸序列。 在一個方面,本發明肽包含在SEQ ID NO: 1中或相對於SEQ ID NO: 1具有0至4個氨基酸突變,例如0至3個,例如1至3個,例如0、1、2、3或4個氨基酸突變的氨基酸序列。 在一個方面,本發明肽包含與SEQ ID NO: 2具有至少70%、至少75%、至少80%、至少85%、至少90%、至少95%、至少98%、至少99%或者100%同一性的氨基酸序列。 在一個方面,本發明肽包含在SEQ ID NO: 2中或相對於SEQ ID NO: 2具有0至4個氨基酸突變,例如0至3個,例如1至3個,例如0、1、2、3或4個氨基酸突變的氨基酸序列。 氨基酸突變可以選自氨基酸的添加、缺失或取代。添加可以是氨基酸序列中兩個氨基酸殘基之間插入氨基酸,或者是在氨基酸序列C-端或者N-端的添加。缺失可以是氨基酸序列中兩個氨基酸殘基之間刪除氨基酸,或者是氨基酸序列C-端或者N-端的缺失。取代可以是氨基酸序列中任意氨基酸殘基的取代。優選地,氨基酸的添加、缺失或取代發生在序列的C-端或者N-端。取代可以是保守取代。保守取代是指性狀相近的氨基酸分子之間的取代,其中的性狀包含分子的離子性、疏水性和分子量等。性狀相近的氨基酸可以按以下分類:脂肪族氨基酸(例如甘氨酸, 丙氨酸, 纈氨酸, 亮氨酸, 異亮氨酸);含羥基或含硫氨基酸(例如絲氨酸, 半胱氨酸, 蘇氨酸, 甲硫氨酸);環狀氨基酸(例如脯氨酸);芳香族氨基酸(例如苯丙氨酸, 酪氨酸, 色氨酸);鹼性氨基酸(例如組氨酸, 賴胺酸, 精氨酸)和酸性或醯胺類氨基酸(例如天冬氨酸, 谷氨酸, 天冬醯胺, 穀氨醯胺)。 本領域技術人員理解,構成肽、多肽或蛋白的二十種常見的氨基酸及其字母縮寫如下表1所示。技術人員可以例如根據下表1或者公知常識確定本發明肽的氨基酸殘基順序和組成。 氨基酸突變可以位於氨基酸序列的C-端和/或N-端。例如,所述肽可以在SEQ ID NO: 1的C-端或者N-端具有1個、2個或3個氨基酸添加。例如,所述肽可以在SEQ ID NO: 2的C-端或者N-端具有1個、2個或3個氨基酸缺失。 在一個方面,本發明的肽可以具有8至20個氨基酸的長度,優選10至18個氨基酸長度,更優選11至14個氨基酸長度。例如,所述肽的長度可以為10、11、12、13、14、15、16、17或18個氨基酸。 在一個方面,本發明的肽可以選自 1) 包含SEQ ID NO: 1的氨基酸序列、基本上由SEQ ID NO: 1的氨基酸序列組成或者由SEQ ID NO: 1的氨基酸序列表示的肽;或者 2) 包含SEQ ID NO: 2的氨基酸序列、基本上由SEQ ID NO: 2的氨基酸序列組成或者由SEQ ID NO: 2的氨基酸序列表示的肽。 在一個方面,本發明的肽由SEQ ID NO: 1的氨基酸序列組成。或者,本發明的肽由SEQ ID NO: 2的氨基酸序列組成。 在一個方面,本發明涉及本文所述的肽在製備用於預防、治療或改善有需要的受試者中神經退行性疾病的組合物中的用途。在一個方面,本發明涉及肽,其用於預防、治療或改善有需要的受試者中神經退行性疾病。在一個方面,本發明涉及一種預防、治療或改善有需要的受試者中神經退行性疾病的方法,所述方法包括將有效量的肽給予需要的受試者。神經退行性疾病可包括認知障礙症。 在一個方面,神經退行性疾病是慢性神經退行性疾病。 在一個方面,本發明涉及本文所述的肽在製備用於預防、治療或改善有需要的受試者中認知障礙症的組合物中的用途。在一個方面,本發明涉及肽,其用於預防、治療或改善有需要的受試者中認知障礙症。在一個方面,本發明涉及一種預防、治療或改善有需要的受試者中認知障礙症的方法,所述方法包括將有效量的肽給予需要的受試者。認知障礙症可以由神經退行性疾病引起。或者,認知障礙症可以伴隨神經退行性疾病。 在一個方面,神經退行性疾病或認知障礙症選自阿爾茨海默病或帕金森病。因此,本發明的肽可以用於預防、治療或改善阿爾茨海默病。此外,本發明的肽可以用於預防、治療或改善帕金森病。 認知可以是機體認識和獲取知識的智慧加工過程,涉及學習、記憶、語言、思維、精神、情感等一系列隨意、心理和社會行為。認知障礙可以包括與上述學習記憶以及思維判斷有關的大腦高級智慧加工過程出現異常,從而引起嚴重學習、記憶障礙,同時伴有失語或失用或失認或失行等改變的病理過程。 在一個方面,本發明涉及本文所述的肽在製備用於改善、增強或恢復有需要的受試者中認知功能或認知能力的組合物中的用途。在一個方面,本發明涉及肽,其用於改善、增強或恢復有需要的受試者中認知功能或認知能力。在一個方面,本發明涉及一種改善、增強或恢復有需要的受試者中認知功能或認知能力的方法,所述方法包括將有效量的肽給予需要的受試者。在該方面中,認知功能或認知能力包括感知能力、思維邏輯能力、記憶能力、語言能力、學習能力、情緒控制能力、社交能力或注意力。在該方面中,所述受試者具有認知功能或認知能力衰退,例如年齡老化而引致的或伴隨年齡老化的認知功能或認知能力衰退。或者,所述受試者可以患有認知障礙症或神經退行性疾病引起,優選由神經退行性疾病引起的認知障礙症,更優選阿爾茨海默病或帕金森病。 在一個方面,認知障礙症特徵在於或表現在於溝通障礙、判斷力差、難以進行簡單任務、遺漏或放錯物件、語言障礙、性格突變、行為能力下降、時間及空間感混亂、理解能力下降、問題解決能力下降、注意力下降、社交能力下降、感知障礙、思維邏輯障礙或記憶障礙。 在一個方面,神經退行性疾病或認知障礙症為阿爾茨海默病。所述阿爾茨海默病特徵在於在大腦特別是海馬體中β-澱粉樣蛋白(Aβ)沉積、神經炎症、和/或膠質細胞異常或活化。阿爾茨海默病的預防、治療或改善通過減少或緩解在大腦中β-澱粉樣蛋白(Aβ)沉積、神經炎症和/或膠質細胞異常或活化來實現。 在一個方面,本發明涉及本文所述的肽在製備用於減少或緩解受試者大腦特別是海馬體中β-澱粉樣蛋白(Aβ)沉積、神經炎症和/或膠質細胞異常或活化的組合物中的用途。所述受試者可以是神經退行性疾病患者或認知障礙症患者,優選阿爾茨海默病患者。 本文所述的肽可以有效減少或緩解受試者大腦例如海馬體中的Aβ或Aβ斑塊沉積。此外,所述肽可以減少或緩解受試者大腦例如海馬體或者皮層中炎症因數的表達或水準。炎症因數可以選自IL-1β、IL-6或TNF-α。所述肽還可以減少或緩解大腦海馬體膠質細胞異常或活化。膠質細胞也可稱為神經膠質細胞,可以選自星形膠質細胞或小膠質細胞,優選小膠質細胞。 在一個方面,神經退行性疾病或認知障礙症為帕金森病。所述帕金森病特徵在於大腦黑質神經元異常或損傷。帕金森病的預防、治療或改善通過減少或緩解大腦黑質神經元異常或損傷來實現。 在一個方面,本發明涉及本文所述的肽在製備用於減少或緩解受試者大腦黑質神經元異常或損傷的組合物中的用途。所述受試者可以是神經退行性疾病患者或認知障礙症患者,優選帕金森病患者。 在一個方面,受試者或患者是哺乳動物。在一個方面,受試者或患者是人,例如中年人或老年人,優選老年人。中年人可以是40至59歲的人或者45至59歲的人。老年人可以是60歲以上、65歲以上、70歲以上、75歲以上或80歲以上的人。 在一個方面,受試者可以是認知功能或認知能力衰退的受試者,例如年齡老化而引致的或伴隨年齡老化的認知功能或認知能力衰退。所述衰退通常是指慢性衰退。 本發明的組合物包含本文所述的肽。本發明的組合物還可以包含藥用輔料、溶媒或載體;或者人體可接受的藥用輔料、溶媒或載體。組合物可以是藥物組合物或者營養組合物。藥物組合物可以是藥物。營養組合物可為營養補充劑、營養補充品、膳食補充劑、飲食補充劑、保健食品、保健品、營養品或健康食品的形式。 本文所述的“組合物”總體上可以包括改善或提高受試者(例如人)的健康況狀或治療所述受試者的疾病的物質或配方。 本文所述的“營養組合物”可以指滿足受試者(例如人)至少部分營養物需求、調節所述受試者身體機能、調理受試者生理功能和/或改善受試者健康狀態的可服用的物質或配方。例如,營養組合物可以是啟動、調節或改善認知功能或認知能力的保健品、功能性食品、膳食補充劑、營養品或健康食品。 本文所述的“藥物組合物”可以指用於預防、治療、緩解、改善、調理或調節受試者(例如人)的疾病、病症、症狀或病狀的可服用的物質或配方。例如,藥物組合物可以是藥物的形式,包括口服藥物,例如藥片、藥丸或膠囊等。 本發明的多肽或組合物的服用可以是口服形式。用於口服的組合物可以使用本領域已知的載體、溶媒或賦形劑以適合於口服施用的劑量進行配製。這樣的載體使得組合物能夠配製成適合於受試者攝入的片劑、丸劑、膠囊、液體、凝膠、糖漿、漿液、混懸劑等。 此外,本發明的多肽或組合物可被製備成注射劑,例如肌肉注射劑、腹膜內注射劑、腹腔注射劑、皮下注射劑或者靜脈注射劑。注射劑可以呈溶液、乳濁液或混懸液形式,以及臨用前配成溶液或混懸液的粉末或濃溶液。本領域技術人員理解,注射劑中還可包含溶劑例如水、等滲劑、緩衝劑、防腐劑、助溶劑、增溶劑、助懸劑和乳化劑等。 術語“有效量”或“治療有效量”是指本發明的肽或組合物有效治療受試者的疾病或病症或者改善或提高其健康狀況的量。改善或提高其健康狀況包括提高或改善認知功能或能力。本發明的肽可以按0.01mg/kg至10mg/kg、0.02 mg/kg至8mg/kg、0.04mg/kg至5mg/kg、0.05mg/kg至1 mg/kg、0.1mg/kg至0.8mg/kg或0.2mg/kg至0.5mg/kg的量給予受試者。例如,本發明的肽可以按0.04mg/kg、0.16 mg/kg或0.64mg/kg的量給予受試者。治療有效量可以是這些劑量。本發明的肽或組合物可以按一天一次的間隔給藥,持續至少1周,例如1至4周。 本發明的肽可以按肽組合的形式給予受試者或製備成組合物。本發明的組合物可包含這樣的肽組合。例如,所述肽組合包含具有SEQ ID NO: 1的氨基酸序列或其變體的第一肽以及具有SEQ ID NO: 2的氨基酸序列或其變體的第二肽。 The object of the present invention includes providing drugs for treating neurodegenerative diseases and cognitive impairment, as well as providing active ingredients or compositions for improving or enhancing the cognitive function of subjects. It is also an object of the present invention to provide medicaments for the treatment of Alzheimer's disease and Parkinson's disease. One of the technical problems of the present invention is solved by providing the peptides of the present invention and compositions comprising said peptides. The present invention relates to a peptide having the amino acid sequence of SEQ ID NO: 1 or 2 or a variant or fragment thereof. "Variant" and "fragment" mean a peptide that has certain amino acid residue changes compared to the peptide of the invention and substantially retains the same or similar biological function or activity as the peptide. A variant of an amino acid sequence may be a variant of an amino acid sequence that has at least a certain percent identity with the amino acid sequence, or a variant of an amino acid sequence that has at least one or more amino acid mutations compared with the amino acid sequence. For example, the variant may be at least 70% identical to the amino acid sequence of SEQ ID NO: 1 or 2. Alternatively, the variant may have 0 to 4 amino acid mutations in the amino acid sequence of SEQ ID NO: 1 or 2. A fragment of an amino acid sequence may be an amino acid sequence having one or more deletions at the C-terminus and/or N-terminus compared to the amino acid sequence. The deletions may be 1-5, such as 1, 2, 3, 4 or 5. Fragments of the amino acid sequence may be 8 to 20 amino acids in length, such as 10 to 15 amino acids, such as 10, 11, 12, 13, 14 or 15 amino acids. In one aspect, the peptides of the invention are selected from: a) peptides comprising an amino acid sequence having at least 70% identity with DEAQETAVSSH (SEQ ID NO: 1); b) peptides comprising in DEAQETAVSSH (SEQ ID NO: 1) having A peptide with an amino acid sequence of 0 to 4 amino acid mutations; c) A peptide comprising an amino acid sequence that is at least 70% identical to DEAQETAVSSHEQD (SEQ ID NO: 2); or d) Contained in DEAQETAVSSHEQD (SEQ ID NO: 2) Peptides with amino acid sequences of 0 to 4 amino acid mutations. In one aspect, the peptides of the invention comprise at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100% identical to SEQ ID NO: 1 sexual amino acid sequence. In one aspect, the peptide of the invention is comprised in SEQ ID NO: 1 or has 0 to 4 amino acid mutations relative to SEQ ID NO: 1, such as 0 to 3, such as 1 to 3, such as 0, 1, 2, Amino acid sequence with 3 or 4 amino acid mutations. In one aspect, the peptides of the invention comprise at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, at least 95%, at least 98%, at least 99%, or 100% identical to SEQ ID NO: 2 sexual amino acid sequence. In one aspect, the peptide of the invention is comprised in SEQ ID NO: 2 or has 0 to 4 amino acid mutations relative to SEQ ID NO: 2, such as 0 to 3, such as 1 to 3, such as 0, 1, 2, Amino acid sequence with 3 or 4 amino acid mutations. Amino acid mutations can be selected from additions, deletions or substitutions of amino acids. The addition can be the insertion of an amino acid between two amino acid residues in the amino acid sequence, or the addition of an amino acid at the C-terminus or N-terminus of the amino acid sequence. The deletion can be the deletion of an amino acid between two amino acid residues in the amino acid sequence, or the deletion of the C-terminus or N-terminus of the amino acid sequence. The substitution may be of any amino acid residue in the amino acid sequence. Preferably, the addition, deletion or substitution of amino acids occurs at the C-terminus or N-terminus of the sequence. Substitutions may be conservative substitutions. Conservative substitution refers to the substitution between amino acid molecules with similar properties, including the ionicity, hydrophobicity and molecular weight of the molecule. Amino acids with similar properties can be classified as follows: aliphatic amino acids (such as glycine, alanine, valine, leucine, isoleucine); hydroxyl-containing or sulfur-containing amino acids (such as serine, cysteine, threonine) amino acids, methionine); cyclic amino acids (such as proline); aromatic amino acids (such as phenylalanine, tyrosine, tryptophan); basic amino acids (such as histidine, lysine) , arginine) and acidic or amide amino acids (such as aspartic acid, glutamic acid, asparagine, glutamine). Those skilled in the art understand that the twenty common amino acids that constitute peptides, polypeptides or proteins and their abbreviations are shown in Table 1 below. The skilled person can determine the amino acid residue sequence and composition of the peptide of the present invention, for example, based on Table 1 below or common knowledge. Amino acid mutations may be located at the C-terminus and/or N-terminus of the amino acid sequence. For example, the peptide may have 1, 2 or 3 amino acids added at the C-terminus or N-terminus of SEQ ID NO: 1. For example, the peptide may have 1, 2 or 3 amino acid deletions at the C-terminus or N-terminus of SEQ ID NO: 2. In one aspect, the peptides of the invention may be from 8 to 20 amino acids in length, preferably from 10 to 18 amino acids in length, more preferably from 11 to 14 amino acids in length. For example, the peptide may be 10, 11, 12, 13, 14, 15, 16, 17 or 18 amino acids in length. In one aspect, the peptide of the invention may be selected from 1) a peptide comprising, consisting essentially of, or represented by the amino acid sequence of SEQ ID NO: 1; or 2) A peptide comprising, consisting essentially of, or represented by the amino acid sequence of SEQ ID NO: 2. In one aspect, the peptide of the invention consists of the amino acid sequence of SEQ ID NO: 1. Alternatively, the peptide of the invention consists of the amino acid sequence of SEQ ID NO: 2. In one aspect, the invention relates to the use of a peptide described herein in the preparation of a composition for preventing, treating or ameliorating a neurodegenerative disease in a subject in need thereof. In one aspect, the invention relates to peptides for use in preventing, treating or ameliorating neurodegenerative diseases in a subject in need thereof. In one aspect, the invention relates to a method of preventing, treating or ameliorating a neurodegenerative disease in a subject in need thereof, comprising administering an effective amount of a peptide to the subject in need thereof. Neurodegenerative diseases can include dementia. In one aspect, the neurodegenerative disease is a chronic neurodegenerative disease. In one aspect, the invention relates to the use of a peptide described herein for the preparation of a composition for preventing, treating or ameliorating dementia in a subject in need thereof. In one aspect, the invention relates to peptides for use in preventing, treating or ameliorating dementia in a subject in need thereof. In one aspect, the invention relates to a method of preventing, treating or ameliorating dementia in a subject in need thereof, comprising administering an effective amount of a peptide to the subject in need thereof. Dementia can be caused by neurodegenerative diseases. Alternatively, dementia can accompany neurodegenerative diseases. In one aspect, the neurodegenerative disease or cognitive disorder is selected from Alzheimer's disease or Parkinson's disease. Therefore, the peptides of the present invention can be used to prevent, treat or improve Alzheimer's disease. In addition, the peptide of the present invention can be used to prevent, treat or improve Parkinson's disease. Cognition can be the intelligent processing process by which the body recognizes and acquires knowledge, involving a series of random, psychological and social behaviors such as learning, memory, language, thinking, spirit, and emotion. Cognitive impairment can include abnormalities in the brain's advanced intellectual processing related to the above-mentioned learning, memory, and thinking judgment, causing severe learning and memory impairments, accompanied by pathological processes such as aphasia, apraxia, agnosia, or aphasia. In one aspect, the invention relates to the use of a peptide described herein in the preparation of a composition for improving, enhancing or restoring cognitive function or cognitive ability in a subject in need thereof. In one aspect, the invention relates to peptides for use in improving, enhancing or restoring cognitive function or cognitive ability in a subject in need thereof. In one aspect, the invention relates to a method of improving, enhancing or restoring cognitive function or cognitive ability in a subject in need thereof, comprising administering to the subject in need thereof an effective amount of a peptide. In this aspect, cognitive function or cognitive ability includes perceptual ability, logical thinking ability, memory ability, language ability, learning ability, emotional control ability, social ability or attention. In this aspect, the subject has a cognitive function or cognitive ability decline, such as a cognitive function or cognitive ability decline caused by or accompanying aging. Alternatively, the subject may suffer from cognitive impairment or a neurodegenerative disease, preferably a cognitive impairment caused by a neurodegenerative disease, more preferably Alzheimer's disease or Parkinson's disease. In one aspect, dementia is characterized or manifested by communication difficulties, poor judgment, difficulty performing simple tasks, missing or misplacing objects, language impairment, personality changes, decreased behavioral abilities, confused sense of time and space, decreased comprehension, Decreased problem-solving skills, decreased concentration, decreased social skills, impairments in perception, logical thinking, or memory impairment. In one aspect, the neurodegenerative disease or cognitive disorder is Alzheimer's disease. Alzheimer's disease is characterized by beta-amyloid (Aβ) deposition, neuroinflammation, and/or glial cell abnormalities or activation in the brain, particularly the hippocampus. Prevention, treatment or improvement of Alzheimer's disease is achieved by reducing or alleviating beta-amyloid (Aβ) deposition, neuroinflammation and/or glial cell abnormalities or activation in the brain. In one aspect, the invention relates to the use of the peptides described herein in the preparation of combinations for reducing or alleviating beta-amyloid (Aβ) deposition, neuroinflammation and/or glial abnormality or activation in the brain of a subject, particularly in the hippocampus. uses in things. The subject may be a neurodegenerative disease patient or a cognitive impairment patient, preferably an Alzheimer's disease patient. The peptides described herein can be effective in reducing or alleviating Aβ or Aβ plaque deposition in a subject's brain, such as the hippocampus. Furthermore, the peptide can reduce or alleviate the expression or levels of inflammatory factors in the subject's brain, such as the hippocampus or cortex. The inflammatory factor can be selected from IL-1β, IL-6 or TNF-α. The peptides may also reduce or alleviate glial cell abnormalities or activation in the brain's hippocampus. Glial cells can also be called glial cells and can be selected from astrocytes or microglia, with microglia being preferred. In one aspect, the neurodegenerative disease or cognitive disorder is Parkinson's disease. Parkinson's disease is characterized by abnormality or damage to neurons in the substantia nigra of the brain. The prevention, treatment or improvement of Parkinson's disease is achieved by reducing or alleviating neuronal abnormalities or damage in the substantia nigra of the brain. In one aspect, the invention relates to the use of a peptide described herein in the preparation of a composition for reducing or alleviating abnormality or damage to neurons in the substantia nigra of the brain of a subject. The subject may be a neurodegenerative disease patient or a cognitive impairment patient, preferably a Parkinson's disease patient. In one aspect, the subject or patient is a mammal. In one aspect, the subject or patient is a human, such as a middle-aged or elderly person, preferably an elderly person. Middle-aged people can be people between 40 and 59 years old or people between 45 and 59 years old. An elderly person can be a person over 60, over 65, over 70, over 75 or over 80. In one aspect, the subject may be a subject with cognitive function or cognitive ability decline, such as cognitive function or cognitive ability decline caused by or accompanying aging. The decline generally refers to chronic decline. Compositions of the invention comprise peptides described herein. The composition of the present invention may also contain pharmaceutical excipients, solvents or carriers; or pharmaceutical excipients, solvents or carriers acceptable to the human body. The composition may be a pharmaceutical composition or a nutritional composition. The pharmaceutical composition may be a drug. The nutritional composition may be in the form of a nutritional supplement, nutritional supplement, dietary supplement, dietary supplement, nutraceutical, nutraceutical, nutraceutical or health food. "Compositions" as described herein may generally include substances or formulations that improve or enhance the health condition of a subject (eg, a human) or treat a disease in the subject. "Nutritional composition" as described herein may refer to meeting at least part of the nutrient requirements of a subject (such as a human), regulating the body functions of the subject, regulating the physiological functions of the subject, and/or improving the health status of the subject Substances or formulations that may be taken. For example, the nutritional composition may be a nutraceutical, functional food, dietary supplement, nutraceutical or health food that initiates, modulates or improves cognitive function or cognitive ability. As used herein, a "pharmaceutical composition" may refer to an ingestible substance or formulation for the prevention, treatment, alleviation, amelioration, conditioning, or modulation of a disease, disorder, symptom, or condition in a subject (eg, a human). For example, pharmaceutical compositions may be in the form of pharmaceuticals, including oral medications, such as tablets, pills, capsules, and the like. Administration of the polypeptides or compositions of the invention may be in oral form. Compositions for oral administration may be formulated in dosages suitable for oral administration using carriers, vehicles or excipients known in the art. Such carriers enable the compositions to be formulated as tablets, pills, capsules, liquids, gels, syrups, slurries, suspensions, and the like suitable for ingestion by a subject. In addition, the polypeptide or composition of the present invention can be prepared as an injection, such as intramuscular injection, intraperitoneal injection, intraperitoneal injection, subcutaneous injection or intravenous injection. Injectables may be in the form of solutions, emulsions or suspensions, as well as powders or concentrated solutions for solution or suspension before use. Those skilled in the art understand that injections may also contain solvents such as water, isotonic agents, buffers, preservatives, co-solvents, solubilizers, suspending agents and emulsifiers. The term "effective amount" or "therapeutically effective amount" refers to an amount of a peptide or composition of the invention that is effective in treating a disease or disorder or ameliorating or enhancing the health condition of a subject. Improving or improving one's health includes improving or improving cognitive functions or abilities. The peptide of the present invention can be expressed as 0.01 mg/kg to 10 mg/kg, 0.02 mg/kg to 8 mg/kg, 0.04 mg/kg to 5 mg/kg, 0.05 mg/kg to 1 mg/kg, or 0.1 mg/kg to 0.8 mg. /kg or 0.2 mg/kg to 0.5 mg/kg is administered to the subject. For example, the peptide of the invention can be administered to a subject in an amount of 0.04 mg/kg, 0.16 mg/kg or 0.64 mg/kg. A therapeutically effective amount may be these dosages. The peptide or composition of the invention may be administered at once daily intervals for at least 1 week, for example 1 to 4 weeks. The peptides of the present invention may be administered to a subject as a combination of peptides or prepared into a composition. Compositions of the invention may comprise such combinations of peptides. For example, the peptide combination includes a first peptide having the amino acid sequence of SEQ ID NO: 1 or a variant thereof and a second peptide having the amino acid sequence of SEQ ID NO: 2 or a variant thereof.

除非另有說明,否則本文使用的所有科學術語都和本領域一般技術人員通常理解的含義一致。下文描述了示例方法和材料,可以使用其等同物。本文提及的所有出版物和其它參考文獻都通過引用整體結合到本文中。 提供以下的實施例是為了進一步闡述本發明。以下實施例無意以任何理由限制本發明的範圍。 實施例 實施例 1 – 本發明肽對阿爾茨海默病模式動物 APP/PS1 小鼠神經保護作用的研究PKT101肽:DEAQETAVSSHEQD(SEQ ID NO: 2) PKT002肽:DEAQETAVSSH(SEQ ID NO: 1) 可通過WO2013/173941和WO2016/165102中描述的方法獲得上述肽。 1. 材料和方法5月齡的APP/PS1雄性小鼠60隻,隨機分為4組,每組15隻,分別每日腹腔注射生理鹽水、PKT101(8mg/kg)、PKT002(8mg/kg)和PKT101/PKT002(8mg/kg)聯合給藥,為期4周。每組腹腔注射終體積為80μL,每週給藥結束後,隨機取出20-35隻小鼠測量體重,體重平均數作為下周給藥劑量的重要依據。 對小鼠進行短期學習記憶(Y迷宮和新物體識別)、條件性恐懼記憶和社交行為學檢測,並通過硫磺素染色和6E10免疫螢光染色檢測大腦皮層和海馬斑塊沉積情況;通過免疫螢光染色觀察皮質和海馬腦區小膠質細胞和星形膠質細胞的活化情況;酶聯免疫吸附實驗(ELISA)檢測炎症因數白介素- 1β(IL-1β)、白介素- 6(IL-6)和腫瘤壞死因數(TNF-α)的含量。 1.1. 行為學檢測 1.1.1. 短期學習記憶 1.1.1.1. Y 迷宮本實驗所使用的Y迷宮裝置,三個臂之間夾角為120度,相互聯通,三個臂尺寸相同,均為長寬高29cm × 8cm × 15cm,在臂內側貼上不同圖案為小鼠作為視覺標記,分別命名為起始臂,新奇臂和其他臂。實驗流程分為兩部分,適應階段和測試階段,兩次間隔1-2h。適應是將新奇臂用擋板隔開,小鼠只能在起始臂和其他臂內自由活動,每隻小鼠適應時間為5min,測試階段是在1-2h後,將新奇臂打開,小鼠可以在三個臂內自由活動,每隻小鼠測試時間為5min。實驗過程中用Topscan軟體即時記錄小鼠進入新奇臂的時間與次數。每次實驗結束後都需用75%酒精清理實驗器具,清除小鼠氣味。 1.1.1.2. 新物體識別本裝置在進行測試前,要對小鼠消除陌生感,每天撫摸小鼠,以免操作時對小鼠產生刺激。第1階段(熟悉期),在裝置中放入2個相同的物體(AB,確保物體沒有氣味,不被推動),物體距離兩側壁10cm,將小鼠從兩物體中間放入裝置中,用攝像頭及軟體來記錄小鼠在每個物體上的探索時間(以嘴或者鼻子接觸到物體和湊近物體約2-3 cm範圍都算對物體的探索),在5min內(許多實驗已經證實熟悉期2min時,動物對新奇事物已經有偏好,而多熟悉3 min則偏好更加明顯)測定動物探索每個物體的次數、時間和距離。第2階段(測試期),第1階段完成後的1h作為檢測記憶的時間間隔,將兩個相同物體中的一個物體替換成一個不同的物體放入裝置中(AC或BC),同樣將小鼠從兩個物體中間放入裝置中,記錄5min內小鼠對新舊物體探索的次數、時間和距離,即小鼠在新舊物體周圍活動的次數、時間和距離,檢測小鼠的認知情況。每次實驗結束後都需用75%酒精清理實驗器具。若小鼠認知能力差,則在新舊物體的探索無差異;若小鼠認知能力正常,則對新事物的探索時間較舊事物長。認知指數(recognition index,RI)計算公式為:RI = 新物體 /(新物體 + 舊物體)×100 %。 1.1.2. 條件性恐懼記憶本實驗第1天將小鼠放入箱內(箱子底部為銅條柵欄,可通電),適應3min後,讓小鼠在箱內繼續停留6min,在此期間,每2min同時給予一次單一頻率聲音刺激(1.0 KHZ,70db,30s)以及不可逃避足底電擊(0.8 mA,2s),共三次,記錄小鼠6min內聲音和電擊誘發僵直行為的總時間,隨後放回飼養籠。每次實驗結束後用75%酒精擦拭箱底。24h後將已建立條件恐懼的小鼠放入原來的箱內,立即給予相同強度的聲音刺激(1.0KHZ,70db,30s),記錄小鼠3min內聲音誘發僵直行為。僵直行為定義為除呼吸外無其他運動行為。記錄環境和聲音誘發僵直時間百分比。 1.1.3. 社交行為 1.1.3.1. 社交偏好階段實驗開始前,將小鼠放在行為測試室適應0.5h,用所測試的長方形箱子等分為Empty、Center和Stranger-1三個區域。將同性別同背景相同月齡的小鼠放進Stranger-1區域裡的金屬籠子裡,另外一側箱子的金屬籠子空著。將測試小鼠從中間(Center)的箱子裡放入,使測試小鼠可以在三個箱子中自由活動5min。拍攝並記錄相關參數:進入每個箱子持續的時間,當小鼠的頭和四爪都進入一個箱子就認為它處在那個箱子中。 1.1.3.2. 社交記憶階段在社交記憶階段實驗中空著的金屬籠子中放入同性別同背景不同月齡的小鼠(Stranger-2),然後記錄5min,觀察測試小鼠在Stranger-1和Stranger-2區域活動的時間。 1.2. 腦組織取材和切片製備 1.2.1. 取材在小鼠行為學檢測結束後及時取材。麻醉後經眼球取血。用生理鹽水由心臟左心室灌流,灌注至肝臟呈土黃色,更換4%多聚甲醛溶液灌注5min,取腦置入4%多聚甲醛,4℃固定過夜。切取海馬和皮層部分的腦組織,進行蔗糖梯度脫水。 1.2.2. 冰凍切片製備應用萊卡冰凍切片機對OCT樹脂包埋的腦組織,進行冠狀切片,片厚10 μm,收集於PBS中用於免疫螢光染色和Aβ病理組織學分析。 1.3. 免疫螢光染色切片用10%胎牛血清PBS溶液室溫下封閉1h。棄去封閉液,加入鼠源性6E10抗體、雞源性膠質纖維酸性蛋白抗體和兔源性鈣離子結合受體分子 1(inized calcium binding adapter molecule 1,Iba1)抗體,4℃孵育過夜。次日洗去一抗,PBS洗3遍,每次5min。加入相應種屬的二抗(山羊抗雞IgG-488; 驢抗兔IgG-488;驢抗鼠IgG-647),室溫避光孵育1h。棄去二抗PBS洗3次,每次5min,DAPI染色室溫10min,棄去DAPI,PBS洗3遍。抗螢光淬滅劑封片,螢光顯微鏡下攝片。 1.4. 硫磺素 S 染色硫磺素S可以標記腦片上的Aβ核心斑塊,脫蠟水化後的組織切片即可染色,採用1%硫磺素染色5min,流水沖洗1min,70%酒精分色30s,再用抗螢光淬滅劑封片,螢光顯微鏡下攝片。 1.5. 圖像分析採用Leica顯微鏡拍攝免疫螢光染色的組織切片,應用ImageJ軟體採用灰度閾值分析方法,統計GFAP、Iba1、6E10、硫磺素的陽性面積百分比,每組5隻小鼠,每隻小鼠選擇3張切片,取其平均值作為該組的統計結果。 1.6. 酶聯免疫吸附實驗 (ELISA) 1.6.1. 取材小鼠麻醉後,斷頭處死,剝離頸部和頭部皮膚,暴露顱骨,沿矢狀縫剪開顱骨,分離顱骨後取出腦組織。將皮層組織和海馬組織分別放入標記好的離心管中,迅速在液氮中冷凍,轉移到-80℃冰箱保存。 1.6.2. 樣品準備冰盒上取皮層和海馬組織10-20mg左右,放入1.5mL離心管中稱量後按品質體積比1:10加入RIPA裂解液,剩餘組織放回原來的管子,用液氮速凍。同時離心管中加入兩枚鋼珠,放入勻漿機配平後勻漿裂解,然後冰上搖晃30 min。取出鋼珠,12000rpm,4℃,15min離心,充分吸取上清,轉移至新的1.5mL離心管中。採用蛋白定量法定量蛋白濃度。 1.6.3. ELISA樣本加入稀釋液稀釋到適當濃度,將稀釋好的樣品加入酶標反應孔,用於測試白介素-1β(IL-1β)、白介素-6(IL-6)和腫瘤壞死因數-α(TNF-α)3種促炎指標,每樣品至少加雙孔,標準品和樣本每孔45μL,隨後在樣本和標準品孔中加入50μL生物素化抗體工作液,用封板紙封住反應孔板,置室溫孵育2h(使用微量振盪器,頻率,300rpm)。提前30min製備酶結合底物,室溫避光放置。棄去孔中液體後在吸水紙上拍乾,洗滌次數5次,洗滌方法:吸乾孔內反應液,將洗滌液加入孔板,放置2min略作搖動。除空白孔外的每孔加100μL酶結合底物工作液,置室溫孵育1h(使用微量振盪器,頻率,300rpm)。棄去孔中液體後在吸水紙上拍乾,洗滌次數5次,除空白孔外的每孔加100 μL顯色底物工作液,室溫避光放置15min。除空白孔外的每孔加入終止液100μL終止反應,於20min內測定實驗結果。450 nm波長下檢測,標準孔和測試孔的吸收值。根據標準孔和已知濃度繪製標準曲線,後由標準曲線換算出測試孔的的實際濃度,最後根據蛋白濃度得出每毫克腦組織中含有的目標蛋白質量。 2. PKT101 PKT002 PKT101/PKT002 聯合給藥後對 APP/PS1 小鼠認知行為的影響應用Y迷宮實驗評價各組小鼠的短期記憶,結果顯示:較對照組APP/PS1小鼠相比,PKT101治療組APP/PS1小鼠進入新奇臂的時間有所增加,而PKT002和PKT101/ PKT002聯合治療組APP/PS1小鼠進入新奇臂的時間顯著性增加(圖1A)。應用新物體識別實驗評價各組小鼠的短期記憶,結果顯示:較對照組相比,上述三種治療組APP/PS1小鼠識別新物體的指數顯著性增加(圖1B)。應用條件恐懼記憶評價各組小鼠的恐懼記憶,結果顯示:PKT101、PKT002和PKT101/ PKT002聯合治療組APP/PS1小鼠恐懼時間百分比均未出現明顯的變化,但相對於對照組有所減少(圖1C);應用三箱實驗評價各組小鼠的社交能力,結果顯示:在社會偏愛階段,PKT101表現出對同性別小鼠(Stranger1)偏愛(圖1D左),提示社會學習能力有所提高;在社會記憶階段,PKT101組中對Stranger1仍有一定偏愛(圖1D右)。實驗結果還見下表2。 3. PKT101 PKT002 PKT101/PKT002 聯合給藥後對 APP/PS1 小鼠 沉積的影響應用硫磺素染色和6E10免疫螢光染色,分別評價各組小鼠皮層和海馬纖維性和彌散性Aβ斑塊沉積情況,硫磺素染色結果顯示:與對照組相比,PKT101、PKT002和PKT101/PKT002聯合治療組APP/PS1小鼠海馬Aβ斑塊均顯著減少,另對皮層中沉積Aβ斑塊沒有影響(圖2A,C)。6E10染色結果顯示,PKT101、PKT002和PKT101/PKT002聯合給藥後對APP/PS1小鼠海馬Aβ沉積顯著性降低,而對皮層中Aβ沉積沒有影響(圖2B,D)。實驗結果還見表3。 4. PKT101 PKT002 PKT101/PKT002 聯合給藥後改善 APP/PS1 小鼠海馬 斑塊周圍膠質細胞活化通過6E10、GFAP和Iba1共標染色分別評價APP/PS1小鼠海馬和皮層Aβ斑塊周圍小膠質細胞和星形膠質細胞活化情況,結果顯示:與對照組相比,PKT101、PKT002和PKT101/PKT002聯合治療組小鼠海馬小膠質細胞活化程度均明顯降低(圖3A-B)。PKT002和PKT101/PKT002聯合給藥治療組顯著降低小鼠海馬星形膠質細胞活化(圖3A,C)。與對照組相比,PKT101、PKT002和PKT101/PKT002聯合給藥治療組小鼠大腦皮層膠質細胞活化均未見明顯改善(圖4A-B)。實驗結果還見表4。 5. PKT101 PKT002 PKT101/PKT002 聯合給藥後對 APP/PS1 小鼠海馬和大腦皮層 IL-1β IL-6 TNF-α 含量的影響應用ELISA檢測各組小鼠海馬和大腦皮層炎症因數IL-1β、IL-6和TNF-α的含量,結果顯示:與對照組相比,PKT101、PKT002和PKT101/PKT002聯合治療小鼠海馬炎症因數IL-6和TNF-α的含量顯著性降低,PKT002和PKT101/PKT002聯合治療組IL-1β的含量顯著性降低(圖5A),而上述三組小鼠大腦皮層IL-6的表達顯著性降低,其他炎症因數IL-1β和TNF-α表達均未見顯著性差異(圖5B)。實驗結果還見表5。 6. 結論PKT101和PKT002單獨治療以及PKT101/PKT002聯合治療均能不同程度改善APP/PS1小鼠短期認知障礙、減低海馬Aβ負荷沉積、膠質細胞活化和炎症因數的含量。 實施例 2 – 本發明肽對 MPTP 亞急性帕金森病小鼠模型藥效學實驗研究PKT101肽:DEAQETAVSSHEQD(SEQ ID NO: 2) PKT002肽:DEAQETAVSSH(SEQ ID NO: 1) 可通過WO2013/173941和WO2016/165102中描述的方法獲得上述肽。 1. 材料和方法應用MPTP(1-甲基-4-苯基-1,2,3,6-四氫吡啶)建立亞急性帕金森病小鼠模型。小鼠稱重後記錄,按照20mg/kg頸後部皮下注射MPTP,每天一次,連續注射5天,最後一次給藥後的第1、3、7和14天時進行後續實驗。 將168隻健健康成年C57BL/6J雄性小鼠,按體重隨機分為八組,為正常對照(Sham)組(n=20)、MPTP模型組(n=28)、PKT101低劑量(0.5mg/kg/d)治療組(n=20)、PKT101中劑量(2mg/kg/d)治療組(n=20)、PKT101高劑量(8mg/kg/d)治療組(n=20)、PKT002(8mg/kg/d)治療組(n=20)、PKT101(8mg/kg/d)+PKT002(8mg/kg/d)合併治療組(n=20)和陽性對照藥司來吉蘭(Selegiline,咪多吡)(0.5 mg/kg/d)治療組(n=20)。從第一次MPTP給藥後1小時開始腹腔注射相應藥物進行治療,持續7或14天,每天一次。MPTP模型組小鼠給予等體積生理鹽水(0.1mL/10g)。Sham組小鼠僅給予等體積生理鹽水(0.1mL/10g)。 在MPTP最後一次給藥後的第1、3、7和14天時進行曠場、爬杆和轉棒實驗,評價小鼠的行為學改變。在MPTP最後一次給藥後的第7和14天時對中腦進行尼氏和TH染色,計數黑質緻密部尼氏和TH陽性細胞數量,評價相應腦區多巴胺能神經元損傷程度。 2. 行為學評價分別在MPTP最後一次給藥後的第1、3、7和14天時進行行為學評價。 2.1. 曠場試驗曠場試驗用於評價小鼠的自主運動能力。開放式曠場裝置由不透明的藍色塑膠製成,大小為60cm×60cm×45 cm。測試時,將每隻小鼠從中心區域放入箱內,同時開始記錄每隻小鼠在5min內的運動軌跡,使用軟體(Clever Sys Inc.,VA,USA)計算總運動路程。每隻小鼠測試結束後,使用75%乙醇擦拭曠場區域,防止上一隻小鼠的氣味對下一隻鼠造成影響。 2.2. 爬杆試驗爬杆試驗用於評估小鼠的運動協調功能。所用杆子直徑lcm,高度50cm,立杆頂端固定有直徑為1.2cm的木球,將立杆用防滑膠帶裹住以防止小鼠滑落。將小鼠頭部向上置於杆頂,小鼠開始向上爬時啟動計時器,記下小鼠從開始運動到越過杆頂所需時間(T-Turn)和從開始運動到爬至底部四爪落地時間(T-TLA)。正式測試前,需對每隻小鼠進行三次連續訓練。測試期間,需連續測量三次,取其中所用最短時間。 2.3. 轉棒測試使用轉棒試驗評估小鼠的運動平衡能力和肢體協調性。將小鼠放於rotarod測試儀滾筒上,將轉速設成2分鐘內從5rpm/min勻速增加到25rpm/min,小鼠從滾筒上掉落至下方感應區域時會有紅外感應器接收信號,記錄小鼠在儀器滾筒上運動的時間。正式測試前,需對每隻小鼠進行三次連續訓練。測試期間,需連續測量三次,取平均掉落時間(潛伏期)。 3. 免疫組化研究分別在MPTP最後一次給藥後的第7和14天取每組一半的小鼠,用4%水合氯醛麻醉、生理鹽水+4%PFA灌注取腦組織。後經由20%(v/v)和30%(v/v)的蔗糖溶液梯度脫水各3天後,將小鼠腦組織用O.C.T.膠包埋後冰凍,冰凍切片機(Leica)連續組織切片(20 μm),收集相關區域腦片置於凍存液(甘油:0.01M PBS = 1:1)中,於-80℃保存待用。 3.1. 尼氏染色尼氏體是胞質內的一種嗜鹼性物質,廣泛見於各種神經元,用於計算腦神經元數量。按照尼氏染色試劑盒進行染色。 3.2. TH(Tyrosine hydroxylase ,酪氨酸羥化酶 ) 染色TH主要標記黑質緻密部多巴胺能神經元,MPTP造模後該腦區TH陽性神經元會顯著減少。取出中腦腦片,0.01M PBS 10min洗片一次後,以3% H 2O 2孵育15min以去除內源性過氧化物酶活性,0.01 M PBS 10min×3洗去H 2O 2,然後用含5% BSA和0.3% Triton X-100的PBS於室溫下封閉1小時,繼而孵育小鼠抗TH一抗(T2928,Sigma,USA)4℃過夜,二抗室溫孵育1小時,PBS洗滌5min×3次後二氨基聯苯氨(diaminobenzidin,DAB)避光顯色。 4. 細胞計數和資料處理及分析免疫組織化學陽性細胞計數採用體式學計數系統(Stereo Investigator,MBF bioscience)計算黑質緻密部的尼氏小體陽性細胞和TH +細胞數,細胞計數在×20物鏡下進行。 所有資料均採用Mean ± S.E.M的表示,採用GraphPad Prism 8.0統計分析軟體進行統計學處理。計量資料採用One-way ANOVA合併Dunnett’s test 分析各組與造模組之間的差異, p< 0.05表示差異具有統計學意義。 5. 實驗結果 5.1. PKT101 PKT002 對小鼠 MPTP 亞急性模型後生存率的影響MPTP亞急性給藥使得小鼠在造模後精神萎靡、厭食、體重下降,並出現豎尾反應,但本次實驗中未出現小鼠的死亡,藥物治療各組小鼠也未出現死亡現象。 5.2. PKT101 PKT002 對小鼠 MPTP 亞急性模型後體重改變的影響MPTP亞急性給藥使得小鼠在造模後進食量小,7天內小鼠體重略降低,14天後進食逐漸增加,體重逐漸恢復。各組處理對小鼠體重無顯著影響。結果如圖6所示。 5.3. PKT101 PKT002 對小鼠 MPTP 亞急性模型後在礦場實驗中自主運動能力的影響如表6和圖7所示,MPTP亞急性模型中,小鼠在造模後1、3、7和14天時出現曠場試驗中總運動路程的減少,表明自主活動能力的下降。陽性對照藥司來吉蘭(Selegiline)在1、3、7和14天均能顯著改善MPTP誘導的總運動路程減低。PKT101和PKT002在14天內也均有顯著的改善作用。在14天時,單PKT101給藥低、中和高劑量增加為模型組的221.1%、253.9%和255.3%;單PKT002給藥增加為模型組的213.0%;PKT101和PKT002合併給藥增加為模型組的214.8%。 5.4. PKT101 PKT002 對小鼠 MPTP 亞急性模型後在爬杆實驗中運動協調能力的影響如表7~8和圖8~9,MPTP亞急性模型中,小鼠在造模後1、3、7和14天時出現爬杆實驗中T-Turn和T-TLA時間的延長,表明運動協調能力的下降。陽性對照藥司來吉蘭(Selegiline)在1、3、7和14天均能顯著改善MPTP誘導的T-Turn和T-TLA時間延長。PKT101和PKT002在14天內也均有顯著的改善作用。對T-Turn,在14天時,單PKT101給藥低、中和高劑量分別減少為模型組的26.1%、24.6%和26.0%;單PKT002給藥減少為模型組的24.4%;PKT101和PKT002合併給藥減少為模型組的21.6%。對T-TLA,在14天時,單PKT101給藥低、中和高劑量分別減少為模型組的68.8%、65.1%和38.6%;單PKT002給藥減少為模型組的46.0%;PKT101和PKT002合併給藥減少為模型組的46.3%。 5.5. PKT101 PKT002 對小鼠 MPTP 亞急性模型後在轉棒實驗中運動平衡能力的影響如表9和圖10,MPTP亞急性模型中,小鼠在造模後1、3、7和14天時出現轉棒實驗中掉落潛伏期的縮短,表明運動平衡能力的下降。陽性對照藥司來吉蘭(Selegiline)在1、3、7和14天均能顯著改善MPTP誘導的潛伏期時間縮短。PKT101和PKT00在14天內均有顯著的改善作用。在14天時,單PKT101給藥低、中和高劑量的潛伏期分別延長為模型組的121.7、119.9%和118.3%;單PKT002給藥延長為模型組的120.3%;PKT101和PKT002合併給藥延長為為模型組的120.9%。 5.6. 立再適對小鼠 MPTP 亞急性模型後黑質緻密部尼氏陽性染色神經元數量的影響如表10和圖11,MPTP亞急性模型中,小鼠在造模後7和14天時出現黑質緻密部尼氏染色陽性神經元數量的減少,表明該腦區神經元的死亡。陽性對照藥司來吉蘭(Selegiline)在7和14天均能顯著改善MPTP誘導的黑質緻密部尼氏染色陽性神經元數量的減少。PKT101和PKT002在7和14天時也均有顯著的改善作用。在7天時,單PKT101給藥低、中和高劑量的尼氏陽性細胞數量分別增加為模型組的120.9%、124.0%和130.9%;單PKT002給藥的尼氏陽性細胞數量增加為模型組的124.5%;PKT101和PKT002合併給藥的尼氏陽性細胞數量增加為模型組的128.0%。在14天時,單PKT101給藥低、中和高劑量的尼氏陽性細胞數量分別增加為模型組的124.9%、130.9%和141.1%;單PKT002給藥的尼氏陽性細胞數量增加為模型組的128.5%;PKT101和PKT002合併給藥的尼氏陽性細胞數量增加為模型組的141.5%。 5.7. PKT101 PKT002 對小鼠 MPTP 亞急性模型後黑質緻密部 TH 陽性染色神經元數量的影響如表11和圖12,MPTP亞急性模型中,小鼠在造模後7和14天時出現黑質緻密部TH陽性神經元數量的減少,表明該腦區多巴胺能神經元的死亡。陽性對照藥司來吉蘭(Selegiline)在7和14天均能顯著改善MPTP誘導的黑質緻密部TH染色陽性神經元數量的減少。PKT101和PKT002在7和14天時也均有顯著的改善作用。在7天時,單PKT101給藥低、中和高劑量的TH陽性細胞數量分別增加為模型組的114.5%、124.9%和129.7%;單PKT002給藥的TH陽性細胞數量增加為模型組的129.9%;PKT101和PKT002合併給藥的TH陽性細胞數量增加為模型組的130.3%。在14天時,單PKT101給藥低、中和高劑量的TH陽性細胞數量分別增加為模型組的125.6%、136.0%和137.5%;單PKT002給藥的TH陽性細胞數量增加為模型組的128.1%;PKT101和PKT002合併給藥的TH陽性細胞數量增加為模型組的137.7%。 6. 結論多肽PKT101和PKT002持續7天和14天治療均能顯著改善MPTP亞急性模型後小鼠行為學障礙,可增加曠場試驗中總運動路程,縮短爬杆實驗T-Turn時間和T-TLA時間,延長轉棒實驗潛伏期,但各組間劑量依賴關係不明顯。同時,PKT101和PKT002均能緩解MPTP誘導的黑質緻密部尼氏陽性和TH陽性神經元數量減少。這些結果表明PKT101和PKT002均具有對MPTP亞急性模型的神經保護作用,PKT101和PKT002合併給藥與分別單獨給藥相比,無明顯差別。 實施例 3 – 本發明肽對神經退行性疾病模型中膠質細胞和神經元損傷保護作用的初步研究 1. 實驗目的在細胞水準上,初步探究PKT001、PKT002對膠質細胞和神經元等損傷的保護作用,討論對神經退行性疾病治療的通用機制。 2. 實驗方案小鼠或人的膠質瘤細胞和神經元細胞等,分別按照1、3、9 ug/ml PKT001、PKT002實驗組和對照組進行預處理24h後,再加入脂多糖(LPS,1 μg/ml)誘導48h,進行膜聯蛋白V(Annexin V)/碘化丙啶(PI)雙染色流式分析凋亡細胞比例,檢測PKT001、PKT002給藥對各個神經系細胞凋亡的影響。 3. 實驗材料 細胞株GL261小鼠膠質細胞瘤細胞,培養基:DMEM +10%FBS+ 1%P/S; LN229人膠質瘤細胞,培養基:DMEM+10%FBS+ 1%P/S; WERI-RB-1 人視網膜神經膠質瘤細胞,培養基RPMI-1640+10%FBS+1%P/S; SK-N-SH 人神經母細胞瘤細胞,培養基:MEM(含NEAA)+10%FBS+1%P/S; HT22 小鼠海馬神經元細胞,培養基:DMEM +10%FBS+1%P/S; neuro-2a(N2A)小鼠腦神經瘤細胞,培養基:MEM(含NEAA)+10%FBS+1%P/S; 培養條件:37℃,5% CO 2. 實驗所用的細胞均處於對數生長期。 4. 實驗步驟 PKT001 PKT002 對神經系細胞凋亡的影響(1)細胞傳代鋪板:取正常培養的細胞,吸去原培養液,加入PBS洗後加入胰酶消化1-3min後終止消化,用移液槍吹打成單個細胞,細胞懸液200g離心5min,用培養基重懸,取20μl 細胞懸液加入20μl台盼藍進行計數,3*10 5個/孔(6孔板)進行鋪板,3個複孔,5% CO 2,37℃過夜培養。 (2)加藥處理: ① Cell; ② Cell+LPS(1 μg/ml); ③ Cell+PKT001(1 μg/ml)+LPS(1 μg/ml); ④ Cell+PKT001(3 μg/ml)+LPS(1 μg/ml); ⑤ Cell+PKT001(9 μg/ml)+LPS(1 μg/ml); ⑥ Cell+PKT002(1 μg/ml)+LPS(1 μg/ml); ⑦ Cell+PKT002(3 μg/ml)+LPS(1 μg/ml); ⑧ Cell+PKT002(9 μg/ml)+LPS(1 μg/ml); 共8組,藥物先給藥預處理24h後,再LPS誘導處理48h,隨後進行相關檢測。 (3)凋亡檢測: ① 收集細胞:200g,4℃離心5min收集細胞。 ② 用預冷的PBS洗滌細胞2次,每次200g,4℃離心5min。 ③ 吸棄PBS,加入100μL 1×Binding Buffer重懸細胞。 ④ 加入5μL Annexin FITC和2μL PI,輕輕混勻。 ⑤ 避光、室溫反應15min。 ⑥ 加入300μL 1×Binding Buffer,混勻後放置於冰上,樣品在1小時內用流式細胞儀檢測。 5. 實驗結果 5.1. PKT001 PKT002 LPS 誘導 GL261 小鼠膠質細胞瘤細胞凋亡的影響應用Annexin V-FITC/PI雙染色流式分析法,實驗結果顯示:與LPS對照組相比,1-9μg/ml PKT001預處理後,LPS誘導48h GL261細胞凋亡比例顯著降低;1-9μg/ml PKT002預處理後,LPS誘導48h GL261細胞凋亡比例顯著降低。結果如圖13所示。 5.2. PKT001 PKT002 LPS 誘導 LN229 人膠質瘤細胞 凋亡的影響與LPS對照組相比,1μg/ml PKT001預處理後,LPS誘導48h LN229細胞凋亡比例未顯著性變化,3μg/ml和9μg/ml PKT001預處理後,LPS誘導48h LN229細胞凋亡比例顯著降低。1-9μg/ml PKT002預處理後,LPS誘導48h LN229細胞凋亡比例顯著降低。結果如圖14所示。 5.3. PKT001 PKT002 LPS 誘導 WERI-RB-1 人視網膜神經膠質瘤細胞 凋亡的影響與LPS對照組相比,1-9μg/ml PKT001預處理後,LPS誘導48h WERI-RB-1細胞凋亡比例顯著降低;1-9μg/ml PKT002預處理後,LPS誘導48h WERI-RB-1細胞凋亡比例顯著降低。結果如圖15所示。 5.4. PKT001 PKT002 LPS 誘導 neuro-2a(N2A) 小鼠腦神經瘤細胞凋亡的影響與LPS對照組相比,1μg/ml和3μg/ml PKT001預處理後,LPS誘導48h N2A細胞凋亡比例未顯著性變化,9μg/ml PKT001預處理後,LPS誘導48h N2A細胞凋亡比例降低,有顯著性差異。1-9μg/ml PKT002預處理後,LPS誘導48h N2A細胞凋亡比例顯著降低。結果如圖16所示。 5.5. PKT001 PKT002 LPS 誘導 HT22 小鼠海馬神經元細胞凋亡的影響實驗結果顯示:與LPS對照組相比,1-9μg/ml PKT001預處理後,LPS誘導48h HT22細胞凋亡比例顯著降低;1-9μg/ml PKT002預處理後,LPS誘導48h HT22細胞凋亡比例顯著降低。結果如圖17所示。 5.6. PKT001 PKT002 LPS 誘導 SK-N-SH 人神經母細胞瘤細胞 凋亡的影響與LPS對照組相比,1μg/ml PKT001預處理後,LPS誘導48h SK-N-SH細胞凋亡比例未顯著性變化,3μg/ml和9μg/ml PKT001預處理後,LPS誘導48h SK-N-SH細胞凋亡比例顯著降低。1-9μg/ml PKT002預處理後,LPS誘導48h SK-N-SH細胞凋亡比例顯著降低。結果如圖18所示。 Unless otherwise defined, all scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art. Example methods and materials are described below, equivalents thereof may be used. All publications and other references mentioned herein are incorporated by reference in their entirety. The following examples are provided to further illustrate the invention. The following examples are not intended to limit the scope of the invention for any reason. Examples Example 1 - Study on the neuroprotective effect of the peptide of the present invention on Alzheimer's disease model animal APP/PS1 mice PKT101 peptide: DEAQETAVSSHEQD (SEQ ID NO: 2) PKT002 peptide: DEAQETAVSSH (SEQ ID NO: 1) Yes The above peptides were obtained by the methods described in WO2013/173941 and WO2016/165102. 1. Materials and Methods 60 5-month-old APP/PS1 male mice were randomly divided into 4 groups, 15 mice in each group, and were intraperitoneally injected with normal saline, PKT101 (8mg/kg), and PKT002 (8mg/kg) every day. Combined administration with PKT101/PKT002 (8mg/kg) for 4 weeks. The final volume of intraperitoneal injection in each group was 80 μL. After weekly administration, 20-35 mice were randomly taken out and measured for weight measurement. The average weight was used as an important basis for the next week's dosage. The mice were tested for short-term learning and memory (Y maze and new object recognition), conditioned fear memory and social behavior, and the plaque deposition in the cerebral cortex and hippocampus was detected through thioflavin staining and 6E10 immunofluorescence staining; Light staining was used to observe the activation of microglia and astrocytes in the cortex and hippocampus; enzyme-linked immunosorbent assay (ELISA) was used to detect the inflammatory factors interleukin-1β (IL-1β), interleukin-6 (IL-6) and tumors. Necrosis factor (TNF-α) content. 1.1. Behavioral testing 1.1.1. Short-term learning and memory 1.1.1.1. Y maze The Y maze device used in this experiment has a 120-degree angle between the three arms, which are connected to each other. The three arms have the same size, both length and width. It is 29cm × 8cm × 15cm high. Different patterns are affixed to the inside of the arms for mice as visual markers, which are named starting arms, novel arms and other arms. The experimental process is divided into two parts, the adaptation phase and the testing phase, with an interval of 1-2 hours. Adaptation is to separate the novel arms with baffles. Mice can only move freely in the starting arm and other arms. The adaptation time of each mouse is 5 minutes. The test phase is to open the novel arms after 1-2 hours. The mice can move freely within the three arms, and the test time for each mouse is 5 minutes. During the experiment, Topscan software was used to instantly record the time and number of times the mice entered the novel arm. After each experiment, 75% alcohol must be used to clean the experimental equipment to remove the mouse odor. 1.1.1.2. New object recognition Before testing this device, the mice must be eliminated from any sense of strangeness and the mice must be stroked every day to avoid irritation to the mice during operation. In the first stage (familiarization period), put two identical objects (AB, make sure the objects have no smell and are not pushed) into the device. The objects are 10cm away from both sides of the walls. Put the mouse into the device from the middle of the two objects. Cameras and software are used to record the mouse's exploration time on each object (touching the object with the mouth or nose and approaching the object within about 2-3 cm are considered exploration of the object), within 5 minutes (many experiments have confirmed that the familiarization period At 2 minutes, the animal already has a preference for novel objects, and after 3 minutes of familiarity, the preference becomes more obvious.) The number, time and distance of the animal exploring each object were measured. In the second stage (test period), 1 hour after the completion of the first stage is used as the time interval to detect memory. One of the two identical objects is replaced with a different object and placed in the device (AC or BC). The small object is also placed in the device (AC or BC). The mouse is put into the device from the middle of two objects, and the number, time and distance of the mouse's exploration of the old and new objects within 5 minutes are recorded, that is, the number, time and distance of the mouse's activities around the new and old objects, and the cognitive status of the mouse is detected. . After each experiment, the experimental equipment needs to be cleaned with 75% alcohol. If the mouse has poor cognitive ability, there will be no difference in the exploration of new and old objects; if the mouse has normal cognitive ability, the exploration time of new objects will be longer than that of old objects. The calculation formula of recognition index (recognition index, RI) is: RI = new object/(new object + old object) × 100%. 1.1.2. Conditioned fear memory. On the first day of this experiment, the mice were put into the box (the bottom of the box is a copper fence that can be powered). After adapting for 3 minutes, the mice were allowed to stay in the box for another 6 minutes. During this period, A single-frequency sound stimulus (1.0 KHZ, 70db, 30s) and an inescapable footshock (0.8 mA, 2s) were given simultaneously every 2min for a total of three times. The total time of sound- and electricshock-induced catalepsy behavior in the mice within 6min was recorded, and then released. Return to the breeding cage. After each experiment, wipe the bottom of the box with 75% alcohol. After 24 hours, the mice with established fear were placed in the original box, and the same intensity of sound stimulation (1.0KHZ, 70db, 30s) was immediately given, and the sound-induced freezing behavior of the mice was recorded within 3 minutes. Catalepsy was defined as no movement other than breathing. Percentage of environmentally and sound-induced freezing time was recorded. 1.1.3. Social behavior 1.1.3.1. Social preference stage Before the start of the experiment, the mice were placed in the behavioral testing room to adapt for 0.5 hours. The rectangular box under test was equally divided into three areas: Empty, Center and Stranger-1. Mice of the same sex, background, and age were placed into the metal cage in the Stranger-1 area, and the metal cage on the other side of the box was empty. Put the test mouse into the center box so that the test mouse can move freely in the three boxes for 5 minutes. Photograph and record relevant parameters: the duration of entering each box. When the mouse's head and four paws enter a box, it is considered to be in that box. 1.1.3.2. Social memory stage In the social memory stage of the experiment, mice of the same sex and background and different ages (Stranger-2) were placed in an empty metal cage, and then recorded for 5 minutes. Observe the test mice’s performance in Stranger-1 and Stranger. -2 hours of regional activities. 1.2. Brain tissue collection and section preparation 1.2.1. Collection of materials Timely after the completion of the mouse behavioral test. After anesthesia, blood was collected through the eyeball. Perfuse with physiological saline from the left ventricle of the heart until the liver turns khaki, replace with 4% paraformaldehyde solution and perfuse for 5 minutes. Remove the brain and place it in 4% paraformaldehyde, and fix it overnight at 4°C. Brain tissues from the hippocampus and cortex were excised and subjected to sucrose gradient dehydration. 1.2.2. Preparation of frozen sections . The OCT resin-embedded brain tissue was sliced coronally using a Leica frozen microtome, with a thickness of 10 μm, and collected in PBS for immunofluorescence staining and Aβ histopathological analysis. 1.3. Immunofluorescence stained sections were blocked with 10% fetal calf serum PBS solution for 1 hour at room temperature. Discard the blocking solution, add mouse-derived 6E10 antibody, chicken-derived glial fibrillary acidic protein antibody, and rabbit-derived calcium binding adapter molecule 1 (inized calcium binding adapter molecule 1, Iba1) antibody, and incubate overnight at 4°C. The next day, the primary antibody was washed away and washed 3 times with PBS for 5 minutes each time. Add secondary antibodies of corresponding species (goat anti-chicken IgG-488; donkey anti-rabbit IgG-488; donkey anti-mouse IgG-647) and incubate at room temperature in the dark for 1 hour. Discard the secondary antibody, wash 3 times with PBS, 5 minutes each time, stain with DAPI for 10 minutes at room temperature, discard DAPI, and wash 3 times with PBS. The slides were sealed with anti-fluorescence quenching agent and photographed under a fluorescence microscope. 1.4. Thioflavin S staining Thioflavin S can mark Aβ core plaques on brain slices. The tissue sections can be stained after dewaxing and hydration. Use 1% thioflavin S for 5 minutes, rinse with running water for 1 minute, and separate with 70% alcohol for 30 seconds. The slides were then sealed with anti-fluorescence quenching agent and photographed under a fluorescence microscope. 1.5. Image analysis: Use a Leica microscope to capture immunofluorescence-stained tissue sections, and use ImageJ software to use grayscale threshold analysis to count the percentage of positive areas for GFAP, Iba1, 6E10, and thioflavin. There are 5 mice in each group, and each The mouse selects 3 slices, and the average value is used as the statistical result of the group. 1.6. Enzyme-linked immunosorbent assay (ELISA) 1.6.1. Take the mice. After anesthesia, decapitate the mice, peel off the skin of the neck and head, expose the skull, cut the skull along the sagittal suture, separate the skull and remove the brain tissue. Put the cortical tissue and hippocampal tissue into marked centrifuge tubes, quickly freeze them in liquid nitrogen, and transfer them to a -80°C refrigerator for storage. 1.6.2. Sample preparation : Take about 10-20 mg of cortex and hippocampus tissue on the ice box, weigh it into a 1.5 mL centrifuge tube, and add RIPA lysis solution at a mass-to-volume ratio of 1:10. Put the remaining tissue back into the original tube, and use Liquid nitrogen quick freezing. At the same time, add two steel balls to the centrifuge tube, put it into the homogenizer to balance, homogenize and lyse, and then shake on ice for 30 minutes. Remove the steel beads, centrifuge at 12000 rpm, 4°C for 15 min, fully absorb the supernatant, and transfer to a new 1.5 mL centrifuge tube. Protein concentration was quantified using protein quantification method. 1.6.3. Add diluent to the ELISA sample to dilute it to an appropriate concentration, and add the diluted sample to the enzyme-labeled reaction well for testing interleukin-1β (IL-1β), interleukin-6 (IL-6) and tumor necrosis factor- α (TNF-α) 3 pro-inflammatory indicators, add at least two wells for each sample, 45 μL of standard and sample in each well, then add 50 μL of biotinylated antibody working solution to the sample and standard wells, and seal the reaction with sealing paper Well plate, incubate at room temperature for 2 hours (use micro shaker, frequency, 300 rpm). Prepare the enzyme-binding substrate 30 minutes in advance and store it at room temperature away from light. Discard the liquid in the wells and pat dry on absorbent paper. Wash 5 times. Washing method: absorb the reaction solution in the wells, add the washing solution to the well plate, and leave it for 2 minutes with slight shaking. Add 100 μL of enzyme-bound substrate working solution to each well except the blank well, and incubate at room temperature for 1 hour (use a micro shaker, frequency, 300 rpm). Discard the liquid in the wells and pat dry on absorbent paper. Wash 5 times. Add 100 μL of chromogenic substrate working solution to each well except the blank wells, and place at room temperature in the dark for 15 minutes. Add 100 μL of stop solution to each well except the blank well to stop the reaction, and measure the experimental results within 20 minutes. Detection at 450 nm wavelength, absorbance values of standard holes and test holes. A standard curve is drawn based on the standard wells and known concentrations, and then the actual concentration of the test wells is converted from the standard curve. Finally, the target protein amount per milligram of brain tissue is obtained based on the protein concentration. 2. Effects of PKT101 , PKT002 and PKT101/PKT002 combined administration on the cognitive behavior of APP/PS1 mice. The Y maze test was used to evaluate the short-term memory of mice in each group. The results showed that compared with the control group of APP/PS1 mice, The time for APP/PS1 mice in the PKT101 treatment group to enter the novel arm increased, while the time for APP/PS1 mice in the PKT002 and PKT101/PKT002 combined treatment groups to enter the novel arm increased significantly (Figure 1A). The new object recognition test was used to evaluate the short-term memory of mice in each group. The results showed that compared with the control group, the index of APP/PS1 mice in the above three treatment groups to recognize new objects increased significantly (Figure 1B). Conditioned fear memory was used to evaluate the fear memory of mice in each group. The results showed that the percentage of fear time of APP/PS1 mice in the PKT101, PKT002 and PKT101/PKT002 combined treatment groups did not change significantly, but it was reduced compared to the control group ( Figure 1C); the three-box experiment was used to evaluate the social ability of mice in each group. The results showed that during the social preference stage, PKT101 showed a preference for same-sex mice (Stranger1) (Figure 1D left), suggesting that social learning ability has been improved. ; In the social memory stage, there is still a certain preference for Stranger1 in the PKT101 group (Figure 1D right). The experimental results are also shown in Table 2 below. 3. Effects of PKT101 , PKT002 and PKT101/PKT002 combined administration on deposition in APP/PS1 mice . Thioflavin staining and 6E10 immunofluorescence staining were used to evaluate fibrous and diffuse Aβ plaques in the cortex and hippocampus of mice in each group. Thioflavin staining results showed that compared with the control group, Aβ plaques in the hippocampus of APP/PS1 mice in the PKT101, PKT002 and PKT101/PKT002 combined treatment groups were significantly reduced, and had no effect on Aβ plaque deposition in the cortex ( Figure 2A,C). The 6E10 staining results showed that the combined administration of PKT101, PKT002 and PKT101/PKT002 significantly reduced Aβ deposition in the hippocampus of APP/PS1 mice, but had no effect on Aβ deposition in the cortex (Figure 2B, D). The experimental results are also shown in Table 3. 4. Combined administration of PKT101 , PKT002 and PKT101/PKT002 improves glial cell activation around plaques in the hippocampus of APP /PS1 mice . The co-labeled staining of 6E10, GFAP and Iba1 was used to evaluate the activation of glial cells around the Aβ plaques in the hippocampus and cortex of APP/PS1 mice, respectively. Activation of microglia and astrocytes, the results showed that compared with the control group, the degree of microglial activation in the hippocampus of mice in the PKT101, PKT002 and PKT101/PKT002 combined treatment groups was significantly reduced (Figure 3A-B). The combined administration of PKT002 and PKT101/PKT002 significantly reduced astrocyte activation in the mouse hippocampus (Figure 3A, C). Compared with the control group, the activation of glial cells in the cerebral cortex of mice in the PKT101, PKT002 and PKT101/PKT002 combined treatment groups showed no significant improvement (Figure 4A-B). The experimental results are also shown in Table 4. 5. Effects of PKT101 , PKT002 and PKT101/PKT002 combined administration on IL-1β , IL-6 and TNF-α contents in the hippocampus and cerebral cortex of APP/PS1 mice. ELISA was used to detect inflammatory factors in the hippocampus and cerebral cortex of mice in each group. The results showed that compared with the control group, the levels of inflammatory factors IL-6 and TNF-α in the hippocampus of mice treated with PKT101, PKT002 and PKT101/PKT002 combination were significantly reduced. The content of IL-1β in the PKT002 and PKT101/PKT002 combined treatment groups was significantly reduced (Figure 5A), while the expression of IL-6 in the cerebral cortex of the above three groups of mice was significantly reduced, and the expression of other inflammatory factors IL-1β and TNF-α was significantly reduced. No significant difference was seen (Fig. 5B). The experimental results are also shown in Table 5. 6. Conclusion PKT101 and PKT002 alone and PKT101/PKT002 combined treatment can improve short-term cognitive impairment, reduce hippocampal Aβ load deposition, glial cell activation and inflammatory factors in APP/PS1 mice to varying degrees. Example 2 - Experimental study on the pharmacodynamics of the peptide of the present invention on MPTP subacute Parkinson's disease mouse model PKT101 peptide: DEAQETAVSSHEQD (SEQ ID NO: 2) PKT002 peptide: DEAQETAVSSH (SEQ ID NO: 1) Can be obtained through WO2013/173941 and The above peptide was obtained by the method described in WO2016/165102. 1. Materials and methods MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) was used to establish a subacute Parkinson's disease mouse model. After the mice were weighed and recorded, 20 mg/kg MPTP was injected subcutaneously into the back of the neck once a day for 5 consecutive days. Follow-up experiments were conducted on days 1, 3, 7 and 14 after the last administration. 168 healthy adult C57BL/6J male mice were randomly divided into eight groups according to body weight: normal control (Sham) group (n=20), MPTP model group (n=28), PKT101 low-dose (0.5mg/ kg/d) treatment group (n=20), PKT101 medium-dose (2mg/kg/d) treatment group (n=20), PKT101 high-dose (8mg/kg/d) treatment group (n=20), PKT002 ( 8mg/kg/d) treatment group (n=20), PKT101 (8mg/kg/d)+PKT002 (8mg/kg/d) combined treatment group (n=20) and the positive control drug Selegiline, Imidopyr) (0.5 mg/kg/d) treatment group (n=20). Treatment began with intraperitoneal injection of the corresponding drug 1 hour after the first MPTP administration and continued once a day for 7 or 14 days. Mice in the MPTP model group were given an equal volume of physiological saline (0.1mL/10g). The mice in the Sham group were only given an equal volume of physiological saline (0.1mL/10g). Open field, rod climbing and rotarod experiments were conducted on days 1, 3, 7 and 14 after the last administration of MPTP to evaluate the behavioral changes of mice. On days 7 and 14 after the last administration of MPTP, Nissl and TH staining was performed on the midbrain, the number of Nissl and TH positive cells in the substantia nigra pars compacta was counted, and the degree of damage to dopaminergic neurons in the corresponding brain areas was evaluated. 2. Behavioral evaluation: Conduct behavioral evaluation on days 1, 3, 7 and 14 after the last administration of MPTP. 2.1. Open field test The open field test is used to evaluate the voluntary movement ability of mice. The open field device is made of opaque blue plastic and measures 60cm × 60cm × 45 cm. During the test, each mouse was placed into the box from the central area, and at the same time, the movement trajectory of each mouse within 5 minutes was recorded, and the total movement distance was calculated using software (Clever Sys Inc., VA, USA). After each mouse test, use 75% ethanol to wipe the open area to prevent the smell of the previous mouse from affecting the next mouse. 2.2. Pole climbing test The pole climbing test is used to evaluate the motor coordination function of mice. The pole used has a diameter of 1cm and a height of 50cm. A wooden ball with a diameter of 1.2cm is fixed on the top of the pole. The pole is wrapped with anti-slip tape to prevent mice from slipping. Place the mouse's head upward on the top of the pole. When the mouse starts to climb up, start the timer. Record the time it takes for the mouse to move over the top of the pole (T-Turn) and the time it takes for the mouse to climb to the bottom of the four paws. Time to Landing (T-TLA). Before formal testing, each mouse needs to be trained three times consecutively. During the test, three consecutive measurements are required, and the shortest time is chosen. 2.3. Rotarod test Use the rotarod test to evaluate the motor balance ability and limb coordination of mice. Place the mouse on the drum of the rotarod tester, and set the rotation speed to increase from 5rpm/min to 25rpm/min at a constant speed within 2 minutes. When the mouse falls from the drum to the sensing area below, an infrared sensor will receive the signal and record it. Time for mice to move on the instrument roller. Before formal testing, each mouse needs to be trained three times consecutively. During the test, three consecutive measurements are required to calculate the average drop time (latency period). 3. Immunohistochemistry study: On days 7 and 14 after the last administration of MPTP, half of the mice from each group were taken, anesthetized with 4% chloral hydrate, and perfused with normal saline + 4% PFA to collect brain tissue. After gradient dehydration with 20% (v/v) and 30% (v/v) sucrose solution for 3 days each, the mouse brain tissue was embedded in OCT gel and frozen, and serial tissue sections were made using a freezing microtome (Leica) ( 20 μm), collect brain slices from relevant areas, place them in cryopreservation solution (glycerol: 0.01M PBS = 1:1), and store them at -80°C until use. 3.1. Nissl staining Nissl bodies are a kind of basophilic substance in the cytoplasm, which are widely found in various neurons and are used to count the number of brain neurons. Stain according to Nissl staining kit. 3.2. TH (Tyrosine hydroxylase , tyrosine hydroxylase ) staining TH mainly marks dopaminergic neurons in the substantia nigra pars compacta. After MPTP modeling, TH-positive neurons in this brain area will be significantly reduced. Remove the midbrain slices, wash them once with 0.01 M PBS for 10 minutes, incubate with 3% H 2 O 2 for 15 minutes to remove endogenous peroxidase activity, wash away the H 2 O 2 with 0.01 M PBS 10 min × 3, and then use PBS containing 5% BSA and 0.3% Triton After 5 min × 3 times, diaminobenzidin (DAB) was used to develop color in the dark. 4. Cell counting, data processing and analysis. Immunohistochemistry positive cells were counted using the Stereo Investigator, MBF bioscience) to calculate the number of Nissl body-positive cells and TH + cells in the substantia nigra pars compacta. The cell count was ×20 Carry out under the objective lens. All data are expressed as Mean ± SEM, and GraphPad Prism 8.0 statistical analysis software was used for statistical processing. Measurement data were analyzed using One-way ANOVA combined with Dunnett's test to analyze the differences between each group and the modeling group. p < 0.05 indicated that the difference was statistically significant. 5. Experimental results 5.1. Effects of PKT101 and PKT002 on the survival rate of mice after subacute MPTP model. Subacute administration of MPTP caused listlessness, anorexia, weight loss, and tail erect reaction in mice after the model was established. However, this time No mice died during the experiment, and no mice in each group of mice died after drug treatment. 5.2. Effects of PKT101 and PKT002 on body weight changes after the subacute model of MPTP in mice . Subacute administration of MPTP caused the mice to eat less after the model was established. The body weight of the mice decreased slightly within 7 days. After 14 days, the food intake gradually increased, and the body weight gradually increased. Recovery. The treatments in each group had no significant effect on the body weight of mice. The results are shown in Figure 6. 5.3. The effects of PKT101 and PKT002 on the autonomous movement ability of mice in the mine experiment after the MPTP subacute model are shown in Table 6 and Figure 7. In the MPTP subacute model, mice were 1, 3, 7 and 7 after the modeling. A decrease in the total distance traveled in the open field test occurred at 14 days, indicating a decrease in the ability to move independently. The positive control drug Selegiline can significantly improve MPTP-induced decrease in total exercise distance on days 1, 3, 7 and 14. PKT101 and PKT002 also had significant improvement within 14 days. At 14 days, the low, medium and high doses of PKT101 alone increased to 221.1%, 253.9% and 255.3% of the model group; PKT002 alone increased to 213.0% of the model group; the combined administration of PKT101 and PKT002 increased to the model 214.8% of the group. 5.4. The effects of PKT101 and PKT002 on the motor coordination ability of mice in the pole climbing experiment after the MPTP subacute model are shown in Tables 7~8 and Figures 8~9. In the MPTP subacute model, mice were 1, 3, The T-Turn and T-TLA times in the pole climbing experiment were prolonged at 7 and 14 days, indicating a decrease in motor coordination ability. The positive control drug Selegiline can significantly improve MPTP-induced T-Turn and T-TLA time prolongation on days 1, 3, 7 and 14. PKT101 and PKT002 also had significant improvement within 14 days. For T-Turn, at 14 days, the low, medium and high doses of single PKT101 administration were reduced to 26.1%, 24.6% and 26.0% of the model group respectively; the single PKT002 administration was reduced to 24.4% of the model group; PKT101 and PKT002 The combined administration was reduced to 21.6% of the model group. For T-TLA, at 14 days, the low, medium and high doses of single PKT101 administration were reduced to 68.8%, 65.1% and 38.6% of the model group respectively; the single PKT002 administration was reduced to 46.0% of the model group; PKT101 and PKT002 The combined dose reduction was 46.3% of the model group. 5.5. The effects of PKT101 and PKT002 on the motor balance ability of mice in the rotarod test after the MPTP subacute model are shown in Table 9 and Figure 10. In the MPTP subacute model, mice were 1, 3, 7 and 14 days after modeling. The shortening of the falling latency in the rotarod test indicates a decrease in motor balance ability. The positive control drug Selegiline can significantly improve the MPTP-induced shortening of the latency time on days 1, 3, 7 and 14. Both PKT101 and PKT00 had significant improvement within 14 days. At 14 days, the incubation period of low, medium and high doses of PKT101 alone was prolonged to 121.7, 119.9% and 118.3% of the model group respectively; the latency of PKT002 alone was prolonged to 120.3% of the model group; the combined administration of PKT101 and PKT002 prolonged It is 120.9% of the model group. 5.6. The effect of Lizazhen on the number of Nissl-positive neurons in the substantia nigra pars compacta in the subacute MPTP model of mice is shown in Table 10 and Figure 11. In the MPTP subacute model, mice were 7 and 14 days after modeling. The decrease in the number of Nissl stain-positive neurons in the substantia nigra pars compacta indicates the death of neurons in this brain area. The positive control drug Selegiline can significantly improve the MPTP-induced decrease in the number of Nissl stain-positive neurons in the substantia nigra pars compacta on both 7 and 14 days. PKT101 and PKT002 also had significant improvement effects at 7 and 14 days. At 7 days, the number of Nissl-positive cells treated with low, medium and high doses of PKT101 alone increased to 120.9%, 124.0% and 130.9% of the model group respectively; the number of Nissl-positive cells treated with PKT002 alone increased to 120.9%, 124.0% and 130.9% of the model group The number of Nissl-positive cells after combined administration of PKT101 and PKT002 increased to 128.0% of the model group. At 14 days, the number of Nissl-positive cells treated with low, medium and high doses of PKT101 alone increased to 124.9%, 130.9% and 141.1% of the model group respectively; the number of Nissl-positive cells treated with PKT002 alone increased to 124.9%, 130.9% and 141.1% of the model group respectively The number of Nissl-positive cells after combined administration of PKT101 and PKT002 increased to 141.5% of the model group. 5.7. The effects of PKT101 and PKT002 on the number of TH- positive neurons in the substantia nigra pars compacta of the mouse MPTP subacute model are shown in Table 11 and Figure 12. In the MPTP subacute model, mice appeared 7 and 14 days after modeling. The decrease in the number of TH-positive neurons in the substantia nigra pars compacta indicates the death of dopaminergic neurons in this brain area. The positive control drug Selegiline could significantly improve the MPTP-induced decrease in the number of TH-stained positive neurons in the substantia nigra pars compacta on both 7 and 14 days. PKT101 and PKT002 also had significant improvement effects at 7 and 14 days. At 7 days, the number of TH-positive cells after single administration of PKT101 at low, medium and high doses increased to 114.5%, 124.9% and 129.7% of the model group respectively; the number of TH-positive cells after single administration of PKT002 increased to 129.9% of the model group. %; the number of TH-positive cells after combined administration of PKT101 and PKT002 increased to 130.3% of that in the model group. At 14 days, the number of TH-positive cells after single administration of PKT101 at low, medium and high doses increased to 125.6%, 136.0% and 137.5% of the model group respectively; the number of TH-positive cells after single administration of PKT002 increased to 128.1% of the model group. %; the number of TH-positive cells after combined administration of PKT101 and PKT002 increased to 137.7% of that in the model group. 6. Conclusion 7-day and 14-day treatment with polypeptides PKT101 and PKT002 can significantly improve the behavioral disorders of mice after the MPTP subacute model, increase the total movement distance in the open field test, and shorten the T-Turn time and T-Turn time in the pole climbing test. TLA time prolonged the incubation period of the rotarod test, but the dose-dependent relationship between the groups was not obvious. At the same time, both PKT101 and PKT002 can alleviate the MPTP-induced decrease in the number of Nissl-positive and TH-positive neurons in the substantia nigra pars compacta. These results indicate that both PKT101 and PKT002 have neuroprotective effects on the subacute model of MPTP. There is no significant difference between combined administration of PKT101 and PKT002 compared with administration alone. Example 3 - Preliminary study on the protective effect of peptides of the present invention on glial cells and neuron damage in neurodegenerative disease models 1. The purpose of the experiment is to initially explore the protective effect of PKT001 and PKT002 on glial cells and neuron damage at the cellular level. , discuss common mechanisms for the treatment of neurodegenerative diseases. 2. Experimental protocol: Mouse or human glioma cells and neuronal cells were pretreated for 24 hours with 1, 3, 9 ug/ml PKT001, PKT002 experimental group and control group respectively, and then lipopolysaccharide (LPS, 1 μg/ml) was induced for 48 hours, and the proportion of apoptotic cells was analyzed by flow cytometry with Annexin V/propidium iodide (PI) double staining to detect the effects of PKT001 and PKT002 administration on the apoptosis of various nervous system cells. 3. Experimental materials cell line GL261 mouse glioblastoma cells, culture medium: DMEM +10%FBS+ 1%P/S; LN229 human glioma cells, culture medium: DMEM+10%FBS+ 1%P/S; WERI-RB- 1 Human retinal glioma cells, culture medium: RPMI-1640+10%FBS+1%P/S; SK-N-SH human neuroblastoma cells, culture medium: MEM (containing NEAA)+10%FBS+1%P /S; HT22 mouse hippocampal neuron cells, culture medium: DMEM +10%FBS+1%P/S; neuro-2a (N2A) mouse brain neuroma cells, culture medium: MEM (containing NEAA)+10%FBS+ 1%P/S; culture conditions: 37°C, 5% CO 2. The cells used in the experiment are all in the logarithmic growth phase. 4. Experimental steps : Effect of PKT001 and PKT002 on apoptosis of nervous system cells (1) Cell subculture and plating: Take the normally cultured cells, aspirate the original culture medium, add PBS to wash, add trypsin for digestion for 1-3 minutes, and then terminate the digestion. Pipette into single cells with a pipette, centrifuge the cell suspension at 200g for 5 minutes, resuspend in culture medium, take 20 μl of cell suspension and add 20 μl of trypan blue for counting, and plate at 3*10 5 cells/well (6-well plate). 3 replicate wells were cultured overnight at 37°C in 5% CO 2 . (2) Medication treatment: ① Cell; ② Cell+LPS(1 μg/ml); ③ Cell+PKT001(1 μg/ml)+LPS(1 μg/ml); ④ Cell+PKT001(3 μg/ml) +LPS(1 μg/ml); ⑤ Cell+PKT001(9 μg/ml)+LPS(1 μg/ml); ⑥ Cell+PKT002(1 μg/ml)+LPS(1 μg/ml); ⑦ Cell+ PKT002 (3 μg/ml) + LPS (1 μg/ml); ⑧ Cell + PKT002 (9 μg/ml) + LPS (1 μg/ml); a total of 8 groups. The drug was first administered and pretreated for 24 hours, and then LPS Induction treatment was carried out for 48 hours, and then relevant detection was carried out. (3) Apoptosis detection: ① Collect cells: 200g, centrifuge at 4°C for 5 minutes to collect cells. ② Wash the cells twice with pre-cooled PBS, 200g each time, and centrifuge at 4°C for 5 minutes. ③ Aspirate the PBS and add 100μL 1×Binding Buffer to resuspend the cells. ④ Add 5μL Annexin FITC and 2μL PI, and mix gently. ⑤ Protect from light and react at room temperature for 15 minutes. ⑥ Add 300μL 1×Binding Buffer, mix well and place on ice. The sample will be detected by flow cytometry within 1 hour. 5. Experimental results 5.1. The effects of PKT001 and PKT002 on the apoptosis of glioblastoma cells induced by LPS in GL261 mice were analyzed using Annexin V-FITC/PI double staining flow cytometry. The experimental results showed that compared with the LPS control group, 1- After pretreatment with 9 μg/ml PKT001, the proportion of apoptosis in GL261 cells induced by LPS at 48 h was significantly reduced; after pretreatment with 1-9 μg/ml PKT002, the proportion of apoptosis in GL261 cells induced by LPS at 48 h was significantly reduced. The results are shown in Figure 13. 5.2. Effects of PKT001 and PKT002 on LPS -induced apoptosis of LN229 human glioma cells . Compared with the LPS control group, after pretreatment with 1 μg/ml PKT001, the proportion of LN229 cell apoptosis induced by LPS did not change significantly at 48 h, at 3 μg/ml and 9 μg. /ml After pretreatment with PKT001, the proportion of LN229 cell apoptosis induced by LPS at 48 h was significantly reduced. After pretreatment with 1-9 μg/ml PKT002, the proportion of LN229 cell apoptosis induced by LPS at 48 h was significantly reduced. The results are shown in Figure 14. 5.3. Effects of PKT001 and PKT002 on LPS -induced apoptosis of WERI-RB-1 human retinal glioma cells . Compared with the LPS control group, after pretreatment with 1-9 μg/ml PKT001, LPS induced apoptosis of WERI-RB-1 cells for 48 h. The apoptosis ratio was significantly reduced; after pretreatment with 1-9 μg/ml PKT002, the apoptosis ratio of WERI-RB-1 cells was significantly reduced 48 h after LPS induction. The results are shown in Figure 15. 5.4. Effects of PKT001 and PKT002 on LPS- induced apoptosis of neuro-2a (N2A) mouse brain neuroma cells . Compared with the LPS control group, LPS induced apoptosis of N2A cells at 48 h after 1 μg/ml and 3 μg/ml PKT001 pretreatment. The proportion did not change significantly. After pretreatment with 9 μg/ml PKT001, the proportion of N2A cell apoptosis induced by LPS at 48 h decreased, and there was a significant difference. After pretreatment with 1-9 μg/ml PKT002, the proportion of N2A cell apoptosis induced by LPS at 48 h was significantly reduced. The results are shown in Figure 16. 5.5. Effects of PKT001 and PKT002 on LPS- induced apoptosis of hippocampal neuron cells in HT22 mice. Experimental results show that compared with the LPS control group, after pretreatment with 1-9 μg/ml PKT001, the proportion of LPS-induced apoptosis in HT22 cells at 48 h was significantly reduced. ; After pretreatment with 1-9 μg/ml PKT002, the proportion of LPS-induced apoptosis in HT22 cells at 48 h was significantly reduced. The results are shown in Figure 17. 5.6. Effects of PKT001 and PKT002 on LPS -induced apoptosis of SK-N-SH human neuroblastoma cells . Compared with the LPS control group, after pretreatment with 1 μg/ml PKT001, the apoptosis ratio of SK-N-SH cells induced by LPS at 48 h. There was no significant change. After pretreatment with PKT001 at 3 μg/ml and 9 μg/ml, the apoptosis rate of SK-N-SH cells induced by LPS at 48 h was significantly reduced. After pretreatment with PKT002 at 1-9 μg/ml, the apoptosis rate of SK-N-SH cells induced by LPS at 48 h was significantly reduced. The results are shown in Figure 18.

[圖1]. PKT101、PKT002和PKT101/PKT002聯合服用對APP/PS1小鼠短期工作記憶、條件恐懼記憶和社交能力的影響。(A)Y迷宮實驗中對照組(Con),PKT101,PKT002和PKT101/PKT002聯合治療組小鼠進入新奇臂時間百分比的統計圖。(B)新物體識別實驗中,四組小鼠新物體識別認知指數統計圖。(C)條件恐懼記憶實驗中,四組小鼠僵直不動時間統計圖。(D)社交偏好和社會記憶實驗中,(左) 小鼠進入Empty室和Stranger-1室的時間百分比統計圖;(右) 小鼠進入Stranger-1室和Stranger-2室的時間百分比統計圖。結果以均值±標準誤的方式表示,n = 13-15每組。資料在D圖中,四組小鼠進入Empty室和Stranger-1的時間百分的比較以及四組小鼠進入Stranger-1室和Stranger-2室的時間百分比的比較使用T檢驗,其他統計圖使用單因素方差分析。Enter number: 進入次數;Recognition index: 認知指數;Time spent: 耗時;% of freezing: 僵直時間百分比。 [圖2]. PKT101、PKT002和PKT101/PKT002聯合服用對APP/PS1小鼠海馬和皮層Aβ斑塊沉積的影響。(A)對照組(Con)、PKT101、PKT002和PKT101/ PKT002聯合治療組小鼠硫磺素S染色的螢光圖。尺規為200 μm。(B)免疫螢光實驗顯示四組小鼠皮層和海馬6E10表達的螢光圖。尺規為200 μm。(C)四組小鼠皮層和海馬硫磺素S陽性表達Aβ斑塊定量統計圖。(D)四組小鼠皮層和海馬6E10陽性表達面積百分比的統計圖。結果以均值±標準誤的方式表示,n = 4-5每組,統計圖使用單因素方差分析。Cortext:皮層;Hippocampus:海馬體;Relative Density:相對密度;Positive area of 6E10+:6E10+的陽性區域。 [圖3]. PKT101、PKT002和PKT101/PKT002聯合服用對APP/PS1小鼠海馬Aβ斑塊周圍膠質細胞活化的影響。(A)免疫螢光實驗顯示對照組(Con)、PKT101、PKT002和PKT101/PKT002聯合治療組組小鼠海馬Iba1,GFAP和6E10表達的螢光圖。尺規為50 μm。(B)四組小鼠海馬Iba1和GFAP陽性表達面積百分比統計圖。結果以均值±標準誤的方式表示,n = 4-5每組,統計圖使用單因素方差分析。Positive area of Iba1+:Iba1+的陽性區域;Positive area of GFAP+:GFAP+的陽性區域。 [圖4]. PKT101、PKT002和PKT101/PKT002聯合服用對APP/PS1小鼠皮層Aβ斑塊周圍膠質細胞活化的影響。(A)免疫螢光實驗顯示對照組(Con)、PKT101、PKT002和PKT101/PKT002聯合治療組小鼠皮層Iba1,GFAP和6E10表達的螢光圖。尺規為50 μm。(B)四組小鼠皮層Iba1和GFAP陽性表達面積百分比統計圖。結果以均值 ± 標準誤的方式表示,n = 5每組,統計圖使用單因素方差分析。Positive area of Iba1+:Iba1+的陽性區域;Positive area of GFAP+:GFAP+的陽性區域。 [圖5]. PKT101、PKT002和PKT101/PKT002聯合服用對APP/PS1小鼠海馬和皮層區IL-1β,IL-6和TNF-α含量的影響。(A)ELISA檢測對照組(Con)、PKT101、PKT002和PKT101/PKT002聯合治療組小鼠海馬中炎症因數IL-1β,IL-6和TNF-α的表達。(B)ELISA檢測四組小鼠皮層中炎症因數IL-1β,IL-6和TNF-α的表達。結果以均值 ± 標準誤的方式表示,n = 5每組,統計圖使用單因素方差分析。Cortext:皮層;Hippocampus:海馬體;Relative expression of IL-1β:IL-1β的相對表達;;Relative expression of IL-6:IL-6的相對表達;;Relative expression of TNF-α:TNF-α的相對表達。 [圖6]. PKT101和PKT002對小鼠MPTP亞急性模型後體重改變的影響。0~7天時,n=20或28;14天時,n=10或14。Sham:正常對照組;Selegiline:司來吉蘭治療組。 [圖7]. PKT101和PKT002對MPTP亞急性模型後小鼠在礦場實驗中總運動路程的影響(1、3、7和14天)。 *** p<0.001, ** p<0.01 vs.Sham; ### p<0.001, ## p<0.01, # p<0.05 vs.MPTP。 [圖8]. PKT101和PKT002對MPTP亞急性模型後小鼠在爬杆實驗中T-Turn的影響(1、3、7和14天)。***p<0.001, **p<0.01 vs. Sham;###p<0.001, ##p<0.01, #p<0.05 vs. MPTP。 [圖9]. PKT101和PKT002對MPTP亞急性模型後小鼠在爬杆實驗中T-TLA的影響(1、3、7和14天)。***p<0.001 vs. Sham; ###p<0.001, ##p<0.01, #p<0.05 vs. MPTP。 [圖10]. PKT101和PKT002對MPTP亞急性模型後小鼠在轉棒實驗中潛伏期的影響(1、3、7和14天)。***p<0.001 vs. Sham; ###p<0.001 vs. MPTP。 [圖11]. PKT101和PKT002對MPTP亞急性模型後小鼠黑質緻密部尼氏陽性神經元數量的影響(7和14天)。***p<0.001 vs. Sham; ##p<0.01, #p<0.05 vs. MPTP; 尺規為250µm。 [圖12]. PKT101和PKT002對MPTP亞急性模型後小鼠黑質緻密部TH陽性神經元數量的影響(7和14天)。*p<0.05 vs. Sham; ###p<0.001, ##p<0.01 vs. MPTP; 尺規為250µm。 [圖13]. PKT001和PKT002預處理對LPS誘導GL261細胞凋亡的影響。 [圖14]. PKT001和PKT002預處理對LPS誘導LN229凋亡的影響。 [圖15]. PKT001和PKT002預處理對LPS誘導WERI-RB-1細胞凋亡的影響。 [圖16]. PKT001和PKT002預處理對LPS誘導N2A細胞凋亡的影響。 [圖17]. PKT001和PKT002預處理對LPS誘導HT22細胞凋亡的影響。 [圖18]. PKT001和PKT002預處理對LPS誘導SK-N-SH細胞凋亡的影響。 [Figure 1]. Effects of combined administration of PKT101, PKT002 and PKT101/PKT002 on short-term working memory, conditioned fear memory and social ability of APP/PS1 mice. (A) Statistical chart showing the percentage of time mice in the control group (Con), PKT101, PKT002 and PKT101/PKT002 combined treatment groups entered the novel arm in the Y maze experiment. (B) In the new object recognition experiment, the statistical chart of the new object recognition cognitive index of the four groups of mice. (C) Statistical chart of freezing time of mice in four groups during the conditioned fear memory experiment. (D) In the social preference and social memory experiment, (left) statistical graph of the percentage of time mice entered the Empty chamber and Stranger-1 chamber; (right) statistical graph of the percentage of time mice entered the Stranger-1 chamber and Stranger-2 chamber . Results are expressed as mean ± SE, n = 13-15 per group. The data is in Figure D. The comparison of the percentage of time for the four groups of mice to enter the Empty chamber and Stranger-1 and the comparison of the percentage of time for the four groups of mice to enter the Stranger-1 chamber and Stranger-2 chamber are using T test. Other statistical charts One-way analysis of variance was used. Enter number: number of entries; Recognition index: cognitive index; Time spent: time spent; % of freezing: percentage of freezing time. [Figure 2]. Effects of combined administration of PKT101, PKT002 and PKT101/PKT002 on Aβ plaque deposition in the hippocampus and cortex of APP/PS1 mice. (A) Fluorescence images of thioflavin S staining of mice in the control group (Con), PKT101, PKT002 and PKT101/PKT002 combined treatment groups. The ruler gauge is 200 μm. (B) Immunofluorescence experiment shows fluorescence images of 6E10 expression in the cortex and hippocampus of four groups of mice. The ruler gauge is 200 μm. (C) Quantitative statistical diagram of Thioflavin S-positive Aβ plaques in the cortex and hippocampus of four groups of mice. (D) Statistical chart of the percentage of 6E10 positive expression areas in the cortex and hippocampus of four groups of mice. Results are expressed as mean ± standard error, n = 4-5 per group, and statistical graphs used one-way analysis of variance. Cortext: cortex; Hippocampus: hippocampus; Relative Density: relative density; Positive area of 6E10+: the positive area of 6E10+. [Figure 3]. Effects of combined administration of PKT101, PKT002 and PKT101/PKT002 on glial cell activation around hippocampal Aβ plaques in APP/PS1 mice. (A) Immunofluorescence experiment shows fluorescence images of Iba1, GFAP and 6E10 expression in the hippocampus of mice in the control group (Con), PKT101, PKT002 and PKT101/PKT002 combined treatment groups. The ruler gauge is 50 μm. (B) Statistical chart of the percentage of positive expression areas of Iba1 and GFAP in the hippocampus of four groups of mice. Results are expressed as mean ± standard error, n = 4-5 per group, and statistical graphs used one-way analysis of variance. Positive area of Iba1+: positive area of Iba1+; Positive area of GFAP+: positive area of GFAP+. [Figure 4]. Effects of combined administration of PKT101, PKT002 and PKT101/PKT002 on glial cell activation around cortical Aβ plaques in APP/PS1 mice. (A) Immunofluorescence experiment shows fluorescence images of Iba1, GFAP and 6E10 expression in the cortex of mice in the control group (Con), PKT101, PKT002 and PKT101/PKT002 combined treatment groups. The ruler gauge is 50 μm. (B) Statistical chart of the percentage of positive expression areas of Iba1 and GFAP in the cortex of four groups of mice. Results are expressed as mean ± standard error, n = 5 per group, and statistical graphs were analyzed using one-way analysis of variance. Positive area of Iba1+: positive area of Iba1+; Positive area of GFAP+: positive area of GFAP+. [Figure 5]. Effects of combined administration of PKT101, PKT002 and PKT101/PKT002 on the contents of IL-1β, IL-6 and TNF-α in the hippocampus and cortex of APP/PS1 mice. (A) ELISA detects the expression of inflammatory factors IL-1β, IL-6 and TNF-α in the hippocampus of mice in the control group (Con), PKT101, PKT002 and PKT101/PKT002 combined treatment groups. (B) ELISA detects the expression of inflammatory factors IL-1β, IL-6 and TNF-α in the cortex of four groups of mice. Results are expressed as mean ± standard error, n = 5 per group, and statistical graphs were analyzed using one-way analysis of variance. Cortext: cortex; Hippocampus: hippocampus; Relative expression of IL-1β: relative expression of IL-1β;; Relative expression of IL-6: relative expression of IL-6;; Relative expression of TNF-α: TNF-α Relative expression. [Figure 6]. Effects of PKT101 and PKT002 on body weight changes after subacute model of MPTP in mice. When 0 to 7 days old, n=20 or 28; when 14 days old, n=10 or 14. Sham: normal control group; Selegiline: Selegiline treatment group. [Figure 7]. Effects of PKT101 and PKT002 on the total movement distance of mice in the mine experiment after MPTP subacute model (1, 3, 7 and 14 days). *** p <0.001, ** p <0.01 vs. Sham; ### p <0.001, ## p <0.01, # p <0.05 vs. MPTP. [Figure 8]. Effects of PKT101 and PKT002 on T-Turn of mice in the pole climbing experiment after subacute model of MPTP (1, 3, 7 and 14 days). ***p<0.001, **p<0.01 vs. Sham; ###p<0.001, ##p<0.01, #p<0.05 vs. MPTP. [Figure 9]. Effects of PKT101 and PKT002 on T-TLA in the rod climbing experiment of mice after subacute model of MPTP (1, 3, 7 and 14 days). ***p<0.001 vs. Sham; ###p<0.001, ##p<0.01, #p<0.05 vs. MPTP. [Figure 10]. Effects of PKT101 and PKT002 on the incubation period of mice in the rotarod experiment after subacute MPTP model (1, 3, 7 and 14 days). ***p<0.001 vs. Sham; ###p<0.001 vs. MPTP. [Figure 11]. Effects of PKT101 and PKT002 on the number of Nissl-positive neurons in the substantia nigra pars compacta of mice after the subacute model of MPTP (7 and 14 days). ***p<0.001 vs. Sham; ##p<0.01, #p<0.05 vs. MPTP; The ruler is 250µm. [Figure 12]. Effects of PKT101 and PKT002 on the number of TH-positive neurons in the substantia nigra pars compacta of mice after the subacute model of MPTP (7 and 14 days). *p<0.05 vs. Sham; ###p<0.001, ##p<0.01 vs. MPTP; Ruler gauge is 250µm. [Figure 13]. Effect of PKT001 and PKT002 pretreatment on LPS-induced apoptosis of GL261 cells. [Figure 14]. Effect of PKT001 and PKT002 pretreatment on LPS-induced apoptosis of LN229. [Figure 15]. Effect of PKT001 and PKT002 pretreatment on LPS-induced apoptosis of WERI-RB-1 cells. [Figure 16]. Effect of PKT001 and PKT002 pretreatment on LPS-induced apoptosis of N2A cells. [Figure 17]. Effect of PKT001 and PKT002 pretreatment on LPS-induced apoptosis of HT22 cells. [Figure 18]. Effect of PKT001 and PKT002 pretreatment on LPS-induced apoptosis of SK-N-SH cells.

TW202340233A_112112599_SEQL.xmlTW202340233A_112112599_SEQL.xml

Claims (15)

一種肽在製備用於預防、治療或改善有需要的受試者中神經退行性疾病或認知障礙症的組合物中的用途,所述肽選自: a) 包含與DEAQETAVSSH (SEQ ID NO: 1)具有至少70%同一性的氨基酸序列的肽; b) 包含在DEAQETAVSSH (SEQ ID NO: 1)中具有0至4個氨基酸突變的氨基酸序列的肽; c) 包含與DEAQETAVSSHEQD (SEQ ID NO: 2)具有至少70%同一性的氨基酸序列的肽;或 d) 包含在DEAQETAVSSHEQD (SEQ ID NO: 2)中具有0至4個氨基酸突變的氨基酸序列的肽。 Use of a peptide selected from the group consisting of: a) A peptide comprising an amino acid sequence that is at least 70% identical to DEAQETAVSSH (SEQ ID NO: 1); b) A peptide comprising an amino acid sequence having 0 to 4 amino acid mutations in DEAQETAVSSH (SEQ ID NO: 1); c) A peptide comprising an amino acid sequence that is at least 70% identical to DEAQETAVSSHEQD (SEQ ID NO: 2); or d) A peptide comprising an amino acid sequence having 0 to 4 amino acid mutations in DEAQETAVSSHEQD (SEQ ID NO: 2). 如請求項1的用途,其中所述認知障礙症由神經退行性疾病引起。The use of claim 1, wherein the cognitive impairment is caused by a neurodegenerative disease. 如請求項1或2的用途,其中神經退行性疾病或認知障礙症選自阿爾茨海默病或帕金森病。The use of claim 1 or 2, wherein the neurodegenerative disease or cognitive impairment is selected from Alzheimer's disease or Parkinson's disease. 一種肽在製備用於改善、增強或恢復有需要的受試者中認知功能或認知能力,優選包括感知能力、思維邏輯能力、記憶能力、語言能力、學習能力、情緒控制能力、社交能力或注意力的組合物中的用途,所述肽選自: a) 包含與DEAQETAVSSH (SEQ ID NO: 1)具有至少70%同一性的氨基酸序列的肽; b) 包含在DEAQETAVSSH (SEQ ID NO: 1)中具有0至4個氨基酸突變的氨基酸序列的肽; c) 包含與DEAQETAVSSHEQD (SEQ ID NO: 2)具有至少70%同一性的氨基酸序列的肽;或 d) 包含在DEAQETAVSSHEQD (SEQ ID NO: 2)中具有0至4個氨基酸突變的氨基酸序列的肽。 A peptide prepared for improving, enhancing or restoring cognitive function or cognitive ability in a subject in need, preferably including perceptual ability, logical thinking ability, memory ability, language ability, learning ability, emotional control ability, social ability or attention. For use in a composition of force, the peptide is selected from: a) A peptide comprising an amino acid sequence that is at least 70% identical to DEAQETAVSSH (SEQ ID NO: 1); b) A peptide comprising an amino acid sequence having 0 to 4 amino acid mutations in DEAQETAVSSH (SEQ ID NO: 1); c) A peptide comprising an amino acid sequence that is at least 70% identical to DEAQETAVSSHEQD (SEQ ID NO: 2); or d) A peptide comprising an amino acid sequence having 0 to 4 amino acid mutations in DEAQETAVSSHEQD (SEQ ID NO: 2). 如請求項4的用途,其中 所述受試者具有認知功能或認知能力衰退,例如年齡老化而引致的或伴隨年齡老化的認知功能或認知能力衰退;或者 所述受試者患有認知障礙症或神經退行性疾病,優選由神經退行性疾病引起的認知障礙症,更優選阿爾茨海默病或帕金森病。 Such as the purpose of request item 4, where The subject has cognitive function or cognitive ability decline, such as cognitive function or cognitive ability decline caused by or accompanying aging; or The subject suffers from cognitive impairment or neurodegenerative disease, preferably cognitive impairment caused by neurodegenerative disease, more preferably Alzheimer's disease or Parkinson's disease. 如請求項1-3和5中任一項的用途,其中認知障礙症特徵在於溝通障礙、判斷力差、難以進行簡單任務、遺漏或放錯物件、語言障礙、性格突變、行為能力下降、時間及空間感混亂、理解能力下降、問題解決能力下降、注意力下降、社交能力下降、感知障礙、思維邏輯障礙或記憶障礙。Such as the use of any of claims 1-3 and 5, wherein dementia is characterized by communication difficulties, poor judgment, difficulty in performing simple tasks, missing or misplacing objects, language impairment, personality changes, behavioral decline, time and confusion in spatial sense, decreased understanding, decreased problem-solving ability, decreased concentration, decreased social skills, perceptual impairment, logical thinking impairment, or memory impairment. 如前述請求項中任一項的用途,其中所述神經退行性疾病或認知障礙症為阿爾茨海默病,其中 a) 所述阿爾茨海默病特徵在於在大腦中β-澱粉樣蛋白(Aβ)沉積、神經炎症、和/或膠質細胞異常或活化;或者 b) 阿爾茨海默病的預防、治療或改善通過減少或緩解在大腦中β-澱粉樣蛋白(Aβ)沉積、神經炎症和/或膠質細胞異常或活化來實現。 The use of any one of the preceding claims, wherein the neurodegenerative disease or cognitive impairment is Alzheimer's disease, wherein a) the Alzheimer's disease is characterized by beta-amyloid (Aβ) deposition, neuroinflammation, and/or glial cell abnormalities or activation in the brain; or b) Prevention, treatment or improvement of Alzheimer's disease is achieved by reducing or alleviating beta-amyloid (Aβ) deposition, neuroinflammation and/or glial cell abnormalities or activation in the brain. 如前述請求項中任一項的用途,其中所述神經退行性疾病或認知障礙症為帕金森病,其中 a) 所述帕金森病特徵在於大腦黑質神經元異常或損傷;或者 b) 帕金森病的預防、治療或改善通過減少或緩解大腦黑質神經元異常或損傷來實現。 The use of any one of the preceding claims, wherein the neurodegenerative disease or cognitive impairment is Parkinson's disease, wherein a) The Parkinson's disease is characterized by abnormality or damage to neurons in the substantia nigra of the brain; or b) The prevention, treatment or improvement of Parkinson's disease is achieved by reducing or alleviating neuronal abnormalities or damage in the substantia nigra of the brain. 如前述請求項中任一項的用途,所述受試者是人,優選老年人。As used in any one of the preceding claims, the subject is a human, preferably an elderly person. 如前述請求項中任一項的用途,所述組合物為藥物組合物或營養組合物,優選所述藥物組合物或營養組合物包含所述肽和藥用輔料、溶媒或載體。As used in any one of the preceding claims, the composition is a pharmaceutical composition or nutritional composition. Preferably, the pharmaceutical composition or nutritional composition contains the peptide and pharmaceutical excipients, solvents or carriers. 如前述請求項中任一項的用途,所述肽選自: a) 包含與DEAQETAVSSH (SEQ ID NO: 1)具有至少80%同一性的氨基酸序列的肽; b) 包含在DEAQETAVSSH (SEQ ID NO: 1)中具有0至3個氨基酸突變的氨基酸序列的肽; c) 包含與DEAQETAVSSHEQD (SEQ ID NO: 2)具有至少80%同一性的氨基酸序列的肽;或 d) 包含在DEAQETAVSSHEQD (SEQ ID NO: 2)中具有0至3個氨基酸突變的氨基酸序列的肽。 As used in any of the preceding claims, the peptide is selected from: a) A peptide comprising an amino acid sequence that is at least 80% identical to DEAQETAVSSH (SEQ ID NO: 1); b) A peptide comprising an amino acid sequence having 0 to 3 amino acid mutations in DEAQETAVSSH (SEQ ID NO: 1); c) A peptide comprising an amino acid sequence that is at least 80% identical to DEAQETAVSSHEQD (SEQ ID NO: 2); or d) A peptide comprising an amino acid sequence having 0 to 3 amino acid mutations in DEAQETAVSSHEQD (SEQ ID NO: 2). 如前述請求項中任一項的用途,所述氨基酸突變選自氨基酸的添加、缺失或取代。As used in any one of the preceding claims, the amino acid mutation is selected from addition, deletion or substitution of amino acids. 如前述請求項中任一項的用途,所述氨基酸突變位於氨基酸序列的C-端和/或N-端,優選是氨基酸序列的C-端和/或N-端的添加或缺失。As used in any one of the preceding claims, the amino acid mutation is located at the C-terminus and/or N-terminus of the amino acid sequence, preferably the addition or deletion of the C-terminus and/or N-terminus of the amino acid sequence. 如前述請求項中任一項的用途,所述肽具有8至20個氨基酸的長度,優選10至18個氨基酸長度,更優選11至14個氨基酸長度。As used in any one of the preceding claims, the peptide has a length of 8 to 20 amino acids, preferably 10 to 18 amino acids in length, more preferably 11 to 14 amino acids in length. 如前述請求項中任一項的用途,所述肽選自 1) 包含SEQ ID NO: 1的氨基酸序列、基本上由SEQ ID NO: 1的氨基酸序列組成或者由SEQ ID NO: 1的氨基酸序列表示的肽;或者 2) 包含SEQ ID NO: 2的氨基酸序列、基本上由SEQ ID NO: 2的氨基酸序列組成或者由SEQ ID NO: 2的氨基酸序列表示的肽。 As used in any of the preceding claims, the peptide is selected from 1) A peptide comprising, consisting essentially of, or represented by the amino acid sequence of SEQ ID NO: 1; or 2) A peptide comprising, consisting essentially of, or represented by the amino acid sequence of SEQ ID NO: 2.
TW112112599A 2022-04-01 2023-03-31 Use of peptide in treating neurodegenerative disease or ameliorating cognitive function TW202340233A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
WOPCT/CN2022/084784 2022-04-01
PCT/CN2022/084784 WO2023184473A1 (en) 2022-04-01 2022-04-01 Use of peptide in treating neurodegenerative diseases or improving cognitive function

Publications (1)

Publication Number Publication Date
TW202340233A true TW202340233A (en) 2023-10-16

Family

ID=88198867

Family Applications (1)

Application Number Title Priority Date Filing Date
TW112112599A TW202340233A (en) 2022-04-01 2023-03-31 Use of peptide in treating neurodegenerative disease or ameliorating cognitive function

Country Status (2)

Country Link
TW (1) TW202340233A (en)
WO (2) WO2023184473A1 (en)

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090232910A1 (en) * 2004-07-22 2009-09-17 Duke University Biomarkers and therapeutics targets for cognitive decline
WO2008031190A1 (en) * 2006-09-15 2008-03-20 Osta Biotechnologies Inc. Alpha-1-antitrypsin as a diagnostic/prognostic indicator for neurodegenerative diseases
ES2332645B1 (en) * 2009-06-30 2010-10-18 Grifols, S.A. USE OF ALFA-1-ANTITRIPSIN FOR THE PREPARATION OF MEDICINES FOR THE TREATMENT OF CHRONIC FATIGUE SYNDROME.
US20130203650A1 (en) * 2010-08-31 2013-08-08 Yissum Research Development Company Of The Hebrew Uviversity Of Jerusalem Polypeptides derived from alpha-1 antitrypsin and methods of use thereof
CN104321337B (en) * 2012-05-25 2017-07-11 永林有限公司 Peptide and application thereof
US10130676B2 (en) * 2015-04-16 2018-11-20 Prime Bio-Drug Development Limited Therapeutic peptides for cerebrovascular diseases
CN107849091B (en) * 2015-04-16 2021-09-10 医诺生药开发有限公司 Peptides and derivatives thereof capable of inhibiting hepatitis C virus replication in human adipose-derived stem cells and hepatocytes
TW201711695A (en) * 2016-08-19 2017-04-01 首創生物藥品發展有限公司 Therapeutic peptides for cerebrovascular diseases
WO2021009288A1 (en) * 2019-07-16 2021-01-21 Fundació Hospital Universitari Vall D'hebron - Institut De Recerca Combination comprising alpha-1 antitrypsin for use in treating ischaemia in a subject
JP2020063311A (en) * 2020-01-31 2020-04-23 プライム・バイオ‐ドラッグ・ディヴェロップメント・リミテッドPrime Bio‐Drug Development Limited Therapeutic peptides for cerebrovascular diseases

Also Published As

Publication number Publication date
WO2023184473A1 (en) 2023-10-05
WO2023186062A1 (en) 2023-10-05

Similar Documents

Publication Publication Date Title
Yang et al. Systemic inflammation induces anxiety disorder through CXCL12/CXCR4 pathway
JP2019533636A (en) AuC-containing material, method for producing the same, and use thereof
US20110053859A1 (en) Methods to reduce the effects of sleep deprivation
Chen et al. Activation of mGluR2/3 underlies the effects of N-acetylcystein on amygdala-associated autism-like phenotypes in a valproate-induced rat model of autism
Chang et al. NeuroHIV and use of addictive substances
CN108472393A (en) Method for treating Alzheimer disease He having related disorders
JP2019518034A (en) Lipids with an odd number of carbon atoms and their use as pharmaceutical compositions or nutraceuticals
Saavedra et al. Long-term activation of hippocampal glial cells and altered emotional behavior in male and female adult rats after different neonatal stressors
Ma et al. The effect of acori graminei rhizoma and extract fractions on spatial memory and hippocampal neurogenesis in amyloid beta 1-42 injected mice
Kim et al. Effects of Panax ginseng CA Meyer extract on the offspring of adult mice with maternal immune activation
Zhang et al. Environmental enrichment reverses maternal sleep deprivation-induced anxiety-like behavior and cognitive impairment in CD-1 mice
Xu et al. Schisantherin B improves the pathological manifestations of mice caused by behavior desperation in different ages-depression with cognitive impairment
Komleva et al. Expression of NLRP3 inflammasomes in neurogenic niche contributes to the effect of spatial learning in physiological conditions but not in Alzheimer’s type neurodegeneration
Asakawa et al. Oral administration of D-serine prevents the onset and progression of colitis in mice
Wang et al. Chrysin alleviates lipopolysaccharide-induced neuron damage and behavioral deficits in mice through inhibition of Fyn
JP2021512086A (en) How to prevent or treat Alzheimer&#39;s disease
Qian et al. Activation of the CD200/CD200R1 axis attenuates neuroinflammation and improves postoperative cognitive dysfunction via the PI3K/Akt/NF-κB signaling pathway in aged mice
WO2012144501A1 (en) Anti-allergy substance, anti-allergy agent, and food
KR20200116054A (en) Composition for preventing or treating neuroinflammation diseases comprising bee venom extract
WO2023186062A1 (en) Use of peptide in treating neurodegenerative disease or ameliorating cognitive function
JP7249433B2 (en) Composition for prevention or treatment of neuroinflammatory disease containing bee venom extract as an active ingredient
US20220110899A1 (en) Compositions comprising amino acids for use and treatment of central nervous system injuries
Gharedaghi et al. Dinitrobenzene sulphonic acid-induced colitis impairs spatial recognition memory in mice: roles of N-methyl D-aspartate receptors and nitric oxide
Zhang et al. Sleep deprivation aggravates lipopolysaccharide-induced anxiety, depression and cognitive impairment: The role of pro-inflammatory cytokines and synaptic plasticity-associated proteins
Cheng et al. Xenon inhalation attenuates neuronal injury and prevents epilepsy in febrile seizure Sprague-Dawley pups