博碩士論文 107324034 詳細資訊




以作者查詢圖書館館藏 以作者查詢臺灣博碩士 以作者查詢全國書目 勘誤回報 、線上人數:72 、訪客IP:3.139.107.131
姓名 徐嘉宏(Chia-Hung Hsu)  查詢紙本館藏   畢業系所 化學工程與材料工程學系
論文名稱 蝦紅素微脂體的製備與安定性之研究
相關論文
★ 在低溫下以四氯化鈦製備高濃度二氧化鈦結晶覆膜液★ 水熱法合成細顆粒鈦酸鋇
★ 合成均一粒徑球形二氧化鈦★ 共沉澱法合成細顆粒鈦酸鋇
★ 中孔型沸石的晶體形狀之研究★ 含釩或鎵金屬之中孔型分子篩的合成與鑑定
★ 奈米級二氧化鈦及鈦酸鋇之合成與鑑定★ 汽機車尾氣在富氧條件下NOx之去除
★ 耐高溫燃燒觸媒的配製及鑑定★ 高效率醋酸乙酯生產製程研究
★ 製備參數對水熱法製備球形奈米鈦酸鋇粉體之影響研究★ Au/FexOy 奈米材料之製備 及CO 氧化的應用
★ 非晶態奈米鐵之製備與催化性質研究★ 奈米含銀二氧化鈦光觸媒之製備與應用
★ 非晶形奈米鎳合金觸媒的製備及其 在對-氯硝基苯液相選擇性氫化反應之研究★ 奈米金/氧化鈰觸媒之製備及在氧化反應之應用
檔案 [Endnote RIS 格式]    [Bibtex 格式]    [相關文章]   [文章引用]   [完整記錄]   [館藏目錄]   至系統瀏覽論文 ( 永不開放)
摘要(中) 本研究以磷脂質和膽固醇,作為微脂體的包覆材料,形成空心球狀的雙分子磷脂層膜,由於生物體的細胞膜結構也是雙分子磷脂層膜,因此與人體具有良好的相容性和生物可分解性,而球狀結構使其能作為藥物和抗氧化劑的載體,且能有效保護其內容物,在醫學、食品、保養品方面廣為大家所用。本研究使用脂質薄膜水合法製備蝦紅素微脂體溶液,將蝦紅素包覆於微脂體中,使其能保護蝦紅素不被溫度和光線分解,傳統上使用氯仿、甲醇和醚類作為合成上的溶劑,而本研究使用乙醇和天然食品作為其材料,在食安問題上較不會有疑慮。本研究首先改變不同比例的磷脂質和膽固醇來製備微脂體溶液,並將各樣品分別至於4 oC和25 oC放置60天,對其做安定性測試,分別在第7、14、30、60天時對其進行測試,利用紫外光-可見光近紅外光光譜儀觀察其濃度和包覆效果的變化,動態光散射分析儀做粒徑測試,並以光學顯微鏡對其外觀進行觀測,最後找出最佳的合成比例。
由於產品之後希望進行商業化生產,經由上述之實驗,找出最佳的包覆比例後,會進行劑量的放大測試,研究上先以放大5倍做測試,在劑量提升下,希望蝦紅素微脂體溶液仍保有小劑量時的效果,實驗上一樣會分別保存4 oC和25 oC環境,並對樣品做濃度、包覆率、粒徑和外觀的測試。
由本實驗結果得知,樣品須保存在4oC的條件下,且在磷脂質和膽固醇以4:1的條件下合成出來的微脂體具有最好的包覆效果(達92 %)和最小的粒徑(96.3 nm),外觀上呈現一顆一顆圓形橘紅色,有效證明蝦紅素被包覆於微脂體中,而在放大規模的實驗中,得到與小劑量時類似的效果,在未來若能有效應用於商業化上,將具有良好發展潛力。
摘要(英) In this study, phospholipids and cholesterol were used as coating materials for liposomes to form a hollow spherical bimolecular phospholipid layer membrane. Since the cell membrane structure of an organism is also a bimolecular phospholipid layer membrane, it has good compatibility with the human body and has high biodegradability. The spherical structure can be used as a carrier for drugs and antioxidants, and can effectively protect its contents. It is widely used in medicine, food, and maintenance products. In this study, a thin film hydration method was used to prepare astaxanthin liposome solution. Astaxanthin was encapsulated in the liposome to protect it from decomposition under high temperature and light. Traditionally, chloroform, methanol and ether were used as a solvent in synthesis. In this study ethanol and natural food were used, so there is no doubt on food safety. In this study, different ratios of phospholipids and cholesterol were used to prepare liposome solutions, and each sample was placed at 4 oC and 25 oC, respectively, for 60 days. The stability test was performed on days 7, 14, 30, and 60, respectively. Ultraviolet-visible near-infrared spectrometer (UV-vis) was used to determine its concentration and coating effect, dynamic light scattering analyzer (DLS) was used to do particle size test, and optical microscope (OM) was used to observe its appearance.
Since the product is expected to be commercialized afterwards, a scale-up test was conducted. In the research, the test was firstly performed at a magnification of 5 times. The product was stored at 4 oC and 25 oC, respectively, and then did the concentration, encapsulate efficiency., particle size and appearance tests.
The results show that the samples must be stored at 4 oC, and the liposomes synthesized under the condition of 4: 1 phospholipid and cholesterol had the best coating effect (up to 92%) and the smallest particles Diameter (96.3 nm). The appearance is round orange red, which can also effectively prove that astaxanthin is coated in liposomes. In the scaled-up test, the product was similar with that at the small scale. The method developed in this study has commercial potential.
關鍵字(中) ★ 蝦紅素
★ 微脂體
★ 包覆
★ 薄膜水合法
關鍵字(英) ★ Astaxanthin
★ liposome
★ encapsulated
★ thin-film hydration method
論文目次 中文摘要 i
Abstract ii
Table of Content iv
List of Table vi
List of Figures vii
Chapter 1 Introduction 1
1.1 Background 1
1.2 Market Introduction 2
1.3 Introduction of Astaxanthin 5
1.4 Research motivation and purpose 6
Chapter 2 Literature Review 8
2.1 Astaxanthin 8
2.1.1 Characteristics of Astaxanthin 8
2.1.2 Production of astaxanthin 9
2.1.3 Antioxidant capacity and application of astaxanthin 11
2.1.4 Safety of Astaxanthin 13
2.2 Liposome technology 14
2.2.1 introduction of liposome 14
2.2.2 Composition of liposomes 18
2.2.3 Methods of liposome preparation 22
Chapter 3 Experimental 26
3.1 Material 26
3.2 Preparation of astaxanthin-loaded liposome solution 26
3.3 Characterization of Astaxanthin-loaded liposome 27
3.3.1 UV-Visible Spectrophotometer 27
3.3.2 Dynamic Light Scattering (DLS) 28
3.3.3 Optical microscopy (OM) 29
Chapter 4 Results and Discussion 31
4.1 Introduction 31
4.2 Effects of phosphatidylcholine /cholesterol on the encapsulation rat 32
4.2.1 Effect on Phosphatidylcholine 33
4.2.2 Effect on cholesterol 35
4.2.3 Effect on polysorbate 80 37
4.2.4 Effect on solvent 39
4.3 particle size of liposome 40
4.4 Appearance study 42
4.5 Scale up test 42
4.5.1 Encapsulation efficiency 43
4.5.2 particle size 44
4.5.3 Appearance study 46
4.6 Effects on homogenizer 46
Chapter 5 Conclusion 49
Reference 51
參考文獻 Alejandra A.M.D, Sanghamitra K, Josefina V.R, “Chemical stability of astaxanthin integrated into a food matrix: Effects of food processing and methods for preservation”, Food Chemistry. 225 (2017) 23-30
Akbarzadeh A, Sadabady R.R, Davaran S, Joo S.W, Zarghami N, Hanifehpour Y, Samiei M, Kouhi M, Koshki K.N, “Liposome: Classification, Preparation, and Applications”, Nanoscale Res Lett. 8 (2013) 120-129
Anantita S, Maruj L, Dalad S, Wanwimol K, “Properties and bioavailability assessment of shrimp astaxanthin loaded liposomes”, Food Science and Biotechnology. 28 (2018) 529-537
Bhatt P.C, Ahmad M, Panda B.P. “Enhanced bioaccumulation of astaxanthin in Phaffia rhodozyma by utilising low-cost agro products as fermentation substrate”, Biocatal. Agric. Biotechnol. 2 (2013) 58-63
Ciapara I.H, Valenzuela L.F, Goycoolea F.M, “Astaxanthin: A Review of Its Chemistry and Applications”, Crit Rev Food Sci Nutr. 46 (2006) 185-196
Chen X.L, Chen R, Guo Z.Y, Li C, Li P.C, “The preparation and stability of the inclusion complex of astaxanthin with β-cyclodextrin”, Food Chemistry. 101 (2007) 1580-1584
Drummond D.C., Noble C.O, Hayes M.E, Park J.W, Kirpotin D.B, “Pharmacokinetics and in vivo drug release rates in liposomal nanocarrier development”, J. Pharm. Sci. ,97 (2008) 4696 – 4740

Eiji Y, “Astaxanthin as a Medical Food”, Functional Foods in Health and Disease. 3 (2013) 254-258
Fardin T, Mohammad S, Jaleh V, Ali N, “Design and characterization of astaxanthin-loaded nanostructured lipid carriers”, Innovative Food Science & Emerging Technologies. 26 (2014) 366-374
Fardin T, Mohammad S, Jaleh V, Ali N, “Stability of astaxanthin-loaded nanostructured lipid carriers as affected by pH, ionic strength, heat treatment, simulated gastric juice and freeze–thawing”, Journal of Food Science and Technology. 54 (2017) 3132-3141
Garza C.B, Barragán B.E, “Thermal and pH stability of spray-dried encapsulated astaxanthin oleoresin from Haematococcus pluvialis using several encapsulation wall materials”, Food Res. Int. 54 (2013) 641-649
Griffin W.C., “Classification of Surface-Active Agents by HLB”, Journal of the Society of Cosmetic Chemists. 1 (1949) 311-326
Goto S, Kogure K, Abe K, Kimata Y, Kitahama K, Yamashita E, Terada H, “Efficient Radical Trapping at the Surface and Inside the Phospholipid Membrane Is Responsible for Highly Potent Antiperoxidative Activity of the Carotenoid Astaxanthin”, Biochim Biophys Acta. 1512 (2001) 251-258
Gouveia L, Empis J, “Relative stabilities of microalgal carotenoids in microalgal extracts, biomass and fish feed: effect of storage conditions”, Innovative Food Science & Emerging Technologies. 4 (2003) 227-233
Higueraciapara I, Félixvalenzuela L, Goycoolea F.M, “Astaxanthin: A review of its chemistry and applications”, Crit. Rev. Food Sci. Nutr, 46 (2006) 185-196
Jeong J.S, Kin I.H, “Effect of astaxanthin produced by Phaffia rhodozyma on growth performance, meat quality, and fecal noxious gas emission in broilers”, Poultry Science 93 (2014) 3138-3144
Mccormack B, Gregoriadis G, “Drugs in cyclodextrins in liposomes: a novel concept in drug delivery”, Int. J. Pharm. 112 (1994) 249 –258
Miaomiao L, Mohamed R.Z, Qipeng Y, Feibao T, Hao L, “Preparation and stability of astaxanthin solid lipid nanoparticles based on stearic acid”, Eu journal of lipid science and technology. 118 (2016) 592-602
Park J.S, Chyun J.H, Kim T.K, Line L.L, Chew B.P, “Astaxanthin decreased oxidative stress and inflammation and enhanced immune response in humans”, Nutr. Metab. 7 (2010) 7-18
Pan L, Zhang S, Gu K, Zhang N, “Preparation of astaxanthin-loaded liposomes: characterization, storage stability and antioxidant activity” CyTA - Journal of Food. 16 (2018) 607-618
Raffy S, Teissie J, “Control of Lipid Membrane Stability by Cholesterol Content”, Biophys. J. 76 (1999) 2072-2080
Rodríguez I, Lee H.K, Sam F. Y., “Microchannel electrophoretic separation of biogenic amines by micellar electrokinetic chromatography”, Electrophoresis. 20 (1999) 118-126
Satoh A, Tsuji S, Okada Y, Murakami N, Urami M, Nakagawa K, Ishikura M, Katagiri M, Koga Y, Shirasawa T, “Preliminary Clinical Evaluation of Toxicity and Efficacy of A New Astaxanthin-rich Haematococcus pluvialis Extract”, Journal of Clinical Biochemistry and Nutrition. 44 (2009) 280-284
Sophie R, Justin T, “Control of Lipid Membrane Stability by Cholesterol Content”, Biophysical journal. 76 (1999) 2072-2080
Tolasa S, Cakli S, Ostermeyer U, “Determination of astaxanthin and canthaxanthin in salmonid”, European Food Research and Technology. 221 (2005) 787-791
Yuan J.P, Peng J, Yin K, Wang J.H, “Potential health‐promoting effects of astaxanthin: A high‐value carotenoid mostly from microalgae”, Molecular of nutrition food research. 55 (2011) 150-165
指導教授 陳郁文 審核日期 2020-7-8
推文 facebook   plurk   twitter   funp   google   live   udn   HD   myshare   reddit   netvibes   friend   youpush   delicious   baidu   
網路書籤 Google bookmarks   del.icio.us   hemidemi   myshare   

若有論文相關問題,請聯絡國立中央大學圖書館推廣服務組 TEL:(03)422-7151轉57407,或E-mail聯絡  - 隱私權政策聲明