Derleme
BibTex RIS Kaynak Göster

Role of Apolipoproteins in Neurodegenerative Diseases

Yıl 2023, Cilt: 5 Sayı: 3, 122 - 133, 31.12.2023
https://doi.org/10.59124/guhes.1383395

Öz

Since lipids are insoluble in water, they are carried in the blood as particles called lipoproteins. Lipoproteins consisting of lipids and proteins are multicomponent complexes. The classification of lipoproteins, which are divided into several main groups such as low density lipoprotein (LDL), high density lipoprotein (HDL), very low density lipoprotein (VLDL) and chylomicrons, is based on their density, size, lipid and apolipoprotein content. Apolipoproteins are the protein component of lipoproteins that carry lipids from the blood to various tissues of the body for metabolism and utilisation. Apolipoproteins play an important role in lipid metabolism. They regulate many metabolic enzymes and interact with lipoprotein receptors. Numerous studies have shown that apolipoprotein phenotype, different allelic polymorphism and apolipoprotein gene mutation can affect metabolism and utilisation of blood lipids and consequently trigger the onset and development of atherosclerosis, hyperlipidaemia, cerebrovascular and cardiovascular diseases. Furthermore, apolipoproteins have been associated with neurodegenerative diseases and different apolipoprotein polymorphisms have been evaluated as risk factors or protective agents in different neurodegenerative diseases. This review presents evidence from some studies linking apolipoproteins with Alzheimer's disease (AD), Parkinson's disease (PD), multiple sclerosis (MS), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS) and frontotemporal degeneration disease.

Etik Beyan

-

Destekleyen Kurum

-

Proje Numarası

-

Teşekkür

-

Kaynakça

  • Aarsland, D., Andersen, K., Larsen, J. P., & Lolk, A. (2003). Prevalence and characteristics of dementia in Parkinson disease: an 8-year prospective study. Archives of neurology, 60(3), 387-392. https://doi.org/10.1001/archneur.60.3.387
  • Adunsky, A., Chesnin, V., Davidson, M., Gerber, Y., Alexander, K., & Haratz, D. (2002). A cross-sectional study of lipids and ApoC levels in Alzheimer's patients with and without cardiovascular disease. The Journals of Gerontology Series A: Biological Sciences and Medical Sciences, 57(11), M757-M761. https://doi.org/10.1093/gerona/57.11.m757
  • Antonios, N., Angiolillo, D. J., & Silliman, S. (2008). Hypertriglyceridemia and ischemic stroke. European neurology, 60(6), 269-278. https://doi.org/10.1159/000157880
  • Bahrami, A., Barreto, G. E., Lombardi, G., Pirro, M., & Sahebkar, A. (2019). Emerging roles for high‐density lipoproteins in neurodegenerative disorders. BioFactors, 45(5), 725-739. https://doi.org/10.1002/biof.1541
  • Balazs, Z., Panzenboeck, U., Hammer, A., Sovic, A., Quehenberger, O., Malle, E., & Sattler, W. (2004). Uptake and transport of high‐density lipoprotein (HDL) and HDL‐associated α‐tocopherol by an in vitro blood–brain barrier model. Journal of neurochemistry, 89(4), 939-950. https://doi.org/10.1111/j.1471-4159.2004.02373.x
  • Bergt, C., Nakano, T., Ditterich, J., DeCarli, C., & Eiserich, J. P. (2006). Oxidized plasma high-density lipoprotein is decreased in Alzheimer's disease. Free Radical Biology and Medicine, 41(10), 1542-1547. https://doi.org/10.1016/j.freeradbiomed.2006.08.019
  • Björkhem, I., & Meaney, S. (2004). Brain cholesterol: long secret life behind a barrier. Arteriosclerosis, thrombosis, and vascular biology, 24(5), 806-815. https://doi.org/10.1161/01.ATV.0000120374.59826.1b.
  • Brown, M. S., & Goldstein, J. L. (1986). A receptor-mediated pathway for cholesterol homeostasis. Science, 232(4746), 34-47. https://doi.org/10.1126/science.3513311
  • Browne, R. W., Weinstock-Guttman, B., Horakova, D., Zivadinov, R., Bodziak, M. L., Tamaño-Blanco, M., Badgett, D., Tyblova, M., Vaneckova, M., Seidl, Z., Krasensky, J., Bergsland, N., Ramasamy, D. P., Hagemeier, J., Havrdova, E., & Ramanathan, M. (2014). Apolipoproteins are associated with new MRI lesions and deep grey matter atrophy in clinically isolated syndromes. Journal of Neurology, Neurosurgery & Psychiatry, 85(8), 859-864. https://doi.org/10.1136/jnnp-2013-307106
  • Calero, M., Rostagno, A., Matsubara, E., Zlokovic, B., Frangione, B., & Ghiso, J. (2000). Apolipoprotein J (clusterin) and Alzheimer's disease. Microscopy research and technique, 50(4), 305-315. https://doi.org/10.1002/1097-0029(20000815)50:4<305::AID-JEMT10>3.0.CO;2-L
  • Chang, K. H., Cheng, M. L., Lo, C. J., Fan, C. M., Wu, Y. R., & Chen, C. M. (2023). Alternations of Lipoprotein Profiles in the Plasma as Biomarkers of Huntington’s Disease. Cells, 12(3), 385. https://doi.org/10.3390/cells12030385
  • Choi, H. J., Byun, M. S., Yi, D., Choe, Y. M., Sohn, B. K., Baek, H. W., Lee, J. H., Kim, H. J., Han, J. Y., Yoon, E. J., Kim, Y. K., Woo, J. I., & Lee, D. Y. (2016). Association between serum triglycerides and cerebral amyloidosis in cognitively normal elderly. The American Journal of Geriatric Psychiatry, 24(8), 604-612. https://doi.org/10.1016/j.jagp.2016.03.001
  • Dai, L., Li, S., Hao, Q., Zhou, R., Zhou, H., Lei, W., Kang, H., Wu, H., Li, Y., & Ma, X. (2023). Low-density lipoprotein: a versatile nanoscale platform for targeted delivery. Nanoscale Advances, 5, 1011-1022. https://doi.org/10.1039/d2na00883a
  • Dominiczak, M. H., & Caslake, M. J. (2011). Apolipoproteins: metabolic role and clinical biochemistry applications. Annals of clinical biochemistry, 48(6), 498-515. https://doi.org/10.1258/acb.2011.011111
  • Eiselein, L., Wilson, D. W., Lamé, M. W., & Rutledge, J. C. (2007). Lipolysis products from triglyceride-rich lipoproteins increase endothelial permeability, perturb zonula occludens-1 and F-actin, and induce apoptosis. American Journal of Physiology-Heart and Circulatory Physiology, 292(6), H2745-H2753. https://doi.org/10.1152/ajpheart.00686.2006
  • Fang, F., Zhan, Y., Hammar, N., Shen, X., Wirdefeldt, K., Walldius, G., & Mariosa, D. (2019). Lipids, Apolipoproteins, and the risk of Parkinson disease: a prospective cohort study and a Mendelian randomization analysis. Circulation Research, 125(6), 643-652. https://doi.org/10.1161/CIRCRESAHA.119.314929
  • Fanning, A. S., & Anderson, J. M. (2009). Zonula occludens‐1 and‐2 are cytosolic scaffolds that regulate the assembly of cellular junctions. Annals of the new York Academy of Sciences, 1165(1), 113-120. https://doi.org/10.1111/j.1749-6632.2009.04440.x
  • Farrer, L. A., Cupples, L. A., Haines, J. L., Hyman, B., Kukull, W. A., Mayeux, R., Myers, R. H., Pericak-Vance, M. A., Risch, N., & Van Duijn, C. M. (1997). Effects of age, sex, and ethnicity on the association between apolipoprotein E genotype and Alzheimer disease: a meta-analysis. Jama, 278(16), 1349-1356. https://doi.org/10.1001/jama.1997.03550160069041
  • Foster, E. M., Dangla-Valls, A., Lovestone, S., Ribe, E. M., & Buckley, N. J. (2019). Clusterin in Alzheimer’s disease: mechanisms, genetics, and lessons from other pathologies. Frontiers in neuroscience, 13, 164. https://doi.org/10.3389/fnins.2019.00164
  • Fyfe-Desmarais, G., Desmarais, F., Rassart, É., & Mounier, C. (2023). Apolipoprotein D in Oxidative Stress and Inflammation. Antioxidants, 12(5), 1027. https://doi.org/10.3390/antiox12051027
  • Gomperts, S. N., Locascio, J. J., Rentz, D., Santarlasci, A., Marquie, M., Johnson, K. A., & Growdon, J. H. (2013). Amyloid is linked to cognitive decline in patients with Parkinson disease without dementia. Neurology, 80(1), 85-91. https://doi.org/10.1212/WNL.0b013e31827b1a07
  • Hely, M. A., Reid, W. G., Adena, M. A., Halliday, G. M., & Morris, J. G. (2008). The Sydney multicenter study of Parkinson's disease: the inevitability of dementia at 20 years. Movement disorders, 23(6), 837-844. https://doi.org/10.1002/mds.21956
  • Hong, S., Prokopenko, D., Dobricic, V., Kilpert, F., Bos, I., Vos, S. J., Tijms, B. M., Andreasson, U., Blennow, K., Vandenberghe, R., Cleynen, I., Gabel, S., Schaeverbeke, J., Scheltens, P., Teunissen, C. E., Niemantsverdriet, E., Engelborghs, S., Frisoni, G., Blin, O., … & Bertram, L. (2020). Genome-wide association study of Alzheimer’s disease CSF biomarkers in the EMIF-AD Multimodal Biomarker Discovery dataset. Translational psychiatry, 10(1), 403. https://doi.org/10.1038/s41398-020-01074-z
  • Huang, X., Chen, P. C., & Poole, C. (2004). APOE-ε2 allele associated with higher prevalence of sporadic Parkinson disease. Neurology, 62(12), 2198-2202. https://doi.org/10.1212/01.WNL.0000130159.28215.6A
  • Huang, Y. C., Wu, Y. R., Tseng, M. Y., Chen, Y. C., Hsieh, S. Y., & Chen, C. M. (2011). Increased prothrombin, apolipoprotein A-IV, and haptoglobin in the cerebrospinal fluid of patients with Huntington's disease. PloS one, 6(1), e15809. https://doi.org/10.1371/journal.pone.0015809
  • Hughes, T. A., Ross, H. F., Musa, S., Bhattacherjee, S., Nathan, R. N., Mindham, R. H. S., & Spokes, E. G. S. (2000). A 10-year study of the incidence of and factors predicting dementia in Parkinson’s disease. Neurology, 54(8), 1596-1603. https://doi.org/10.1212/wnl.54.8.1596
  • Johansson, P., Almqvist, E. G., Bjerke, M., Wallin, A., Johansson, J. O., Andreasson, U., Blennow, K., Zetterberg, H., & Svensson, J. (2017). Reduced cerebrospinal fluid concentration of apolipoprotein AI in patients with Alzheimer’s disease. Journal of Alzheimer's Disease, 59(3), 1017-1026. https://doi.org/10.3233/JAD-170226
  • Kawano, M., Kawakami, M., Otsuka, M., Yashima, H., Yaginuma, T., & Ueki, A. (1995). Marked decrease of plasma apolipoprotein AI and AII in Japanese patients with late-onset non-familial Alzheimer's disease. Clinica chimica acta; international journal of clinical chemistry, 239(2), 209-211. https://doi.org/10.1016/0009-8981(95)06115-t
  • Kim, W. S., He, Y., Phan, K., Ahmed, R. M., Rye, K. A., Piguet, O., Hodges, J. R., & Halliday, G. M. (2018). Altered high density lipoprotein composition in behavioral variant frontotemporal dementia. Frontiers in neuroscience, 12, 847. https://doi.org/10.3389/fnins.2018.00847
  • Koizumi, K., Hattori, Y., Ahn, S. J., Buendia, I., Ciacciarelli, A., Uekawa, K., Wang, G., Hiller, A., Zhao, L., Voss, H. U., Paul, S. M., Schaffer, C., Park, L., & Iadecola, C. (2018). Apoε4 disrupts neurovascular regulation and undermines white matter integrity and cognitive function. Nature communications, 9(1), 3816. https://doi.org/10.1038/s41467-018-06301-2
  • Kuriyama, M., Takahashi, K., Yamano, T., Hokezu, Y., Togo, S., Osame, M., & Igakura, T. (1994). Low levels of serum apolipoprotein AI and AII in senile dementia. Psychiatry and Clinical Neurosciences, 48(3), 589-593. https://doi.org/10.1111/j.1440-1819.1994.tb03019.x
  • Leduc, M. S., Shimmin, L. C., Klos, K. L., Hanis, C., Boerwinkle, E., & Hixson, J. E. (2008). Comprehensive evaluation of apolipoprotein H gene (APOH) variation identifies novel associations with measures of lipid metabolism in GENOA* s⃞. Journal of lipid research, 49(12), 2648-2656. https://doi.org/10.1194/jlr.M800155-JLR200
  • Lehnert, S., Jesse, S., Rist, W., Steinacker, P., Soininen, H., Herukka, S. K., Tumani, H., Lenter, M., Oeckl, P., Ferger, B., Hengerer, B., & Otto, M. (2012). iTRAQ and multiple reaction monitoring as proteomic tools for biomarker search in cerebrospinal fluid of patients with Parkinson's disease dementia. Experimental neurology, 234(2), 499-505. https://doi.org/10.1016/j.expneurol.2012.01.024
  • Lenzi, C., Ramazzina, I., Russo, I., Filippini, A., Bettuzzi, S., & Rizzi, F. (2020). The down-regulation of clusterin expression enhances the αsynuclein aggregation process. International Journal of Molecular Sciences, 21(19), 7181. https://doi.org/10.3390/ijms21197181
  • Leoni, E., Bremang, M., Mitra, V., Zubiri, I., Jung, S., Lu, C. H., Adiutori, R., Lombardi, V., Russell, C., Koncarevic, S., Ward, M., Pike, I., & Malaspina, A. (2019). Combined tissue-fluid proteomics to unravel phenotypic variability in amyotrophic lateral sclerosis. Scientific Reports, 9(1), 4478. https://doi.org/10.1038/s41598-019-40632-4
  • Lin, Y., Lu, L., Zhou, M., Liu, H., Ye, P., Zhang, W., Qiu, J., Zhang, Z., Yang, X., Ding, L., Guo, W., Mo, M., Zhu, X., Zhang, X., Chen, X., & Xu, P. (2021). Association of CLU gene polymorphism with Parkinson's disease in the Chinese Han population. The Journal of Gene Medicine, 23(2), e3302. https://doi.org/10.1002/jgm.3302
  • Liu, H. C., Hu, C. J., Chang, J. G., Sung, S. M., Lee, L. S., Yuan, R. Y., & Leu, S. J. (2006). Proteomic identification of lower apolipoprotein AI in Alzheimer’s disease. Dementia and geriatric cognitive disorders, 21(3), 155-161. https://doi.org/10.1159/000090676
  • Liu, T., Chen, J. M., Zhang, D., Zhang, Q., Peng, B., Xu, L., & Tang, H. (2021). ApoPred: identification of apolipoproteins and their subfamilies with multifarious features. Frontiers in Cell and Developmental Biology, 8, 621144. https://doi.org/10.3389/fcell.2020.621144
  • Maarouf, C. L., Beach, T. G., Adler, C. H., Shill, H. A., Sabbagh, M. N., Wu, T., Walker, D. G., Kokjohn, T. A., Roher, A. E., & Consortium, A. P. (2012). Cerebrospinal fluid biomarkers of neuropathologically diagnosed Parkinson’s disease subjects. Neurological research, 34(7), 669-676. https://doi.org/10.1179/1743132812Y.0000000063
  • Mata , I. F., Leverenz, J. B., Weintraub, D., Trojanowski, J. Q., Hurtig, H. I., Van Deerlin, V. M., Ritz, B., Rausch, R., Rhodes, S. L., Factor, S. A., Wood-Siverio, C., Quinn, J. F., Chung, K. A., Peterson, A. L., Espay, A. J., Revilla, F. J., Devoto, J., Hu, S., Cholerton, B. A., Wan, J. Y., Montine, T. J., Edwards, K. L., & Zabetian, C. P. (2014). APOE, MAPT, and SNCA genes and cognitive performance in Parkinson disease. JAMA neurology, 71(11), 1405-1412. https://doi.org/10.1001/jamaneurol.2014.1455
  • Mathieu, C., Pappu, R. V., & Taylor, J. P. (2020). Beyond aggregation: Pathological phase transitions in neurodegenerative disease. Science, 370(6512), 56-60. https://doi.org/10.1126/science.abb8032
  • Merched , A., Xia, Y., Visvikis, S., Serot, J. M., & Siest, G. (2000). Decreased high-density lipoprotein cholesterol and serum apolipoprotein AI concentrations are highly correlated with the severity of Alzheimer’s disease☆. Neurobiology of aging, 21(1), 27-30. https://doi.org/10.1016/s0197-4580(99)00103-7
  • McComb, M., Parambi, R., Browne, R. W., Bodziak, M. L., Jakimovski, D., Bergsland, N., Maceski, A., Weinstock-Guttman, B., Jens Kuhle, J., Zivadinov, R., & Ramanathan, M. (2020). Apolipoproteins AI and E are associated with neuroaxonal injury to gray matter in multiple sclerosis. Multiple sclerosis and related disorders, 45, 102389. https://doi.org/10.1016/j.msard.2020.102389
  • Misra, A., Chakrabarti, S. S., Gambhir, I. S., Baghel, M. S., & Patil, Y. R. (2021). Tandem mass spectrometry-based identification of protein profiles in the cerebrospinal fluid of Alzheimer’s dementia patients. https://doi.org/10.21203/rs.3.rs-574644/v1
  • Muenchhoff, J., Song, F., Poljak, A., Crawford, J. D., Mather, K. A., Kochan, N. A., Yang, Z., Trollor, J. N., Reppermund, S., Maston, K., Theobald, A., Kirchner-Adelhardt, S., Kwok, J. B., Richmond, R. L., McEvoy, M., Attia, J., Schofield, P. W., Brodaty, H., & Sachdev, P. S. (2017). Plasma apolipoproteins and physical and cognitive health in very old individuals. Neurobiology of Aging, 55, 49-60. https://doi.org/10.1016/j.neurobiolaging.2017.02.017
  • Murali, N., Browne, R. W., Fellows Maxwell, K., Bodziak, M. L., Jakimovski, D., Hagemeier, J., Bergsland, N., Weinstock-Guttman, B., Zivadinov, R., & Ramanathan, M. (2020). Cholesterol and neurodegeneration: longitudinal changes in serum cholesterol biomarkers are associated with new lesions and gray matter atrophy in multiple sclerosis over 5 years of follow‐up. European Journal of Neurology, 27(1), 188-e4. https://doi.org/10.1111/ene.14055
  • Nakaya, Y., Schaefer, E. J., & Brewer Jr, H. B. (1980). Activation of human post heparin lipoprotein lipase by apolipoprotein H (β2-glycoprotein I). Biochemical and biophysical research communications, 95(3), 1168-1172. https://doi.org/10.1016/0006-291X(80)91595-8
  • Namba, Y., Tsuchiya, H., & Ikeda, K. (1992). Apolipoprotein B immunoreactivity in senile plaque and vascular amyloids and neurofibrillary tangles in the brains of patients with Alzheimer's disease. Neuroscience letters, 134(2), 264-266. https://doi.org/10.1016/0304-3940(92)90531-b
  • Navarro, A., Rioseras, B., Del Valle, E., Martínez-Pinilla, E., Astudillo, A., & Tolivia, J. (2018). Expression pattern of myelin-related apolipoprotein D in human multiple sclerosis lesions. Frontiers in Aging Neuroscience, 10, 254. https://doi.org/10.3389/fnagi.2018.00254
  • Öhrfelt, A., Andreasson, U., Simon, A., Zetterberg, H., Edman, Å., Potter, W., Holder, D., Devanarayan, V., Seeburger, J., Smith, A. D., Blennow, K., & Wallin, A. (2011). Screening for new biomarkers for subcortical vascular dementia and Alzheimer’s disease. Dementia and geriatric cognitive disorders extra, 1(1), 31-42. https://doi.org/10.1159/000323417
  • Park, J. H., Hong, K. S., Lee, E. J., Lee, J., & Kim, D. E. (2011). High levels of apolipoprotein B/AI ratio are associated with intracranial atherosclerotic stenosis. Stroke, 42(11), 3040-3046. https://doi.org/10.1161/STROKEAHA.111.620104
  • Paula-Lima, A. C., Tricerri, M. A., Brito-Moreira, J., Bomfim, T. R., Oliveira, F. F., Magdesian, M. H., Grinberg, L. T., Panizzutti, R., & Ferreira, S. T. (2009). Human apolipoprotein A–I binds amyloid-β and prevents Aβ-induced neurotoxicity. The international journal of biochemistry & cell biology, 41(6), 1361-1370. https://doi.org/10.1016/j.biocel.2008.12.003
  • Přikrylová Vranová, H., Mareš, J., Nevrlý, M., Stejskal, D., Zapletalová, J., Hluštík, P., & Kaňovský, P. (2010). CSF markers of neurodegeneration in Parkinson’s disease. Journal of neural transmission, 117, 1177-1181. https://doi.org/10.1007/s00702-010-0462-z
  • Ramasamy, I. (2014). Recent advances in physiological lipoprotein metabolism. Clinical Chemistry and Laboratory Medicine (CCLM), 52(12), 1695-1727. https://doi.org/10.1515/cclm-2013-0358
  • Rassart, E., Bedirian, A., Do Carmo, S., Guinard, O., Sirois, J., Terrisse, L., & Milne, R. (2000). Apolipoprotein d. Biochimica et Biophysica Acta (BBA)-Protein Structure and Molecular Enzymology, 1482(1-2), 185-198. https://doi.org/10.1016/S0167-4838(00)00162-X
  • Real, R., Martinez-Carrasco, A., Reynolds, R. H., Lawton, M. A., Tan, M. M., Shoai, M., Corvol, J. C., Ryten, M., Bresner, C., Hubbard, L., Brice, A., Lesage, S., Faouzi, J., Elbaz, A., Artaud, F., Williams, N., Hu, M. T. M., Ben-Shlomo, Y., Grosset, D. G., ... & Morris, H. R. (2023). Association between the LRP1B and APOE loci and the development of Parkinson’s disease dementia. Brain, 146(5), 1873-1887. https://doi.org/10.1093/brain/awac414
  • Reichert, C. O., de Freitas, F. A., Sampaio-Silva, J., Rokita-Rosa, L., Barros, P. D. L., Levy, D., & Bydlowski, S. P. (2020a). Ferroptosis mechanisms involved in neurodegenerative diseases. International journal of molecular sciences, 21(22), 8765. https://doi.org/10.3390/ijms21228765
  • Reichert, C. O., Levy, D., & Bydlowski, S. P. (2020b). Paraoxonase role in human neurodegenerative diseases. Antioxidants, 10(1), 11. https://doi.org/10.3390/antiox10010011
  • Reindl, M., Knipping, G., Wicher, I., Dilitz, E., Egg, R., Deisenhammer, F., & Berger, T. (2001). Increased intrathecal production of apolipoprotein D in multiple sclerosis. Journal of neuroimmunology, 119(2), 327-332. https://doi.org/10.1016/s0165-5728(01)00378-2
  • Richardson, T. G., Sanderson, E., Palmer, T. M., Ala-Korpela, M., Ference, B. A., Davey Smith, G., & Holmes, M. V. (2020). Evaluating the relationship between circulating lipoprotein lipids and apolipoproteins with risk of coronary heart disease: A multivariable Mendelian randomisation analysis. PLoS medicine, 17(3), e1003062. https://doi.org/10.1371/journal.pmed.1003062
  • Ross MH, & Pawlina W. (2011). Histology: A text and atlas with correlated cell and molecular biology. Baltimore, Philadelphia: Lippincott Williams & Wilkins.
  • Saczynski, J. S., White, L., Peila, R. L., Rodriguez, B. L., & Launer, L. J. (2007). The relation between apolipoprotein AI and dementia: the Honolulu-Asia aging study. American journal of epidemiology, 165(9), 985-992. https://doi.org/10.1093/aje/kwm027
  • Sasaki , K., Doh-ura, K., Wakisaka, Y., & Iwaki, T. (2002). Clusterin/apolipoprotein J is associated with cortical Lewy bodies: immunohistochemical study in cases with α-synucleinopathies. Acta neuropathologica, 104, 225-230. https://doi.org/10.1007/s00401-002-0546-4
  • Shih, Y. H., Tsai, K. J., Lee, C. W., Shiesh, S. C., Chen, W. T., Pai, M. C., & Kuo, Y. M. (2014). Apolipoprotein C-III is an amyloid-β-binding protein and an early marker for Alzheimer's disease. Journal of Alzheimer's Disease, 41(3), 855-865. https://doi.org/10.3233/JAD-140111
  • Su, W. H., Shi, Z. H., Liu, S. L., Wang, X. D., Liu, S., & Ji, Y. (2017). Updated meta-analysis of the role of APOE ε2/ε3/ε4 alleles in frontotemporal lobar degeneration. Oncotarget, 8(27), 43721. https://doi.org/10.18632/oncotarget.17341
  • Sweeney, M. D., Sagare, A. P., & Zlokovic, B. V. (2018). Blood–brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nature Reviews Neurology, 14(3), 133-150. https://doi.org/10.1038/nrneurol.2017.188
  • Thompson, A. G., Talbot, K., & Turner, M. R. (2022). Higher blood high density lipoprotein and apolipoprotein A1 levels are associated with reduced risk of developing amyotrophic lateral sclerosis. Journal of Neurology, Neurosurgery & Psychiatry, 93(1), 75-81. https://doi.org/10.1136/jnnp-2021-327133
  • Tóth, M. E., Dukay, B., Hoyk, Z., & Sántha, M. (2020). Cerebrovascular changes and neurodegeneration related to hyperlipidemia: characteristics of the human ApoB-100 transgenic mice. Current Pharmaceutical Design, 26(13), 1486-1494. https://doi.org/10.2174/1381612826666200218101818
  • Tsuang, D., Leverenz, J. B., Lopez, O. L., Hamilton, R. L., Bennett, D. A., Schneider, J. A., Buchman, A. S., Larson, E. B., Crane, P. K., Kaye, J. A., Kramer, P., Woltjer, R., Trojanowski, J. O., Weintraub, D., Chen-Plotkin, A. S., Irwin, D. J., Rick, J., Schellenberg, G. D., Watson, G. S., … & Zabetian, C. P. (2013). APOE ϵ4 increases risk for dementia in pure synucleinopathies. JAMA neurology, 70(2), 223-228. https://doi.org/10.1001/jamaneurol.2013.600
  • Turan, T. N., Makki, A. A., Tsappidi, S., Cotsonis, G., Lynn, M. J., Cloft, H. J., & Chimowitz, M. I. (2010). Risk factors associated with severity and location of intracranial arterial stenosis. Stroke, 41(8), 1636-1640. https://doi.org/10.1161/STROKEAHA.110.584672
  • van Luijn, M. M., van Meurs, M., Stoop, M. P., Verbraak, E., Wierenga-Wolf, A. F., Melief, M. J., Kreft, K. L., Verdijk, R. M., ’t Hart, B. A., Luider, T. M., Laman, J. D., & Hintzen, R. Q. (2016). Elevated expression of the cerebrospinal fluid disease markers chromogranin a and clusterin in astrocytes of multiple sclerosis white matter lesions. Journal of Neuropathology & Experimental Neurology, 75(1), 86-98. https://doi.org/10.1093/jnen/nlv004
  • Vitali, C., Wellington, C. L., & Calabresi, L. (2014). HDL and cholesterol handling in the brain. Cardiovascular research, 103(3), 405-413. https://doi.org/10.1093/cvr/cvu148
  • Waldner, A., Dassati, S., Redl, B., Smania, N., & Gandolfi, M. (2018). Apolipoprotein D concentration in human plasma during aging and in Parkinson’s disease: a cross-sectional study. Parkinson’s Disease, 2018. https://doi.org/10.1155/2018/3751516
  • Wang, L., Sapuri-Butti, A. R., Aung, H. H., Parikh, A. N., & Rutledge, J. C. (2008). Triglyceride-rich lipoprotein lipolysis increases aggregation of endothelial cell membrane microdomains and produces reactive oxygen species. American Journal of Physiology-Heart and Circulatory Physiology, 295(1), H237-H244. https://doi.org/10.1152/ajpheart.01366.2007
  • Wei, Q., Wang, H., Tian, Y., Xu, F., Chen, X., & Wang, K. (2013). Reduced serum levels of triglyceride, very low density lipoprotein cholesterol and apolipoprotein B in Parkinson’s disease patients. PLoS One, 8(9), e75743. https://doi.org/10.1371/journal.pone.0075743
  • Weinstock-Guttman, B., Zivadinov, R., Mahfooz, N., Carl, E., Drake, A., Schneider, J., Teter, B., Hussein, S., Mehta, B., Weiskopf, M., Durfee, J., Bergsland, N., & Ramanathan, M. (2011). Serum lipid profiles are associated with disability and MRI outcomes in multiple sclerosis. Journal of neuroinflammation, 8(1), 1-7. https://doi.org/10.1186/1742-2094-8-127
  • Wisniewski, T., Golabek, A. A., Kida, E., Wisniewski, K. E., & Frangione, B. (1995). Conformational mimicry in Alzheimer's disease. Role of apolipoproteins in amyloidogenesis. The American journal of pathology, 147(2), 238. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1869828/pdf/amjpathol00044-0015.pdf
  • Zhang, X., & Alzheimer’s Disease Neuroimaging Initiative. (2020). Higher plasma APOC-III was associated with a slower reduction of β-amyloid levels in cerebrospinal fluid among older individuals without dementia. Neuropsychiatric Disease and Treatment, 1139-1144. https://doi.org/10.2147/NDT.S238985
  • Zhang, X., Yin, X., Yu, H., Liu, X., Yang, F., Yao, J., Jin, H., & Yang, P. (2012). Quantitative proteomic analysis of serum proteins in patients with Parkinson’s disease using an isobaric tag for relative and absolute quantification labeling, two-dimensional liquid chromatography, and tandem mass spectrometry. Analyst, 137(2), 490-495. https://doi.org/10.1039/c1an15551b
  • Zlokovic, B. V., Martel, C. L., Matsubara, E., McComb, J. G., Zheng, G., McCluskey, R. T., Frangione, B., & Ghiso, J. (1996). Glycoprotein 330/megalin: probable role in receptor-mediated transport of apolipoprotein J alone and in a complex with Alzheimer disease amyloid beta at the blood-brain and blood-cerebrospinal fluid barriers. Proceedings of the National Academy of Sciences, 93(9), 4229-4234. https://doi.org/10.1073/pnas.93.9.4229
Yıl 2023, Cilt: 5 Sayı: 3, 122 - 133, 31.12.2023
https://doi.org/10.59124/guhes.1383395

Öz

Proje Numarası

-

Kaynakça

  • Aarsland, D., Andersen, K., Larsen, J. P., & Lolk, A. (2003). Prevalence and characteristics of dementia in Parkinson disease: an 8-year prospective study. Archives of neurology, 60(3), 387-392. https://doi.org/10.1001/archneur.60.3.387
  • Adunsky, A., Chesnin, V., Davidson, M., Gerber, Y., Alexander, K., & Haratz, D. (2002). A cross-sectional study of lipids and ApoC levels in Alzheimer's patients with and without cardiovascular disease. The Journals of Gerontology Series A: Biological Sciences and Medical Sciences, 57(11), M757-M761. https://doi.org/10.1093/gerona/57.11.m757
  • Antonios, N., Angiolillo, D. J., & Silliman, S. (2008). Hypertriglyceridemia and ischemic stroke. European neurology, 60(6), 269-278. https://doi.org/10.1159/000157880
  • Bahrami, A., Barreto, G. E., Lombardi, G., Pirro, M., & Sahebkar, A. (2019). Emerging roles for high‐density lipoproteins in neurodegenerative disorders. BioFactors, 45(5), 725-739. https://doi.org/10.1002/biof.1541
  • Balazs, Z., Panzenboeck, U., Hammer, A., Sovic, A., Quehenberger, O., Malle, E., & Sattler, W. (2004). Uptake and transport of high‐density lipoprotein (HDL) and HDL‐associated α‐tocopherol by an in vitro blood–brain barrier model. Journal of neurochemistry, 89(4), 939-950. https://doi.org/10.1111/j.1471-4159.2004.02373.x
  • Bergt, C., Nakano, T., Ditterich, J., DeCarli, C., & Eiserich, J. P. (2006). Oxidized plasma high-density lipoprotein is decreased in Alzheimer's disease. Free Radical Biology and Medicine, 41(10), 1542-1547. https://doi.org/10.1016/j.freeradbiomed.2006.08.019
  • Björkhem, I., & Meaney, S. (2004). Brain cholesterol: long secret life behind a barrier. Arteriosclerosis, thrombosis, and vascular biology, 24(5), 806-815. https://doi.org/10.1161/01.ATV.0000120374.59826.1b.
  • Brown, M. S., & Goldstein, J. L. (1986). A receptor-mediated pathway for cholesterol homeostasis. Science, 232(4746), 34-47. https://doi.org/10.1126/science.3513311
  • Browne, R. W., Weinstock-Guttman, B., Horakova, D., Zivadinov, R., Bodziak, M. L., Tamaño-Blanco, M., Badgett, D., Tyblova, M., Vaneckova, M., Seidl, Z., Krasensky, J., Bergsland, N., Ramasamy, D. P., Hagemeier, J., Havrdova, E., & Ramanathan, M. (2014). Apolipoproteins are associated with new MRI lesions and deep grey matter atrophy in clinically isolated syndromes. Journal of Neurology, Neurosurgery & Psychiatry, 85(8), 859-864. https://doi.org/10.1136/jnnp-2013-307106
  • Calero, M., Rostagno, A., Matsubara, E., Zlokovic, B., Frangione, B., & Ghiso, J. (2000). Apolipoprotein J (clusterin) and Alzheimer's disease. Microscopy research and technique, 50(4), 305-315. https://doi.org/10.1002/1097-0029(20000815)50:4<305::AID-JEMT10>3.0.CO;2-L
  • Chang, K. H., Cheng, M. L., Lo, C. J., Fan, C. M., Wu, Y. R., & Chen, C. M. (2023). Alternations of Lipoprotein Profiles in the Plasma as Biomarkers of Huntington’s Disease. Cells, 12(3), 385. https://doi.org/10.3390/cells12030385
  • Choi, H. J., Byun, M. S., Yi, D., Choe, Y. M., Sohn, B. K., Baek, H. W., Lee, J. H., Kim, H. J., Han, J. Y., Yoon, E. J., Kim, Y. K., Woo, J. I., & Lee, D. Y. (2016). Association between serum triglycerides and cerebral amyloidosis in cognitively normal elderly. The American Journal of Geriatric Psychiatry, 24(8), 604-612. https://doi.org/10.1016/j.jagp.2016.03.001
  • Dai, L., Li, S., Hao, Q., Zhou, R., Zhou, H., Lei, W., Kang, H., Wu, H., Li, Y., & Ma, X. (2023). Low-density lipoprotein: a versatile nanoscale platform for targeted delivery. Nanoscale Advances, 5, 1011-1022. https://doi.org/10.1039/d2na00883a
  • Dominiczak, M. H., & Caslake, M. J. (2011). Apolipoproteins: metabolic role and clinical biochemistry applications. Annals of clinical biochemistry, 48(6), 498-515. https://doi.org/10.1258/acb.2011.011111
  • Eiselein, L., Wilson, D. W., Lamé, M. W., & Rutledge, J. C. (2007). Lipolysis products from triglyceride-rich lipoproteins increase endothelial permeability, perturb zonula occludens-1 and F-actin, and induce apoptosis. American Journal of Physiology-Heart and Circulatory Physiology, 292(6), H2745-H2753. https://doi.org/10.1152/ajpheart.00686.2006
  • Fang, F., Zhan, Y., Hammar, N., Shen, X., Wirdefeldt, K., Walldius, G., & Mariosa, D. (2019). Lipids, Apolipoproteins, and the risk of Parkinson disease: a prospective cohort study and a Mendelian randomization analysis. Circulation Research, 125(6), 643-652. https://doi.org/10.1161/CIRCRESAHA.119.314929
  • Fanning, A. S., & Anderson, J. M. (2009). Zonula occludens‐1 and‐2 are cytosolic scaffolds that regulate the assembly of cellular junctions. Annals of the new York Academy of Sciences, 1165(1), 113-120. https://doi.org/10.1111/j.1749-6632.2009.04440.x
  • Farrer, L. A., Cupples, L. A., Haines, J. L., Hyman, B., Kukull, W. A., Mayeux, R., Myers, R. H., Pericak-Vance, M. A., Risch, N., & Van Duijn, C. M. (1997). Effects of age, sex, and ethnicity on the association between apolipoprotein E genotype and Alzheimer disease: a meta-analysis. Jama, 278(16), 1349-1356. https://doi.org/10.1001/jama.1997.03550160069041
  • Foster, E. M., Dangla-Valls, A., Lovestone, S., Ribe, E. M., & Buckley, N. J. (2019). Clusterin in Alzheimer’s disease: mechanisms, genetics, and lessons from other pathologies. Frontiers in neuroscience, 13, 164. https://doi.org/10.3389/fnins.2019.00164
  • Fyfe-Desmarais, G., Desmarais, F., Rassart, É., & Mounier, C. (2023). Apolipoprotein D in Oxidative Stress and Inflammation. Antioxidants, 12(5), 1027. https://doi.org/10.3390/antiox12051027
  • Gomperts, S. N., Locascio, J. J., Rentz, D., Santarlasci, A., Marquie, M., Johnson, K. A., & Growdon, J. H. (2013). Amyloid is linked to cognitive decline in patients with Parkinson disease without dementia. Neurology, 80(1), 85-91. https://doi.org/10.1212/WNL.0b013e31827b1a07
  • Hely, M. A., Reid, W. G., Adena, M. A., Halliday, G. M., & Morris, J. G. (2008). The Sydney multicenter study of Parkinson's disease: the inevitability of dementia at 20 years. Movement disorders, 23(6), 837-844. https://doi.org/10.1002/mds.21956
  • Hong, S., Prokopenko, D., Dobricic, V., Kilpert, F., Bos, I., Vos, S. J., Tijms, B. M., Andreasson, U., Blennow, K., Vandenberghe, R., Cleynen, I., Gabel, S., Schaeverbeke, J., Scheltens, P., Teunissen, C. E., Niemantsverdriet, E., Engelborghs, S., Frisoni, G., Blin, O., … & Bertram, L. (2020). Genome-wide association study of Alzheimer’s disease CSF biomarkers in the EMIF-AD Multimodal Biomarker Discovery dataset. Translational psychiatry, 10(1), 403. https://doi.org/10.1038/s41398-020-01074-z
  • Huang, X., Chen, P. C., & Poole, C. (2004). APOE-ε2 allele associated with higher prevalence of sporadic Parkinson disease. Neurology, 62(12), 2198-2202. https://doi.org/10.1212/01.WNL.0000130159.28215.6A
  • Huang, Y. C., Wu, Y. R., Tseng, M. Y., Chen, Y. C., Hsieh, S. Y., & Chen, C. M. (2011). Increased prothrombin, apolipoprotein A-IV, and haptoglobin in the cerebrospinal fluid of patients with Huntington's disease. PloS one, 6(1), e15809. https://doi.org/10.1371/journal.pone.0015809
  • Hughes, T. A., Ross, H. F., Musa, S., Bhattacherjee, S., Nathan, R. N., Mindham, R. H. S., & Spokes, E. G. S. (2000). A 10-year study of the incidence of and factors predicting dementia in Parkinson’s disease. Neurology, 54(8), 1596-1603. https://doi.org/10.1212/wnl.54.8.1596
  • Johansson, P., Almqvist, E. G., Bjerke, M., Wallin, A., Johansson, J. O., Andreasson, U., Blennow, K., Zetterberg, H., & Svensson, J. (2017). Reduced cerebrospinal fluid concentration of apolipoprotein AI in patients with Alzheimer’s disease. Journal of Alzheimer's Disease, 59(3), 1017-1026. https://doi.org/10.3233/JAD-170226
  • Kawano, M., Kawakami, M., Otsuka, M., Yashima, H., Yaginuma, T., & Ueki, A. (1995). Marked decrease of plasma apolipoprotein AI and AII in Japanese patients with late-onset non-familial Alzheimer's disease. Clinica chimica acta; international journal of clinical chemistry, 239(2), 209-211. https://doi.org/10.1016/0009-8981(95)06115-t
  • Kim, W. S., He, Y., Phan, K., Ahmed, R. M., Rye, K. A., Piguet, O., Hodges, J. R., & Halliday, G. M. (2018). Altered high density lipoprotein composition in behavioral variant frontotemporal dementia. Frontiers in neuroscience, 12, 847. https://doi.org/10.3389/fnins.2018.00847
  • Koizumi, K., Hattori, Y., Ahn, S. J., Buendia, I., Ciacciarelli, A., Uekawa, K., Wang, G., Hiller, A., Zhao, L., Voss, H. U., Paul, S. M., Schaffer, C., Park, L., & Iadecola, C. (2018). Apoε4 disrupts neurovascular regulation and undermines white matter integrity and cognitive function. Nature communications, 9(1), 3816. https://doi.org/10.1038/s41467-018-06301-2
  • Kuriyama, M., Takahashi, K., Yamano, T., Hokezu, Y., Togo, S., Osame, M., & Igakura, T. (1994). Low levels of serum apolipoprotein AI and AII in senile dementia. Psychiatry and Clinical Neurosciences, 48(3), 589-593. https://doi.org/10.1111/j.1440-1819.1994.tb03019.x
  • Leduc, M. S., Shimmin, L. C., Klos, K. L., Hanis, C., Boerwinkle, E., & Hixson, J. E. (2008). Comprehensive evaluation of apolipoprotein H gene (APOH) variation identifies novel associations with measures of lipid metabolism in GENOA* s⃞. Journal of lipid research, 49(12), 2648-2656. https://doi.org/10.1194/jlr.M800155-JLR200
  • Lehnert, S., Jesse, S., Rist, W., Steinacker, P., Soininen, H., Herukka, S. K., Tumani, H., Lenter, M., Oeckl, P., Ferger, B., Hengerer, B., & Otto, M. (2012). iTRAQ and multiple reaction monitoring as proteomic tools for biomarker search in cerebrospinal fluid of patients with Parkinson's disease dementia. Experimental neurology, 234(2), 499-505. https://doi.org/10.1016/j.expneurol.2012.01.024
  • Lenzi, C., Ramazzina, I., Russo, I., Filippini, A., Bettuzzi, S., & Rizzi, F. (2020). The down-regulation of clusterin expression enhances the αsynuclein aggregation process. International Journal of Molecular Sciences, 21(19), 7181. https://doi.org/10.3390/ijms21197181
  • Leoni, E., Bremang, M., Mitra, V., Zubiri, I., Jung, S., Lu, C. H., Adiutori, R., Lombardi, V., Russell, C., Koncarevic, S., Ward, M., Pike, I., & Malaspina, A. (2019). Combined tissue-fluid proteomics to unravel phenotypic variability in amyotrophic lateral sclerosis. Scientific Reports, 9(1), 4478. https://doi.org/10.1038/s41598-019-40632-4
  • Lin, Y., Lu, L., Zhou, M., Liu, H., Ye, P., Zhang, W., Qiu, J., Zhang, Z., Yang, X., Ding, L., Guo, W., Mo, M., Zhu, X., Zhang, X., Chen, X., & Xu, P. (2021). Association of CLU gene polymorphism with Parkinson's disease in the Chinese Han population. The Journal of Gene Medicine, 23(2), e3302. https://doi.org/10.1002/jgm.3302
  • Liu, H. C., Hu, C. J., Chang, J. G., Sung, S. M., Lee, L. S., Yuan, R. Y., & Leu, S. J. (2006). Proteomic identification of lower apolipoprotein AI in Alzheimer’s disease. Dementia and geriatric cognitive disorders, 21(3), 155-161. https://doi.org/10.1159/000090676
  • Liu, T., Chen, J. M., Zhang, D., Zhang, Q., Peng, B., Xu, L., & Tang, H. (2021). ApoPred: identification of apolipoproteins and their subfamilies with multifarious features. Frontiers in Cell and Developmental Biology, 8, 621144. https://doi.org/10.3389/fcell.2020.621144
  • Maarouf, C. L., Beach, T. G., Adler, C. H., Shill, H. A., Sabbagh, M. N., Wu, T., Walker, D. G., Kokjohn, T. A., Roher, A. E., & Consortium, A. P. (2012). Cerebrospinal fluid biomarkers of neuropathologically diagnosed Parkinson’s disease subjects. Neurological research, 34(7), 669-676. https://doi.org/10.1179/1743132812Y.0000000063
  • Mata , I. F., Leverenz, J. B., Weintraub, D., Trojanowski, J. Q., Hurtig, H. I., Van Deerlin, V. M., Ritz, B., Rausch, R., Rhodes, S. L., Factor, S. A., Wood-Siverio, C., Quinn, J. F., Chung, K. A., Peterson, A. L., Espay, A. J., Revilla, F. J., Devoto, J., Hu, S., Cholerton, B. A., Wan, J. Y., Montine, T. J., Edwards, K. L., & Zabetian, C. P. (2014). APOE, MAPT, and SNCA genes and cognitive performance in Parkinson disease. JAMA neurology, 71(11), 1405-1412. https://doi.org/10.1001/jamaneurol.2014.1455
  • Mathieu, C., Pappu, R. V., & Taylor, J. P. (2020). Beyond aggregation: Pathological phase transitions in neurodegenerative disease. Science, 370(6512), 56-60. https://doi.org/10.1126/science.abb8032
  • Merched , A., Xia, Y., Visvikis, S., Serot, J. M., & Siest, G. (2000). Decreased high-density lipoprotein cholesterol and serum apolipoprotein AI concentrations are highly correlated with the severity of Alzheimer’s disease☆. Neurobiology of aging, 21(1), 27-30. https://doi.org/10.1016/s0197-4580(99)00103-7
  • McComb, M., Parambi, R., Browne, R. W., Bodziak, M. L., Jakimovski, D., Bergsland, N., Maceski, A., Weinstock-Guttman, B., Jens Kuhle, J., Zivadinov, R., & Ramanathan, M. (2020). Apolipoproteins AI and E are associated with neuroaxonal injury to gray matter in multiple sclerosis. Multiple sclerosis and related disorders, 45, 102389. https://doi.org/10.1016/j.msard.2020.102389
  • Misra, A., Chakrabarti, S. S., Gambhir, I. S., Baghel, M. S., & Patil, Y. R. (2021). Tandem mass spectrometry-based identification of protein profiles in the cerebrospinal fluid of Alzheimer’s dementia patients. https://doi.org/10.21203/rs.3.rs-574644/v1
  • Muenchhoff, J., Song, F., Poljak, A., Crawford, J. D., Mather, K. A., Kochan, N. A., Yang, Z., Trollor, J. N., Reppermund, S., Maston, K., Theobald, A., Kirchner-Adelhardt, S., Kwok, J. B., Richmond, R. L., McEvoy, M., Attia, J., Schofield, P. W., Brodaty, H., & Sachdev, P. S. (2017). Plasma apolipoproteins and physical and cognitive health in very old individuals. Neurobiology of Aging, 55, 49-60. https://doi.org/10.1016/j.neurobiolaging.2017.02.017
  • Murali, N., Browne, R. W., Fellows Maxwell, K., Bodziak, M. L., Jakimovski, D., Hagemeier, J., Bergsland, N., Weinstock-Guttman, B., Zivadinov, R., & Ramanathan, M. (2020). Cholesterol and neurodegeneration: longitudinal changes in serum cholesterol biomarkers are associated with new lesions and gray matter atrophy in multiple sclerosis over 5 years of follow‐up. European Journal of Neurology, 27(1), 188-e4. https://doi.org/10.1111/ene.14055
  • Nakaya, Y., Schaefer, E. J., & Brewer Jr, H. B. (1980). Activation of human post heparin lipoprotein lipase by apolipoprotein H (β2-glycoprotein I). Biochemical and biophysical research communications, 95(3), 1168-1172. https://doi.org/10.1016/0006-291X(80)91595-8
  • Namba, Y., Tsuchiya, H., & Ikeda, K. (1992). Apolipoprotein B immunoreactivity in senile plaque and vascular amyloids and neurofibrillary tangles in the brains of patients with Alzheimer's disease. Neuroscience letters, 134(2), 264-266. https://doi.org/10.1016/0304-3940(92)90531-b
  • Navarro, A., Rioseras, B., Del Valle, E., Martínez-Pinilla, E., Astudillo, A., & Tolivia, J. (2018). Expression pattern of myelin-related apolipoprotein D in human multiple sclerosis lesions. Frontiers in Aging Neuroscience, 10, 254. https://doi.org/10.3389/fnagi.2018.00254
  • Öhrfelt, A., Andreasson, U., Simon, A., Zetterberg, H., Edman, Å., Potter, W., Holder, D., Devanarayan, V., Seeburger, J., Smith, A. D., Blennow, K., & Wallin, A. (2011). Screening for new biomarkers for subcortical vascular dementia and Alzheimer’s disease. Dementia and geriatric cognitive disorders extra, 1(1), 31-42. https://doi.org/10.1159/000323417
  • Park, J. H., Hong, K. S., Lee, E. J., Lee, J., & Kim, D. E. (2011). High levels of apolipoprotein B/AI ratio are associated with intracranial atherosclerotic stenosis. Stroke, 42(11), 3040-3046. https://doi.org/10.1161/STROKEAHA.111.620104
  • Paula-Lima, A. C., Tricerri, M. A., Brito-Moreira, J., Bomfim, T. R., Oliveira, F. F., Magdesian, M. H., Grinberg, L. T., Panizzutti, R., & Ferreira, S. T. (2009). Human apolipoprotein A–I binds amyloid-β and prevents Aβ-induced neurotoxicity. The international journal of biochemistry & cell biology, 41(6), 1361-1370. https://doi.org/10.1016/j.biocel.2008.12.003
  • Přikrylová Vranová, H., Mareš, J., Nevrlý, M., Stejskal, D., Zapletalová, J., Hluštík, P., & Kaňovský, P. (2010). CSF markers of neurodegeneration in Parkinson’s disease. Journal of neural transmission, 117, 1177-1181. https://doi.org/10.1007/s00702-010-0462-z
  • Ramasamy, I. (2014). Recent advances in physiological lipoprotein metabolism. Clinical Chemistry and Laboratory Medicine (CCLM), 52(12), 1695-1727. https://doi.org/10.1515/cclm-2013-0358
  • Rassart, E., Bedirian, A., Do Carmo, S., Guinard, O., Sirois, J., Terrisse, L., & Milne, R. (2000). Apolipoprotein d. Biochimica et Biophysica Acta (BBA)-Protein Structure and Molecular Enzymology, 1482(1-2), 185-198. https://doi.org/10.1016/S0167-4838(00)00162-X
  • Real, R., Martinez-Carrasco, A., Reynolds, R. H., Lawton, M. A., Tan, M. M., Shoai, M., Corvol, J. C., Ryten, M., Bresner, C., Hubbard, L., Brice, A., Lesage, S., Faouzi, J., Elbaz, A., Artaud, F., Williams, N., Hu, M. T. M., Ben-Shlomo, Y., Grosset, D. G., ... & Morris, H. R. (2023). Association between the LRP1B and APOE loci and the development of Parkinson’s disease dementia. Brain, 146(5), 1873-1887. https://doi.org/10.1093/brain/awac414
  • Reichert, C. O., de Freitas, F. A., Sampaio-Silva, J., Rokita-Rosa, L., Barros, P. D. L., Levy, D., & Bydlowski, S. P. (2020a). Ferroptosis mechanisms involved in neurodegenerative diseases. International journal of molecular sciences, 21(22), 8765. https://doi.org/10.3390/ijms21228765
  • Reichert, C. O., Levy, D., & Bydlowski, S. P. (2020b). Paraoxonase role in human neurodegenerative diseases. Antioxidants, 10(1), 11. https://doi.org/10.3390/antiox10010011
  • Reindl, M., Knipping, G., Wicher, I., Dilitz, E., Egg, R., Deisenhammer, F., & Berger, T. (2001). Increased intrathecal production of apolipoprotein D in multiple sclerosis. Journal of neuroimmunology, 119(2), 327-332. https://doi.org/10.1016/s0165-5728(01)00378-2
  • Richardson, T. G., Sanderson, E., Palmer, T. M., Ala-Korpela, M., Ference, B. A., Davey Smith, G., & Holmes, M. V. (2020). Evaluating the relationship between circulating lipoprotein lipids and apolipoproteins with risk of coronary heart disease: A multivariable Mendelian randomisation analysis. PLoS medicine, 17(3), e1003062. https://doi.org/10.1371/journal.pmed.1003062
  • Ross MH, & Pawlina W. (2011). Histology: A text and atlas with correlated cell and molecular biology. Baltimore, Philadelphia: Lippincott Williams & Wilkins.
  • Saczynski, J. S., White, L., Peila, R. L., Rodriguez, B. L., & Launer, L. J. (2007). The relation between apolipoprotein AI and dementia: the Honolulu-Asia aging study. American journal of epidemiology, 165(9), 985-992. https://doi.org/10.1093/aje/kwm027
  • Sasaki , K., Doh-ura, K., Wakisaka, Y., & Iwaki, T. (2002). Clusterin/apolipoprotein J is associated with cortical Lewy bodies: immunohistochemical study in cases with α-synucleinopathies. Acta neuropathologica, 104, 225-230. https://doi.org/10.1007/s00401-002-0546-4
  • Shih, Y. H., Tsai, K. J., Lee, C. W., Shiesh, S. C., Chen, W. T., Pai, M. C., & Kuo, Y. M. (2014). Apolipoprotein C-III is an amyloid-β-binding protein and an early marker for Alzheimer's disease. Journal of Alzheimer's Disease, 41(3), 855-865. https://doi.org/10.3233/JAD-140111
  • Su, W. H., Shi, Z. H., Liu, S. L., Wang, X. D., Liu, S., & Ji, Y. (2017). Updated meta-analysis of the role of APOE ε2/ε3/ε4 alleles in frontotemporal lobar degeneration. Oncotarget, 8(27), 43721. https://doi.org/10.18632/oncotarget.17341
  • Sweeney, M. D., Sagare, A. P., & Zlokovic, B. V. (2018). Blood–brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nature Reviews Neurology, 14(3), 133-150. https://doi.org/10.1038/nrneurol.2017.188
  • Thompson, A. G., Talbot, K., & Turner, M. R. (2022). Higher blood high density lipoprotein and apolipoprotein A1 levels are associated with reduced risk of developing amyotrophic lateral sclerosis. Journal of Neurology, Neurosurgery & Psychiatry, 93(1), 75-81. https://doi.org/10.1136/jnnp-2021-327133
  • Tóth, M. E., Dukay, B., Hoyk, Z., & Sántha, M. (2020). Cerebrovascular changes and neurodegeneration related to hyperlipidemia: characteristics of the human ApoB-100 transgenic mice. Current Pharmaceutical Design, 26(13), 1486-1494. https://doi.org/10.2174/1381612826666200218101818
  • Tsuang, D., Leverenz, J. B., Lopez, O. L., Hamilton, R. L., Bennett, D. A., Schneider, J. A., Buchman, A. S., Larson, E. B., Crane, P. K., Kaye, J. A., Kramer, P., Woltjer, R., Trojanowski, J. O., Weintraub, D., Chen-Plotkin, A. S., Irwin, D. J., Rick, J., Schellenberg, G. D., Watson, G. S., … & Zabetian, C. P. (2013). APOE ϵ4 increases risk for dementia in pure synucleinopathies. JAMA neurology, 70(2), 223-228. https://doi.org/10.1001/jamaneurol.2013.600
  • Turan, T. N., Makki, A. A., Tsappidi, S., Cotsonis, G., Lynn, M. J., Cloft, H. J., & Chimowitz, M. I. (2010). Risk factors associated with severity and location of intracranial arterial stenosis. Stroke, 41(8), 1636-1640. https://doi.org/10.1161/STROKEAHA.110.584672
  • van Luijn, M. M., van Meurs, M., Stoop, M. P., Verbraak, E., Wierenga-Wolf, A. F., Melief, M. J., Kreft, K. L., Verdijk, R. M., ’t Hart, B. A., Luider, T. M., Laman, J. D., & Hintzen, R. Q. (2016). Elevated expression of the cerebrospinal fluid disease markers chromogranin a and clusterin in astrocytes of multiple sclerosis white matter lesions. Journal of Neuropathology & Experimental Neurology, 75(1), 86-98. https://doi.org/10.1093/jnen/nlv004
  • Vitali, C., Wellington, C. L., & Calabresi, L. (2014). HDL and cholesterol handling in the brain. Cardiovascular research, 103(3), 405-413. https://doi.org/10.1093/cvr/cvu148
  • Waldner, A., Dassati, S., Redl, B., Smania, N., & Gandolfi, M. (2018). Apolipoprotein D concentration in human plasma during aging and in Parkinson’s disease: a cross-sectional study. Parkinson’s Disease, 2018. https://doi.org/10.1155/2018/3751516
  • Wang, L., Sapuri-Butti, A. R., Aung, H. H., Parikh, A. N., & Rutledge, J. C. (2008). Triglyceride-rich lipoprotein lipolysis increases aggregation of endothelial cell membrane microdomains and produces reactive oxygen species. American Journal of Physiology-Heart and Circulatory Physiology, 295(1), H237-H244. https://doi.org/10.1152/ajpheart.01366.2007
  • Wei, Q., Wang, H., Tian, Y., Xu, F., Chen, X., & Wang, K. (2013). Reduced serum levels of triglyceride, very low density lipoprotein cholesterol and apolipoprotein B in Parkinson’s disease patients. PLoS One, 8(9), e75743. https://doi.org/10.1371/journal.pone.0075743
  • Weinstock-Guttman, B., Zivadinov, R., Mahfooz, N., Carl, E., Drake, A., Schneider, J., Teter, B., Hussein, S., Mehta, B., Weiskopf, M., Durfee, J., Bergsland, N., & Ramanathan, M. (2011). Serum lipid profiles are associated with disability and MRI outcomes in multiple sclerosis. Journal of neuroinflammation, 8(1), 1-7. https://doi.org/10.1186/1742-2094-8-127
  • Wisniewski, T., Golabek, A. A., Kida, E., Wisniewski, K. E., & Frangione, B. (1995). Conformational mimicry in Alzheimer's disease. Role of apolipoproteins in amyloidogenesis. The American journal of pathology, 147(2), 238. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC1869828/pdf/amjpathol00044-0015.pdf
  • Zhang, X., & Alzheimer’s Disease Neuroimaging Initiative. (2020). Higher plasma APOC-III was associated with a slower reduction of β-amyloid levels in cerebrospinal fluid among older individuals without dementia. Neuropsychiatric Disease and Treatment, 1139-1144. https://doi.org/10.2147/NDT.S238985
  • Zhang, X., Yin, X., Yu, H., Liu, X., Yang, F., Yao, J., Jin, H., & Yang, P. (2012). Quantitative proteomic analysis of serum proteins in patients with Parkinson’s disease using an isobaric tag for relative and absolute quantification labeling, two-dimensional liquid chromatography, and tandem mass spectrometry. Analyst, 137(2), 490-495. https://doi.org/10.1039/c1an15551b
  • Zlokovic, B. V., Martel, C. L., Matsubara, E., McComb, J. G., Zheng, G., McCluskey, R. T., Frangione, B., & Ghiso, J. (1996). Glycoprotein 330/megalin: probable role in receptor-mediated transport of apolipoprotein J alone and in a complex with Alzheimer disease amyloid beta at the blood-brain and blood-cerebrospinal fluid barriers. Proceedings of the National Academy of Sciences, 93(9), 4229-4234. https://doi.org/10.1073/pnas.93.9.4229
Toplam 80 adet kaynakça vardır.

Ayrıntılar

Birincil Dil İngilizce
Konular Klinik Tıp Bilimleri (Diğer)
Bölüm Makaleler
Yazarlar

Pınar Kaçamak 0000-0001-5650-9664

Çiğdem Elmas 0000-0002-8857-0918

Proje Numarası -
Yayımlanma Tarihi 31 Aralık 2023
Gönderilme Tarihi 30 Ekim 2023
Kabul Tarihi 27 Aralık 2023
Yayımlandığı Sayı Yıl 2023 Cilt: 5 Sayı: 3

Kaynak Göster

APA Kaçamak, P., & Elmas, Ç. (2023). Role of Apolipoproteins in Neurodegenerative Diseases. Journal of Gazi University Health Sciences Institute, 5(3), 122-133. https://doi.org/10.59124/guhes.1383395