Natural Products from Plants and Algae: History
Please note this is an old version of this entry, which may differ significantly from the current revision.

Neurodegenerative disorders including Parkinson’s disease (PD), Huntington’s disease (HD) and the most frequent, Alzheimer’s disease (AD), represent one of the most urgent medical needs worldwide. Despite a significantly developed understanding of disease development and pathology, treatments that stop AD progression are not yet available. The approval of sodium oligomannate (GV-971) for AD treatment in China emphasized the potential value of natural products for the treatment of neurodegenerative disorders. Many current clinical studies include the administration of a natural compound as a single and combination treatment. The most prominent mechanisms of action are anti-inflammatory and anti-oxidative activities, thus preserving cellular survival. 

  • Alzheimer’s disease
  • neurodegeneration
  • drug development
  • clinical studies

1. Introduction

Neurodegenerative diseases are a group of disorders in which neuronal function and survival are seriously affected. Many of these diseases, including Parkinson’s, Huntington’s and Alzheimer’s Disease (AD), are caused by structural changes and the deposition of proteins; therefore, they are also assigned to the group of protein misfolding diseases or amyloidoses [1][2][3]. AD is by far the most common cause of neurodegeneration and dementia. It is estimated that AD currently affects 55 million people worldwide (World-Alzheimer-Report-2021. Available online: https://www.alzint.org/u/World-Alzheimer-Report-2021.pdf, accessed on 4 February 2022). Characteristic symptoms of the disease are progressive memory loss, impaired cognitive function and paranoia. The histopathological hallmarks of AD, extracellular amyloid deposits (“amyloid plaques”), which mainly consist of the peptide Aβ, and intraneuronal neurofibrillary tangles of the hyperphosphorylated protein tau, mainly affect the cerebral cortex and the hippocampus [4][5]. Numerous studies suggest that the disease is initiated by the deposition of Aβ, which starts presumably years or decades before the first symptomatic changes [6]. The slow Aβ deposition triggers a downstream cascade (the amyloid cascade), which involves pathologic tau formation and hyperphosphorylation, widespread neuroinflammation and, finally, neuronal death [7][8]. Although the intense research during the last decades enabled a much better understanding of the crucial events in AD pathogenesis, a curative therapy that halts the progression of the disease is not yet available. Most of the so-called disease-modifying experimental drugs are targeting events of the amyloid cascade such as the generation and aggregation of Aβ and the phosphorylation of tau or the cellular metabolism and energy homeostasis [9]. The drug development in AD is faced with several challenges which has resulted in numerous setbacks in recent years [10]. For instance, the enzymes responsible for Aβ formation also have physiological substrates and functions. This complicates the suppression of amyloid peptide formation without interfering with other proteolytical degradation processes. Prominent examples are the γ-secretase complex and the β-secretase BACE1, which play a role in the formation of Aβ peptides [11][12][13]. Moreover, several reports suggest that Aβ1–40/42 and tau also have physiological functions, which leads one to question whether these represent druggable targets [14][15][16][17]. Also, many of the amyloidogenic proteins are localized in the cell nucleus or cytosol, which makes an effective suppression of the aggregation or the breakdown of the conglomerates, e.g., by antibodies, even more difficult [18]. Third, the efficient passage of the blood-brain barrier is needed and thus the pharmaceuticals are required to meet various physicochemical parameters [19][20]. Hence, methods are currently being examined (e.g., focused ultrasound) to make the blood-brain barrier more permeable [21].
Finally, major factors hampering the development and testing of new drugs are based on the clinical presentation of dementia and the currently available diagnostic biomarkers. AD patients frequently also show the presence of Lewy bodies and thus, significant pathological overlap with patients with dementia with Lewy bodies (DLB). As a result, the clinical testing of new active ingredients does not take place in “pure” Alzheimer’s patient populations. Accordingly, attempts are being made (using imaging methods and genetic analyses, among others) to conduct clinical studies in narrowly defined patient populations at an early stage of the disease [22][23][24]. Previously, numerous approaches were therefore undertaken in patients with a possibly too advanced a disease stage [23][25]. In addition, the available diagnostic biomarkers often do not specifically reflect the neurodegenerative disease or provide enough correlation with the clinical status of the patients. These imponderables could be responsible for the failure of different therapeutic approaches in the clinical phase. As mentioned above, alterations in biomarkers precede the symptoms of the disease [6][26], i.e., the measured value of a biomarker cannot be directly correlated with an effect on cognition. An example of this is the antibody bapineuzumab, which caused a significant change in phospho-tau in CSF in phase 2, but missed clinical endpoints [27].
All of these factors finally led to the numerous failures of disease-modifying drugs in AD clinical trials. The very recent accelerated approval of Aducanumab to treat AD may thus represent a first sign of success. However, the complexity also triggered the intense investigations of other fields, such as drugs from natural sources and nutraceuticals (Table 1). One potential reason is that food supplements may have the status as being generally regarded as safe (GRAS) and thus can be quickly applied in clinical testing, and eventually in combination with experimental drugs. Most of these substances are addressing protective mechanisms to cells by, e.g., anti-oxidative effects. However, there are also compounds in testing which are dedicated to disease-modification by, for example, their influence on immune cells. A prominent example is represented by oligomannate from red algae, which obtained approval for AD therapy in China and is currently being tested in additional clinical trials. Due to the emerging role in clinical testing, this entry focuses on the current treatment strategies which are based on natural products. 

This entry is adapted from the peer-reviewed paper 10.3390/biom12050694

References

  1. Rostagno, A.; Holton, J.L.; Lashley, T.; Revesz, T.; Ghiso, J. Cerebral amyloidosis: Amyloid subunits, mutants and phenotypes. Cell. Mol. Life Sci. 2009, 67, 581–600.
  2. Sipe, J.D.; Benson, M.D.; Buxbaum, J.N.; Ikeda, S.-I.; Merlini, G.; Saraiva, M.J.; Westermark, P. Amyloid fibril protein nomenclature: 2010 recommendations from the nomenclature committee of the International Society of Amyloidosis. Amyloid 2010, 17, 101–104.
  3. Hazenberg, B.P. Amyloidosis: A Clinical Overview. Rheum. Dis. Clin. N. Am. 2013, 39, 323–345.
  4. Braak, H.; Braak, E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol. 1991, 82, 239–259.
  5. Thal, D.R.; Rüb, U.; Orantes, M.; Braak, H. Phases of Aβ-deposition in the human brain and its relevance for the development of AD. Neurology 2002, 58, 1791–1800.
  6. Hadjichrysanthou, C.; Evans, S.; Bajaj, S.; Siakallis, L.C.; McRae-McKee, K.; de Wolf, F.; Anderson, R.M.; Initiative, T.A.D.N. The dynamics of biomarkers across the clinical spectrum of Alzheimer’s disease. Alzheimer’s Res. Ther. 2020, 12, 1102.
  7. Hardy, J.A.; Higgins, G.A. Alzheimer’s disease: The amyloid cascade hypothesis. Science 1992, 256, 184–185.
  8. Uddin, S.; Kabir, T.; Rahman, S.; Behl, T.; Jeandet, P.; Ashraf, G.M.; Najda, A.; Bin-Jumah, M.N.; El-Seedi, H.R.; Abdel-Daim, M.M. Revisiting the Amyloid Cascade Hypothesis: From Anti-Aβ Therapeutics to Auspicious New Ways for Alzheimer’s Disease. Int. J. Mol. Sci. 2020, 21, 5858.
  9. Cummings, J.; Lee, G.; Zhong, K.; Fonseca, J.; Taghva, K. Alzheimer’s disease drug development pipeline. Alzheimer’s Dementia Transl. Res. Clin. Interv. 2021, 7, e12179.
  10. Long, J.M.; Holtzman, D.M. Alzheimer Disease: An Update on Pathobiology and Treatment Strategies. Cell 2019, 179, 312–339.
  11. Lichtenthaler, S.F.; Tschirner, S.K.; Steiner, H. Secretases in Alzheimer’s disease: Novel insights into proteolysis of APP and TREM. Curr. Opin. Neurobiol. 2021, 72, 101–110.
  12. Imbimbo, B.P.; Watling, M. Investigational BACE inhibitors for the treatment of Alzheimer’s disease. Expert Opin. Investig. Drugs 2019, 28, 967–975.
  13. Luo, J.E.; Li, Y.-M. Turning the tide on Alzheimer’s disease: Modulation of γ-secretase. Cell Biosci. 2022, 12, 2.
  14. Puzzo, D.; Privitera, L.; Leznik, E.; Fà, M.; Staniszewski, A.; Palmeri, A.; Arancio, O. Picomolar Amyloid-β Positively Modulates Synaptic Plasticity and Memory in Hippocampus. J. Neurosci. 2008, 28, 14537–14545.
  15. Puzzo, D.; Privitera, L.; Fa’, M.; Staniszewski, A.; Hashimoto, G.; Aziz, F.; Sakurai, M.; Ribe, E.M.; Troy, C.M.; Mercken, M.; et al. Endogenous amyloid-β is necessary for hippocampal synaptic plasticity and memory. Ann. Neurol. 2011, 69, 819–830.
  16. Puzzo, D.; Arancio, O. Amyloid-β Peptide: Dr. Jekyll or Mr. Hyde? J. Alzheimer’s Dis. 2013, 33, S111–S120.
  17. Iqbal, K.; Liu, F.; Gong, C.-X. Tau and neurodegenerative disease: The story so far. Nat. Rev. Neurol. 2016, 12, 15–27.
  18. E Golde, T. Open questions for Alzheimer’s disease immunotherapy. Alzheimer’s Res. Ther. 2014, 6, 3.
  19. Rankovic, Z. CNS Drug Design: Balancing Physicochemical Properties for Optimal Brain Exposure. J. Med. Chem. 2015, 58, 2584–2608.
  20. Hubbard, R.E. Structure-based drug discovery and protein targets in the CNS. Neuropharmacology 2011, 60, 7–23.
  21. Liu, X.; Naomi, S.S.M.; Sharon, W.L.; Russell, E.J. The Applications of Focused Ultrasound (FUS) in Alzheimer’s Disease Treatment: A Systematic Review on Both Animal and Human Studies. Aging Dis. 2021, 12, 1977.
  22. Ringman, J.M.; Network, D.I.A.; Goate, A.; Masters, C.L.; Cairns, N.J.; Danek, A.; Graff-Radford, N.; Ghetti, B.; Morris, J.C. Genetic Heterogeneity in Alzheimer Disease and Implications for Treatment Strategies. Curr. Neurol. Neurosci. Rep. 2014, 14, 499.
  23. Schneider, L.S.; Mangialasche, F.; Andreasen, N.; Feldman, H.; Giacobini, E.; Jones, R.; Mantua, V.; Mecocci, P.; Pani, L.; Winblad, B.; et al. Clinical trials and late-stage drug development for Alzheimer’s disease: An appraisal from 1984 to 2014. J. Intern. Med. 2014, 275, 251–283.
  24. Duara, R.; Barker, W. Heterogeneity in Alzheimer’s Disease Diagnosis and Progression Rates: Implications for Therapeutic Trials. Neurotherapeutics 2022, 1–18.
  25. Giacobini, E.; Gold, G. Alzheimer disease therapy—Moving from amyloid-β to tau. Nat. Rev. Neurol. 2013, 9, 677–686.
  26. Bateman, R.J.; Xiong, C.; Benzinger, T.L.S.; Fagan, A.M.; Goate, A.; Fox, N.C.; Marcus, D.S.; Cairns, N.J.; Xie, X.; Blazey, T.M.; et al. Clinical and Biomarker Changes in Dominantly Inherited Alzheimer’s Disease. N. Engl. J. Med. 2012, 367, 795–804.
  27. Zetterberg, H.; Blennow, K.; Rinne, J.O.; Salloway, S.; Wei, J.; Black, R.; Grundman, M.; Liu, E.; for the AAB-001 201/202 Investigators. Effect of Immunotherapy with Bapineuzumab on Cerebrospinal Fluid Biomarker Levels in Patients With Mild to Moderate Alzheimer Disease. Arch. Neurol. 2012, 69, 1002–1010.
  28. Maurer, S.V.; Williams, C.L. The Cholinergic System Modulates Memory and Hippocampal Plasticity via Its Interactions with Non-Neuronal Cells. Front. Immunol. 2017, 8, 1489.
  29. Giacobini, E. Cholinesterase inhibitors: New roles and therapeutic alternatives. Pharmacol. Res. 2004, 50, 433–440.
  30. Folch, J.; Petrov, D.; Ettcheto, M.; Abad, S.; López, E.S.; García, M.L.; Olloquequi, J.; Beas-Zarate, C.; Auladell, C.; Camins, A. Current Research Therapeutic Strategies for Alzheimer’s Disease Treatment. Neural Plast. 2016, 2016, 8501693.
  31. Proskurnina, N.; Areshknina, L. Alkaloids of Galanthus woronovi. Allg. Chem. 1947, 17, 1216–1219.
  32. Mucke, H.A. The case of galantamine: Repurposing and late blooming of a cholinergic drug. Future Sci. OA 2015, 1, FSO73.
  33. Proskurnina, N.; Yakovieva, A. Alkaloids of Galanthus woronovi. Structure of galanthine. Ber. Akad. Wiss. UdSSR 1953, 90, 565–567.
  34. Uyeo, S.; Kobayashi, S. Lycoris Alkaloids. XXIV.: Isolation and Characterization of Lycoremine. Pharm. Bull. 1953, 1, 139–142.
  35. Irwin, R.; Smith, H. Cholinesterase inhibition by galanthamine and lycoramine. Biochem. Pharmacol. 1960, 3, 147–148.
  36. Lombardo, G.; Arena, G. The use of galantamine bromhydrate (nivaline) in the paralytic sequelae of poliomyelitis, neuraxitis and in muscular dystrophy. Minerva Pediatr. 1962, 30, 724–728.
  37. Stoyanov, E.; Vulchanova, S. The clinical application of nivalin as an antidote of curare. Nauchni Tr. Viss. Meditsinski Inst. Sofiia 1963, 42, 45–48.
  38. Baraka, A. Reversal of Central Anticholinergic Syndrome by Galanthamine. JAMA 1977, 238, 2293–2294.
  39. Czollner, L.; Frantsits, W.; Küenburg, B.; Hedenig, U.; Fröhlich, J.; Jordis, U. New kilogram-synthesis of the anti-alzheimer drug (−)-galanthamine. Tetrahedron Lett. 1998, 39, 2087–2088.
  40. Albuquerque, E.X.; Santos, M.D.; Alkondon, M.; Pereira, E.F.R.; Maelicke, A. Modulation of Nicotinic Receptor Activity in the Central Nervous System: A Novel Approach to the Treatment of Alzheimer Disease. Alzheimer Dis. Assoc. Disord. 2001, 15, S19–S25.
  41. Dajas-Bailador, F.A.; Heimala, K.; Wonnacott, S. The Allosteric Potentiation of Nicotinic Acetylcholine Receptors by Galantamine Is Transduced into Cellular Responses in Neurons: Ca2+ Signals and Neurotransmitter Release. Mol. Pharmacol. 2003, 64, 1217–1226.
  42. Samochocki, M.; Höffle, A.; Fehrenbacher, A.; Jostock, R.; Ludwig, J.; Christner, C.; Radina, M.; Zerlin, M.; Ullmer, C.; Pereira, E.F.R.; et al. Galantamine Is an Allosterically Potentiating Ligand of Neuronal Nicotinic but Not of Muscarinic Acetylcholine Receptors. J. Pharmacol. Exp. Ther. 2003, 305, 1024–1036.
  43. Schilström, B.; Ivanov, V.B.; Wiker, C.; Svensson, T.H. Galantamine Enhances Dopaminergic Neurotransmission In Vivo Via Allosteric Potentiation of Nicotinic Acetylcholine Receptors. Neuropsychopharmacology 2007, 32, 43–53.
  44. Lilienfeld, S. Galantamine—A Novel Cholinergic Drug with a Unique Dual Mode of Action for the Treatment of Patients with Alzheimer’s Disease. CNS Drug Rev. 2002, 8, 159–176.
  45. Tang, X.C.; Han, Y.F. Pharmacological Profile of Huperzine A, a Novel Acetylcholinesterase Inhibitor from Chinese Herb. CNS Drug Rev. 1999, 5, 281–300.
  46. Wang, Y.; Yue, D.; Tang, X. Anti-Cholinesterase Activity of Huerpzine A. Acta Pharmacol. Sin. 1986, 7, 110–113.
  47. Wang, H.; Tang, X. Anticholinesterase Effects of Huperzine A, E2020, and Tacrine in Rats. Zhongguo Yao Li Xue Bao Acta Pharmacol. Sin. 1998, 19, 27–30.
  48. Wang, R.; Tang, X.C. Neuroprotective Effects of Huperzine A. Neurosignals 2005, 14, 71–82.
  49. Zhang, H.Y.; Liang, Y.Q.; Tang, X.C.; He, X.C.; Bai, D.L. Stereoselectivities of enantiomers of huperzine A in protection against β-amyloid25–35-induced injury in PC12 and NG108-15 cells and cholinesterase inhibition in mice. Neurosci. Lett. 2002, 317, 143–146.
  50. Orhan, G.; Orhan, I.; Subutay-Oztekin, N.; Ak, F.; Sener, B. Contemporary Anticholinesterase Pharmaceuticals of Natural Origin and Their Synthetic Analogues for the Treatment of Alzheimers Disease. Recent Patents CNS Drug Discov. 2009, 4, 43–51.
  51. Zhan, Z.-J.; Bian, H.-L.; Wang, J.-W.; Shan, W.-G. Synthesis of physostigmine analogues and evaluation of their anticholinesterase activities. Bioorganic Med. Chem. Lett. 2010, 20, 1532–1534.
  52. McKinney, M.; Miller, J.H.; Yamada, F.; Tuckmantel, W.; Kozikowski, A.P. Potencies and stereoselectivities of enantiomers of huperzine A for inhibition of rat cortical acetylcholinesterase. Eur. J. Pharmacol. 1991, 203, 303–305.
  53. Dvir, H.; Jiang, H.L.; Wong, D.M.; Harel, M.; Chetrit, M.; He, X.C.; Jin, G.Y.; Yu, G.L.; Tang, X.C.; Silman, I.; et al. X-ray Structures of Torpedo californica Acetylcholinesterase Complexed with (+)-Huperzine A and (−)-Huperzine B: Structural Evidence for an Active Site Rearrangement. Biochemistry 2002, 41, 10810–10818.
  54. Zhang, H.-Y.; Brimijoin, S.; Tang, X.-C. Apoptosis induced by beta-amyloid25-35 in acetylcholinesterase-overexpressing neuroblastoma cells. Acta Pharmacol. Sin. 2003, 24, 853–858.
  55. Yan, X.F.; Lu, W.H.; Lou, W.J.; Tang, X.C. Effects of Huperzine A and B on Skeletal Muscle and the Electroenphalogram. Acta Pharmacol. Sin. 1987, 8, 117–123.
  56. Zhang, R.W.; Tang, X.C.; Han, Y.Y.; Sang, G.W.; Zhang, Y.D.; Ma, Y.X.; Zhang, C.L.; Yang, R.M. Drug evaluation of huperzine A in the treatment of senile memory disorders. Acta Pharmacol. Sin. 1991, 12, 250–252.
  57. Xu, S.S.; Gao, Z.X.; Weng, Z.; Du, Z.M.; Xu, W.A.; Yang, J.S.; Zhang, M.L.; Tong, Z.H.; Fang, Y.S.; Chai, X.S. Efficacy of Tablet Huperzine A on Memory, Cognition and Behavior in Alzheimer´s Disease. Acta Pharmacol. Sin. 1995, 16, 195–391.
  58. Xu, S.S.; Cai, Z.Y.; Qu, Z.W.; Yang, R.M.; Cai, Y.L.; Wang, G.Q.; Su, X.Q.; Zhong, X.S.; Cheng, R.Y.; A Xu, W.; et al. Huperzine-A in capsules and tablets for treating patients with Alzheimer disease. Acta Pharmacol. Sin. 1999, 20, 486–490.
  59. Liao, B.; Newmark, H.; Zhou, R. Neuroprotective Effects of Ginseng Total Saponin and Ginsenosides Rb1 and Rg1 on Spinal Cord Neurons In Vitro. Exp. Neurol. 2002, 173, 224–234.
  60. Gillis, C. Panax ginseng pharmacology: A nitric oxide link? Biochem. Pharmacol. 1997, 54, 1–8.
  61. Pena, I.D.; Yoon, S.Y.; Kim, H.J.; Park, S.; Hong, E.Y.; Ryu, J.H.; Park, I.H.; Cheong, J.H. Effects of ginseol k-g3, an Rg3-enriched fraction, on scopolamine-induced memory impairment and learning deficit in mice. J. Ginseng Res. 2014, 38, 1–7.
  62. Rausch, W.-D.; Liu, S.; Gille, G.; Radad, K. Neuroprotective effects of ginsenosides. Acta Neurobiol. Exp. 2006, 66, 369–375.
  63. Nah, S.-Y.; Kim, D.-H.; Rhim, H. Ginsenosides: Are Any of them Candidates for Drugs Acting on the Central Nervous System? CNS Drug Rev. 2007, 13, 381–404.
  64. Radad, K.; Moldzio, R.; Rausch, W.-D. Ginsenosides and Their CNS Targets. CNS Neurosci. Ther. 2011, 17, 761–768.
  65. Lee, D.W.; Andersen, J.K. Iron elevations in the aging Parkinsonian brain: A consequence of impaired iron homeostasis? J. Neurochem. 2010, 112, 332–339.
  66. Selkoe, D.J.; Schenk, D. Alzheimer’s Disease: Molecular Understanding Predicts Amyloid-Based Therapeutics. Annu. Rev. Pharmacol. Toxicol. 2003, 43, 545–584.
  67. Dedov, V.N.; Griffiths, F.M.; Garner, B.; Halliday, G.M.; Double, K.L. Lipid content determines aggregation of neuromelanin granules in vitro. J. Neural. Transm. Suppl. 2007, 72, 35–38.
  68. Tanzi, R.E.; Bertram, L. Alzheimer´s Disease: The latest suspect. Nature 2008, 454, 707–708.
  69. Xu, B.-B.; Liu, C.-Q.; Gao, X.; Zhang, W.-Q.; Wang, S.-W.; Cao, Y.-L. Possible mechanisms of the protection of ginsenoside Re against MPTP-induced apoptosis in substantia nigra neurons of Parkinson’s disease mouse model. J. Asian Nat. Prod. Res. 2005, 7, 215–224.
  70. Oh, J.; Kim, J.-S. Compound K derived from ginseng: Neuroprotection and cognitive improvement. Food Funct. 2016, 7, 4506–4515.
  71. Fang, F.; Chen, X.; Huang, T.; Lue, L.-F.; Luddy, J.S.; Yan, S.S. Multi-faced neuroprotective effects of Ginsenoside Rg1 in an Alzheimer mouse model. Biochim. Biophys. Acta (BBA)-Mol. Basis Dis. 2012, 1822, 286–292.
  72. Hong-Cai, W.; Yu-Meng, J.; Xue, Z.; Ning, W.; Di, G.; Ming, G.; Jing-Yu, Z.; Zi-Chao, Y. Protective Effects of Ginsenoside Rb1 on Aβ Amyloid-Induced Hippocampal Neuronal Injury in Rats. J. Jilin Univ. Med. Ed. 2012, 38, 447–450.
  73. Huang, X.; Li, N.; Pu, Y.; Zhang, T.; Wang, B. Neuroprotective Effects of Ginseng Phytochemicals: Recent Perspectives. Molecules 2019, 24, 2939.
  74. Zhao, H.-H.; Di, J.; Liu, W.-S.; Liu, H.-L.; Lai, H.; Lü, Y.-L. Involvement of GSK3 and PP2A in ginsenoside Rb1’s attenuation of aluminum-induced tau hyperphosphorylation. Behav. Brain Res. 2013, 241, 228–234.
  75. Oken, B.S.; Storzbach, D.M.; Kaye, J.A. The efficacy of Ginkgo biloba on cognitive function in Alzheimer disease. Arch. Neurol. 1998, 55, 1409–1415.
  76. Vellas, B.; Coley, N.; Ousset, P.-J.; Berrut, G.; Dartigues, J.-F.; Dubois, B.; Grandjean, H.; Pasquier, F.; Piette, F.; Robert, P.; et al. Long-term use of standardised ginkgo biloba extract for the prevention of Alzheimer’s disease (GuidAge): A randomised placebo-controlled trial. Lancet Neurol. 2012, 11, 851–859.
  77. Singh, S.K.; Srivastav, S.; Castellani, R.J.; Plascencia-Villa, G.; Perry, G. Neuroprotective and Antioxidant Effect of Ginkgo biloba Extract Against AD and Other Neurological Disorders. Neurotherapeutics 2019, 16, 666–674.
  78. Smith, P.F.; Maclennan, K.; Darlington, C.L. The neuroprotective properties of the Ginkgo biloba leaf: A review of the possible relationship to platelet-activating factor (PAF). J. Ethnopharmacol. 1996, 50, 131–139.
  79. Shi, C.; Liu, J.; Wu, F.; Yew, D.T. Ginkgo biloba Extract in Alzheimer’s Disease: From Action Mechanisms to Medical Practice. Int. J. Mol. Sci. 2010, 11, 107–123.
  80. Augustin, S.; Rimbach, G.; Augustin, K.; Schliebs, R.; Wolffram, S.; Cermak, R. Effect of a short- and long-term treatment with Ginkgo biloba extract on Amyloid Precursor Protein Levels in a transgenic mouse model relevant to Alzheimer’s disease. Arch. Biochem. Biophys. 2009, 481, 177–182.
  81. Löffler, T.; Lee, S.K.; Nöldner, M.; Chatterjee, S.S.; Hoyer, S.; Schliebs, R. Effect of Ginkgo biloba extract (EGb761) on glucose metabolism-related markers in streptozotocin-damaged rat brain. J. Neural Transm. 2001, 108, 1457–1474.
  82. Le Bars, P.L.; Katz, M.M.; Berman, N.; Itil, T.M.; Freedman, A.M.; Schatzberg, A.F. A Placebo-Controlled, Double-blind, Randomized Trial of an Extract of Ginkgo Biloba for Dementia. JAMA 1997, 278, 1327–1332.
  83. Mix, J.A.; Crews, W.D. A double-blind, placebo-controlled, randomized trial of Ginkgo biloba extract EGb 761® in a sample of cognitively intact older adults: Neuropsychological findings. Hum. Psychopharmacol. Clin. Exp. 2002, 17, 267–277.
  84. Kanowski, S.; Herrmann, W.M.; Stephan, K.; Wierich, W.; Hörr, R. Proof of Efficacy of the Ginkgo Biloba Special Extract EGb 761 in Outpatients Suffering from Mild to Moderate Primary Degenerative Dementia of the Alzheimer Type or Multi-infarct Dementia. Pharmacopsychiatry 2007, 29, 47–56.
  85. Napryeyenko, O.; Borzenko, I. Ginkgo biloba Special Extract in Dementia with Neuropsychiatric Features. Arzneimittelforschung 2007, 57, 4–11.
  86. Gao, Y.; Zhang, L.; Jiao, W. Marine glycan-derived therapeutics in China. Prog. Mol. Biol. Transl. Sci. 2019, 163, 113–134.
  87. Wang, X.; Sun, G.; Feng, T.; Zhang, J.; Huang, X.; Wang, T.; Xie, Z.; Chu, X.; Yang, J.; Wang, H.; et al. Sodium oligomannate therapeutically remodels gut microbiota and suppresses gut bacterial amino acids-shaped neuroinflammation to inhibit Alzheimer’s disease progression. Cell Res. 2019, 29, 787–803.
  88. Syed, Y.Y. Sodium Oligomannate: First Approval. Drugs 2020, 80, 441–444.
  89. Xiao, S.; Chan, P.; Wang, T.; Hong, Z.; Wang, S.; Kuang, W.; He, J.; Pan, X.; Zhou, Y.; Ji, Y.; et al. A 36-week multicenter, randomized, double-blind, placebo-controlled, parallel-group, phase 3 clinical trial of sodium oligomannate for mild-to-moderate Alzheimer’s dementia. Alzheimer’s Res. Ther. 2021, 13, 62.
  90. Isaka, S.; Cho, K.; Nakazono, S.; Abu, R.; Ueno, M.; Kim, D.; Oda, T. Antioxidant and anti-inflammatory activities of porphyran isolated from discolored nori (Porphyra yezoensis). Int. J. Biol. Macromol. 2015, 74, 68–75.
  91. Kim, M.; Li, Y.-X.; Dewapriya, P.; Ryu, B.; Kim, S.-K. Floridoside suppresses pro-inflammatory responses by blocking MAPK signaling in activated microglia. BMB Rep. 2013, 46, 398–403.
  92. Cui, Y.-Q.; Jia, Y.-J.; Zhang, T.; Zhang, Q.-B.; Wang, X.-M. Fucoidan Protects against Lipopolysaccharide-Induced Rat Neuronal Damage and Inhibits the Production of Proinflammatory Mediators in Primary Microglia. CNS Neurosci. Ther. 2012, 18, 827–833.
  93. Park, H.Y.; Han, M.H.; Park, C.; Jin, C.-Y.; Kim, G.-Y.; Choi, I.-W.; Kim, N.D.; Nam, T.-J.; Kwon, T.K.; Choi, Y.H. Anti-inflammatory effects of fucoidan through inhibition of NF-κB, MAPK and Akt activation in lipopolysaccharide-induced BV2 microglia cells. Food Chem. Toxicol. 2011, 49, 1745–1752.
  94. Yao, Z.-A.; Xu, L.; Wu, H.-G. Immunomodulatory Function of κ-Carrageenan Oligosaccharides Acting on LPS-Activated Microglial Cells. Neurochem. Res. 2014, 39, 333–343.
More
This entry is offline, you can click here to edit this entry!