Anti-Inflammatory Activities of Marine Algae in Neurodegenerative Diseases
Abstract
:1. Introduction
2. Methods
3. Molecular Mechanisms of Neuroinflammation
4. Marine Algae
4.1. Carbohydrates
4.2. Lipids
4.3. Proteins and Amino Acids
4.4. Phenols
4.5. Isoprenoids
5. Marine Algae and Neuroinflammation
6. Conclusions
Funding
Conflicts of Interest
Abbreviations
AChE | Acetylcholinesterase |
AD | Alzheimer’s disease |
ALS | Amyotrophic lateral sclerosis |
AP-1 | Activating protein 1 |
A- | Amyloid beta |
BACE1 | Beta-secretase 1 |
BBB | Blood‒brain barrier |
BChE | Butyrylcholinesterase |
CNS | Central nervous system |
COX-2 | Cyclooxygenase-2 |
EPA | Eicosapentaenoic acid |
HD | Huntington’s disease |
IL | Interleukin |
iNOS | Inducible nitric oxide synthase |
JNK | c-Jun N-terminal kinases |
LPS | Lipopolysaccharide |
MAPKs | Mitogen-activated protein kinases cascade |
MS | Multiple sclerosis |
NF-κB | Nuclear factor κB |
NGF | Nerve growth factor |
nNOS | Neuronal nitric oxide synthase |
NO | Nitric oxide |
NSAIDs | Non-steroidal anti-inflammatory drugs |
PD | Parkinson’s disease |
PGE2 | Prostaglandin E2 |
PUFA | Polyunsaturated fatty acids |
TBI | Traumatic brain injury |
TGFβ | Transforming growth factor beta |
TLRs | Toll-like receptor |
TNF-α | Tumor necrosis factor alpha |
References
- Höglund, K.; Salter, H. Molecular biomarkers of neurodegeneration. Expert Rev. Mol. Diagn. 2013, 13, 845–861. [Google Scholar] [CrossRef] [PubMed]
- Angeloni, C.; Prata, C.; Dalla Sega, F.V.; Piperno, R.; Hrelia, S. Traumatic brain injury and NADPH oxidase: A deep relationship. Oxid. Med. Cell Longev. 2015, 2015, 370312. [Google Scholar] [CrossRef] [PubMed]
- Tarozzi, A.; Angeloni, C.; Malaguti, M.; Morroni, F.; Hrelia, S.; Hrelia, P. Sulforaphane as a potential protective phytochemical against neurodegenerative diseases. Oxid. Med. Cell Longev. 2013, 2013, 415078. [Google Scholar] [CrossRef] [PubMed]
- Hammond, T.R.; Marsh, S.E.; Stevens, B. Immune Signaling in Neurodegeneration. Immunity 2019, 50, 955–974. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Frank-Cannon, T.C.; Alto, L.T.; McAlpine, F.E.; Tansey, M.G. Does neuroinflammation fan the flame in neurodegenerative diseases? Mol. Neurodegener 2009, 4, 47. [Google Scholar] [CrossRef] [PubMed]
- Morales, I.; Guzmán-Martínez, L.; Cerda-Troncoso, C.; Farías, G.A.; Maccioni, R.B. Neuroinflammation in the pathogenesis of Alzheimer’s disease. A rational framework for the search of novel therapeutic approaches. Front. Cell Neurosci. 2014, 8, 112. [Google Scholar] [CrossRef] [PubMed]
- Glass, C.K.; Saijo, K.; Winner, B.; Marchetto, M.C.; Gage, F.H. Mechanisms underlying inflammation in neurodegeneration. Cell 2010, 140, 918–934. [Google Scholar] [CrossRef]
- Spencer, J.P.; Vafeiadou, K.; Williams, R.J.; Vauzour, D. Neuroinflammation: modulation by flavonoids and mechanisms of action. Mol. Asp. Med. 2012, 33, 83–97. [Google Scholar] [CrossRef]
- Schain, M.; Kreisl, W.C. Neuroinflammation in Neurodegenerative Disorders-a Review. Curr. Neurol. Neurosci. Rep. 2017, 17, 25. [Google Scholar] [CrossRef]
- Sofroniew, M.V. Multiple roles for astrocytes as effectors of cytokines and inflammatory mediators. Neuroscientist 2014, 20, 160–172. [Google Scholar] [CrossRef]
- Neal, M.; Richardson, J.R. Epigenetic regulation of astrocyte function in neuroinflammation and neurodegeneration. Biochim Biophys Acta. Mol. Basis Dis. 2018, 1864, 432–443. [Google Scholar] [CrossRef] [PubMed]
- Motori, E.; Puyal, J.; Toni, N.; Ghanem, A.; Angeloni, C.; Malaguti, M.; Cantelli-Forti, G.; Berninger, B.; Conzelmann, K.K.; Götz, M.; et al. Inflammation-induced alteration of astrocyte mitochondrial dynamics requires autophagy for mitochondrial network maintenance. Cell Metab 2013, 18, 844–859. [Google Scholar] [CrossRef] [PubMed]
- Decheng, B.; Qiuxian, L.; Qingguo, H.; Nan, C.; Hanxing, H.; Weishan, F.; Jiang, Y.; Xiaofan, L.; Hong, X.; Xiuting, L.; et al. Seleno-polymannuronate attenuates neuroinflammation by suppressing microglial and astrocytic activation. J. Funct. Foods 2018, 51, 113–120. [Google Scholar]
- Ning, C.; Wang, H.D.; Gao, R.; Chang, Y.C.; Hu, F.; Meng, X.; Huang, S.Y. Marine-derived protein kinase inhibitors for neuroinflammatory diseases. Biomed. Eng. Online 2018, 17, 46. [Google Scholar] [CrossRef] [PubMed]
- Park, E.J.; Pezzuto, J.M. Antioxidant marine products in cancer chemoprevention. Antioxid Redox Signal. 2013, 19, 115–138. [Google Scholar] [CrossRef] [PubMed]
- Pejin, B.; Jovanovic, K.K.; Savic, A.G. New antitumour natural products from marine red algae: Covering the period from 2003 to 2012. Mini. Rev. Med. Chem. 2015, 15, 720–730. [Google Scholar] [CrossRef] [PubMed]
- Jin, Q.; Yu, H.; Li, P. The Evaluation and Utilization of Marine-derived Bioactive Compounds with Anti-obesity Effect. Curr. Med. Chem. 2018, 25, 861–878. [Google Scholar] [CrossRef] [PubMed]
- Huang, C.; Zhang, Z.; Cui, W. Marine-Derived Natural Compounds for the Treatment of Parkinson’s Disease. Mar. Drugs 2019, 17, 221. [Google Scholar] [CrossRef]
- Newman, D.J.; Cragg, G.M. Advanced preclinical and clinical trials of natural products and related compounds from marine sources. Curr. Med. Chem. 2004, 11, 1693–1713. [Google Scholar] [CrossRef] [PubMed]
- Brown, E.S.; Allsopp, P.J.; Magee, P.J.; Gill, C.I.; Nitecki, S.; Strain, C.R.; McSorley, E.M. Seaweed and human health. Nutr. Rev. 2014, 72, 205–216. [Google Scholar] [CrossRef] [PubMed]
- Fernando, I.P.S.; Nah, J.W.; Jeon, Y.J. Potential anti-inflammatory natural products from marine algae. Env. Toxicol Pharm. 2016, 48, 22–30. [Google Scholar] [CrossRef] [PubMed]
- Jin, D.Q.; Lim, C.S.; Sung, J.Y.; Choi, H.G.; Ha, I.; Han, J.S. Ulva conglobata, a marine algae, has neuroprotective and anti-inflammatory effects in murine hippocampal and microglial cells. Neurosci. Lett. 2006, 402, 154–158. [Google Scholar] [CrossRef] [PubMed]
- Yu, D.K.; Lee, B.; Kwon, M.; Yoon, N.; Shin, T.; Kim, N.G.; Choi, J.S.; Kim, H.R. Phlorofucofuroeckol B suppresses inflammatory responses by down-regulating nuclear factor κB activation via Akt, ERK, and JNK in LPS-stimulated microglial cells. Int. Immunopharmacol 2015, 28, 1068–1075. [Google Scholar] [CrossRef]
- Jiang, X.; Chen, L.; Shen, L.; Chen, Z.; Xu, L.; Zhang, J.; Yu, X. Trans-astaxanthin attenuates lipopolysaccharide-induced neuroinflammation and depressive-like behavior in mice. Brain Res. 2016, 1649, 30–37. [Google Scholar] [CrossRef] [PubMed]
- Fan, Y.; Hu, J.; Li, J.; Yang, Z.; Xin, X.; Wang, J.; Ding, J.; Geng, M. Effect of acidic oligosaccharide sugar chain on scopolamine-induced memory impairment in rats and its related mechanisms. Neurosci. Lett. 2005, 374, 222–226. [Google Scholar] [CrossRef]
- Scarpignato, C.; Dolak, W.; Lanas, A.; Matzneller, P.; Renzulli, C.; Grimaldi, M.; Zeitlinger, M.; Bjarnason, I. Rifaximin Reduces the Number and Severity of Intestinal Lesions Associated With Use of Nonsteroidal Anti-Inflammatory Drugs in Humans. Gastroenterology 2017, 152, 980–982.e983. [Google Scholar] [CrossRef]
- Yuan, G.; Wahlqvist, M.L.; He, G.; Yang, M.; Li, D. Natural products and anti-inflammatory activity. Asia Pac. J. Clin. Nutr. 2006, 15, 143–152. [Google Scholar]
- Adams, K.L.; Gallo, V. The diversity and disparity of the glial scar. Nat. Neurosci. 2018, 21, 9–15. [Google Scholar] [CrossRef]
- Nayak, D.; Roth, T.L.; McGavern, D.B. Microglia development and function. Annu. Rev. Immunol. 2014, 32, 367–402. [Google Scholar] [CrossRef]
- Ginhoux, F.; Greter, M.; Leboeuf, M.; Nandi, S.; See, P.; Gokhan, S.; Mehler, M.F.; Conway, S.J.; Ng, L.G.; Stanley, E.R.; et al. Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 2010, 330, 841–845. [Google Scholar] [CrossRef]
- Subhramanyam, C.S.; Wang, C.; Hu, Q.; Dheen, S.T. Microglia-mediated neuroinflammation in neurodegenerative diseases. Semin Cell Dev. Biol. 2019. [Google Scholar] [CrossRef] [PubMed]
- Schwartz, M.; Kipnis, J.; Rivest, S.; Prat, A. How do immune cells support and shape the brain in health, disease, and aging? J. Neurosci. 2013, 33, 17587–17596. [Google Scholar] [CrossRef] [PubMed]
- Ueno, M.; Fujita, Y.; Tanaka, T.; Nakamura, Y.; Kikuta, J.; Ishii, M.; Yamashita, T. Layer V cortical neurons require microglial support for survival during postnatal development. Nat. Neurosci. 2013, 16, 543–551. [Google Scholar] [CrossRef] [PubMed]
- Takahashi, K.; Rochford, C.D.; Neumann, H. Clearance of apoptotic neurons without inflammation by microglial triggering receptor expressed on myeloid cells-2. J. Exp. Med. 2005, 201, 647–657. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liao, H.; Bu, W.Y.; Wang, T.H.; Ahmed, S.; Xiao, Z.C. Tenascin-R plays a role in neuroprotection via its distinct domains that coordinate to modulate the microglia function. J. Biol. Chem. 2005, 280, 8316–8323. [Google Scholar] [CrossRef] [PubMed]
- Drögemüller, K.; Helmuth, U.; Brunn, A.; Sakowicz-Burkiewicz, M.; Gutmann, D.H.; Mueller, W.; Deckert, M.; Schlüter, D. Astrocyte gp130 expression is critical for the control of Toxoplasma encephalitis. J. Immunol. 2008, 181, 2683–2693. [Google Scholar] [CrossRef] [PubMed]
- Cho, M.L.; Min, S.Y.; Chang, S.H.; Kim, K.W.; Heo, S.B.; Lee, S.H.; Park, S.H.; Cho, C.S.; Kim, H.Y. Transforming growth factor beta 1(TGF-beta1) down-regulates TNFalpha-induced RANTES production in rheumatoid synovial fibroblasts through NF-kappaB-mediated transcriptional repression. Immunol. Lett. 2006, 105, 159–166. [Google Scholar] [CrossRef]
- Wyss-Coray, T.; Mucke, L. Inflammation in neurodegenerative disease—A double-edged sword. Neuron 2002, 35, 419–432. [Google Scholar] [CrossRef]
- Hoshino, K.; Takeuchi, O.; Kawai, T.; Sanjo, H.; Ogawa, T.; Takeda, Y.; Takeda, K.; Akira, S. Cutting edge: Toll-like receptor 4 (TLR4)-deficient mice are hyporesponsive to lipopolysaccharide: evidence for TLR4 as the Lps gene product. J. Immunol. 1999, 162, 3749–3752. [Google Scholar]
- Tang, Y.; Le, W. Differential Roles of M1 and M2 Microglia in Neurodegenerative Diseases. Mol. Neurobiol. 2016, 53, 1181–1194. [Google Scholar] [CrossRef]
- Colton, C.A.; Gilbert, D.L. Production of superoxide anions by a CNS macrophage, the microglia. FEBS Lett. 1987, 223, 284–288. [Google Scholar] [CrossRef] [Green Version]
- Graeber, M.B.; Streit, W.J.; Kreutzberg, G.W. Axotomy of the rat facial nerve leads to increased CR3 complement receptor expression by activated microglial cells. J. Neurosci. Res. 1988, 21, 18–24. [Google Scholar] [CrossRef] [PubMed]
- Chen, W.W.; Zhang, X.; Huang, W.J. Role of neuroinflammation in neurodegenerative diseases. Mol. Med. Rep. 2016, 13, 3391–3396. [Google Scholar] [CrossRef] [PubMed]
- Lull, M.E.; Block, M.L. Microglial activation and chronic neurodegeneration. Neurotherapeutics 2010, 7, 354–365. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Licastro, F.; Hrelia, S.; Porcellini, E.; Malaguti, M.; Di Stefano, C.; Angeloni, C.; Carbone, I.; Simoncini, L.; Piperno, R. Peripheral Inflammatory Markers and Antioxidant Response during the Post-Acute and Chronic Phase after Severe Traumatic Brain Injury. Front. Neurol. 2016, 7, 189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tansey, M.G.; McCoy, M.K.; Frank-Cannon, T.C. Neuroinflammatory mechanisms in Parkinson’s disease: potential environmental triggers, pathways, and targets for early therapeutic intervention. Exp. Neurol. 2007, 208, 1–25. [Google Scholar] [CrossRef] [PubMed]
- Das Sarma, J. Microglia-mediated neuroinflammation is an amplifier of virus-induced neuropathology. J. Neurovirol. 2014, 20, 122–136. [Google Scholar] [CrossRef]
- Chen, H.; Chan, D.C. Mitochondrial dynamics--fusion, fission, movement, and mitophagy--in neurodegenerative diseases. Hum. Mol. Genet. 2009, 18, R169–R176. [Google Scholar] [CrossRef]
- Xiang, W.; Chao, Z.Y.; Feng, D.Y. Role of Toll-like receptor/MYD88 signaling in neurodegenerative diseases. Rev. Neurosci. 2015, 26, 407–414. [Google Scholar] [CrossRef]
- Lehnardt, S.; Massillon, L.; Follett, P.; Jensen, F.E.; Ratan, R.; Rosenberg, P.A.; Volpe, J.J.; Vartanian, T. Activation of innate immunity in the CNS triggers neurodegeneration through a Toll-like receptor 4-dependent pathway. Proc. Natl. Acad. Sci USA 2003, 100, 8514–8519. [Google Scholar] [CrossRef] [Green Version]
- Balducci, C.; Frasca, A.; Zotti, M.; La Vitola, P.; Mhillaj, E.; Grigoli, E.; Iacobellis, M.; Grandi, F.; Messa, M.; Colombo, L.; et al. Toll-like receptor 4-dependent glial cell activation mediates the impairment in memory establishment induced by β-amyloid oligomers in an acute mouse model of Alzheimer’s disease. Brain Behav. Immun. 2017, 60, 188–197. [Google Scholar] [CrossRef] [PubMed]
- Capiralla, H.; Vingtdeux, V.; Zhao, H.; Sankowski, R.; Al-Abed, Y.; Davies, P.; Marambaud, P. Resveratrol mitigates lipopolysaccharide- and Aβ-mediated microglial inflammation by inhibiting the TLR4/NF-κB/STAT signaling cascade. J. Neurochem. 2012, 120, 461–472. [Google Scholar] [CrossRef] [PubMed]
- Lv, R.; Du, L.; Liu, X.; Zhou, F.; Zhang, Z.; Zhang, L. Rosmarinic acid attenuates inflammatory responses through inhibiting HMGB1/TLR4/NF-κB signaling pathway in a mouse model of Parkinson’s disease. Life Sci. 2019, 223, 158–165. [Google Scholar] [CrossRef] [PubMed]
- Caso, J.R.; Pradillo, J.M.; Hurtado, O.; Lorenzo, P.; Moro, M.A.; Lizasoain, I. Toll-like receptor 4 is involved in brain damage and inflammation after experimental stroke. Circulation 2007, 115, 1599–1608. [Google Scholar] [CrossRef] [PubMed]
- Lu, Y.C.; Yeh, W.C.; Ohashi, P.S. LPS/TLR4 signal transduction pathway. Cytokine 2008, 42, 145–151. [Google Scholar] [CrossRef] [PubMed]
- Hanada, T.; Yoshimura, A. Regulation of cytokine signaling and inflammation. Cytokine Growth Factor Rev. 2002, 13, 413–421. [Google Scholar] [CrossRef]
- Shabab, T.; Khanabdali, R.; Moghadamtousi, S.Z.; Kadir, H.A.; Mohan, G. Neuroinflammation pathways: a general review. Int J. Neurosci 2017, 127, 624–633. [Google Scholar] [CrossRef]
- Kawai, T.; Akira, S. Signaling to NF-kappaB by Toll-like receptors. Trends Mol. Med. 2007, 13, 460–469. [Google Scholar] [CrossRef]
- Qian, Y.; Liu, C.; Hartupee, J.; Altuntas, C.Z.; Gulen, M.F.; Jane-Wit, D.; Xiao, J.; Lu, Y.; Giltiay, N.; Liu, J.; et al. The adaptor Act1 is required for interleukin 17-dependent signaling associated with autoimmune and inflammatory disease. Nat. Immunol 2007, 8, 247–256. [Google Scholar] [CrossRef]
- Colombo, E.; Di Dario, M.; Capitolo, E.; Chaabane, L.; Newcombe, J.; Martino, G.; Farina, C. Fingolimod may support neuroprotection via blockade of astrocyte nitric oxide. Ann. Neurol. 2014, 76, 325–337. [Google Scholar] [CrossRef]
- Mayo, L.; Trauger, S.A.; Blain, M.; Nadeau, M.; Patel, B.; Alvarez, J.I.; Mascanfroni, I.D.; Yeste, A.; Kivisäkk, P.; Kallas, K.; et al. Regulation of astrocyte activation by glycolipids drives chronic CNS inflammation. Nat. Med. 2014, 20, 1147–1156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Füchtbauer, L.; Groth-Rasmussen, M.; Holm, T.H.; Løbner, M.; Toft-Hansen, H.; Khorooshi, R.; Owens, T. Angiotensin II Type 1 receptor (AT1) signaling in astrocytes regulates synaptic degeneration-induced leukocyte entry to the central nervous system. Brain Behav. Immun. 2011, 25, 897–904. [Google Scholar] [CrossRef] [PubMed]
- Brambilla, R.; Bracchi-Ricard, V.; Hu, W.H.; Frydel, B.; Bramwell, A.; Karmally, S.; Green, E.J.; Bethea, J.R. Inhibition of astroglial nuclear factor kappaB reduces inflammation and improves functional recovery after spinal cord injury. J. Exp. Med. 2005, 202, 145–156. [Google Scholar] [CrossRef] [PubMed]
- Fu, E.S.; Zhang, Y.P.; Sagen, J.; Candiotti, K.A.; Morton, P.D.; Liebl, D.J.; Bethea, J.R.; Brambilla, R. Transgenic inhibition of glial NF-kappa B reduces pain behavior and inflammation after peripheral nerve injury. Pain 2010, 148, 509–518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Youssef, M.; Ibrahim, A.; Akashi, K.; Hossain, M.S. PUFA-Plasmalogens Attenuate the LPS-Induced Nitric Oxide Production by Inhibiting the NF-kB, p38 MAPK and JNK Pathways in Microglial Cells. Neuroscience 2019, 397, 18–30. [Google Scholar] [CrossRef] [PubMed]
- Yan, Z.; Gibson, S.A.; Buckley, J.A.; Qin, H.; Benveniste, E.N. Role of the JAK/STAT signaling pathway in regulation of innate immunity in neuroinflammatory diseases. Clin. Immunol. 2018, 189, 4–13. [Google Scholar] [CrossRef] [PubMed]
- Yeung, Y.T.; Aziz, F.; Guerrero-Castilla, A.; Arguelles, S. Signaling Pathways in Inflammation and Anti-inflammatory Therapies. Curr. Pharm Des. 2018, 24, 1449–1484. [Google Scholar] [CrossRef]
- Meares, G.P.; Liu, Y.; Rajbhandari, R.; Qin, H.; Nozell, S.E.; Mobley, J.A.; Corbett, J.A.; Benveniste, E.N. PERK-dependent activation of JAK1 and STAT3 contributes to endoplasmic reticulum stress-induced inflammation. Mol. Cell Biol. 2014, 34, 3911–3925. [Google Scholar] [CrossRef]
- Chen, H.; O’Reilly, E.J.; Schwarzschild, M.A.; Ascherio, A. Peripheral inflammatory biomarkers and risk of Parkinson’s disease. Am. J. Epidemiol. 2008, 167, 90–95. [Google Scholar] [CrossRef]
- Lee, E.J.; Woo, M.S.; Moon, P.G.; Baek, M.C.; Choi, I.Y.; Kim, W.K.; Junn, E.; Kim, H.S. Alpha-synuclein activates microglia by inducing the expressions of matrix metalloproteinases and the subsequent activation of protease-activated receptor-1. J. Immunol. 2010, 185, 615–623. [Google Scholar] [CrossRef]
- Qin, H.; Buckley, J.A.; Li, X.; Liu, Y.; Fox, T.H.; Meares, G.P.; Yu, H.; Yan, Z.; Harms, A.S.; Li, Y.; et al. Inhibition of the JAK/STAT Pathway Protects Against α-Synuclein-Induced Neuroinflammation and Dopaminergic Neurodegeneration. J. Neurosci. 2016, 36, 5144–5159. [Google Scholar] [CrossRef] [PubMed]
- Huang, Z.; Wang, Y.; Hu, G.; Zhou, J.; Mei, L.; Xiong, W.C. YAP Is a Critical Inducer of SOCS3, Preventing Reactive Astrogliosis. Cereb. Cortex 2016, 26, 2299–2310. [Google Scholar] [CrossRef] [PubMed]
- Piccolo, S.; Dupont, S.; Cordenonsi, M. The biology of YAP/TAZ: hippo signaling and beyond. Physiol. Rev. 2014, 94, 1287–1312. [Google Scholar] [CrossRef] [PubMed]
- Blunt, J.W.; Carroll, A.R.; Copp, B.R.; Davis, R.A.; Keyzers, R.A.; Prinsep, M.R. Marine natural products. Nat. Prod. Rep. 2018, 35, 8–53. [Google Scholar] [CrossRef] [Green Version]
- Correa, I.; Drews, P.; Botelho, S.; de Souza, M.S.; Tavano, V.M. Deep Learning for Microalgae Classification. In Proceedings of the 16th IEEE International Conference on Machine Learning and Applications (ICMLA), Cancun, Mexico, 18–21 December 2017. [Google Scholar] [CrossRef]
- Peng, Y.; Hu, J.; Yang, B.; Lin, X.-P.; Zhou, X.-F.; Yang, X.-W.; Liu, Y. Chemical composition of seaweeds, Seaweed Sustainability: Food and Non-Food Applications; Academic Press: Salt Lake City, USA, 2015; pp. 79–124. [Google Scholar]
- Goo, B.G.; Baek, G.; Choi, D.J.; Park, Y.I.; Synytsya, A.; Bleha, R.; Seong, D.H.; Lee, C.G.; Park, J.K. Characterization of a renewable extracellular polysaccharide from defatted microalgae Dunaliella tertiolecta. Bioresour. Technol. 2013, 129, 343–350. [Google Scholar] [CrossRef] [PubMed]
- Kurniawati, H.A.; Ismadji, S.; Liu, J.C. Microalgae harvesting by flotation using natural saponin and chitosan. Bioresour. Technol. 2014, 166, 429–434. [Google Scholar] [CrossRef] [Green Version]
- Zhou, R.; Shi, X.Y.; Bi, D.C.; Fang, W.S.; Wei, G.B.; Xu, X. Alginate-Derived Oligosaccharide Inhibits Neuroinflammation and Promotes Microglial Phagocytosis of β-Amyloid. Mar. Drugs 2015, 13, 5828–5846. [Google Scholar] [CrossRef]
- O’Sullivan, L.; Murphy, B.; McLoughlin, P.; Duggan, P.; Lawlor, P.G.; Hughes, H.; Gardiner, G.E. Prebiotics from marine macroalgae for human and animal health applications. Mar. Drugs 2010, 8, 2038–2064. [Google Scholar] [CrossRef]
- Rea, K.; Dinan, T.G.; Cryan, J.F. The microbiome: A key regulator of stress and neuroinflammation. Neurobiol. Stress 2016, 4, 23–33. [Google Scholar] [CrossRef] [Green Version]
- Wang, H.D.; Chen, C.C.; Huynh, P.; Chang, J.S. Exploring the potential of using algae in cosmetics. Bioresour. Technol. 2015, 184, 355–362. [Google Scholar] [CrossRef]
- Yun, E.J.; Choi, I.G.; Kim, K.H. Red macroalgae as a sustainable resource for bio-based products. Trends Biotechnol. 2015, 33, 247–249. [Google Scholar] [CrossRef] [PubMed]
- Chen, H.; Yan, X.; Zhu, P.; Lin, J. Antioxidant activity and hepatoprotective potential of agaro-oligosaccharides in vitro and in vivo. Nutr. J. 2006, 5, 31. [Google Scholar] [CrossRef] [PubMed]
- Enoki, T.; Okuda, S.; Kudo, Y.; Takashima, F.; Sagawa, H.; Kato, I. Oligosaccharides from agar inhibit pro-inflammatory mediator release by inducing heme oxygenase 1. Biosci. Biotechnol. Biochem. 2010, 74, 766–770. [Google Scholar] [CrossRef] [PubMed]
- Bin, B.H.; Kim, S.T.; Bhin, J.; Lee, T.R.; Cho, E.G. The Development of Sugar-Based Anti-Melanogenic Agents. Int. J. Mol. Sci 2016, 17, 583. [Google Scholar] [CrossRef] [PubMed]
- Fernando, I.P.S.; Sanjeewa, K.K.A.; Samarakoon, K.W.; Lee, W.W.; Kim, H.S.; Kang, N.; Ranasinghe, P.; Lee, H.S.; Jeon, Y.J. A fucoidan fraction purified from Chnoospora minima; a potential inhibitor of LPS-induced inflammatory responses. Int. J. Biol Macromol. 2017, 104, 1185–1193. [Google Scholar] [CrossRef] [PubMed]
- Raposo, M.F.; de Morais, R.M.; Bernardo de Morais, A.M. Bioactivity and applications of sulphated polysaccharides from marine microalgae. Mar. Drugs 2013, 11, 233–252. [Google Scholar] [CrossRef] [PubMed]
- Ho, S.H.; Chen, C.Y.; Chang, J.S. Effect of light intensity and nitrogen starvation on CO2 fixation and lipid/carbohydrate production of an indigenous microalga Scenedesmus obliquus CNW-N. Bioresour. Technol. 2012, 113, 244–252. [Google Scholar] [CrossRef]
- Cheng, Y.-S.; Zheng, Y.; Labavitch, J.M.; VanderGheynst, J.S. The impact of cell wall carbohydrate composition on the chitosan flocculation of Chlorella. Process. Biochem. 2011, 46, 1927–1933. [Google Scholar] [CrossRef]
- Kumari, P.; Kumar, M.; Reddy, C.R.K. Algal lipids, fatty acids and sterols. Functional ingredients from algae for foods and nutraceuticals; Woodhead Publishing: Cambridge, UK, 2013; pp. 87–134. [Google Scholar]
- Manuelli, M.; Della Guardia, L.; Cena, H. Enriching Diet with n-3 PUFAs to Help Prevent Cardiovascular Diseases in Healthy Adults: Results from Clinical Trials. Int. J. Mol. Sci 2017, 18, 1552. [Google Scholar] [CrossRef]
- Layé, S.; Nadjar, A.; Joffre, C.; Bazinet, R.P. Anti-Inflammatory Effects of Omega-3 Fatty Acids in the Brain: Physiological Mechanisms and Relevance to Pharmacology. Pharm. Rev. 2018, 70, 12–38. [Google Scholar] [CrossRef]
- Van Ginneken, V.J.; Helsper, J.P.; de Visser, W.; van Keulen, H.; Brandenburg, W.A. Polyunsaturated fatty acids in various macroalgal species from North Atlantic and tropical seas. Lipids Health Dis. 2011, 10, 104. [Google Scholar] [CrossRef] [PubMed]
- Santos, M.A.Z.; Colepicolo, P.; Pupo, D.; Fujii, M.T.; de Pereira, C.M.P.; Mesko, M.F. Antarctic red macroalgae: a source of polyunsaturated fatty acids. J. Appl. Phycol. 2017, 29, 759–767. [Google Scholar] [CrossRef]
- Jung, H.A.; Jin, S.E.; Ahn, B.R.; Lee, C.M.; Choi, J.S. Anti-inflammatory activity of edible brown alga Eisenia bicyclis and its constituents fucosterol and phlorotannins in LPS-stimulated RAW264.7 macrophages. Food Chem. Toxicol. 2013, 59, 199–206. [Google Scholar] [CrossRef] [PubMed]
- Sun, Y.; Lin, Y.; Cao, X.; Xiang, L.; Qi, J. Sterols from Mytilidae show anti-aging and neuroprotective effects via anti-oxidative activity. Int. J. Mol. Sci 2014, 15, 21660–21673. [Google Scholar] [CrossRef] [PubMed]
- Chen, H.; Han, C.; Wu, J.; Liu, X.; Zhan, Y.; Chen, J.; Chen, Y.; Gu, R.; Zhang, L.; Chen, S.; et al. Accessible Method for the Development of Novel Sterol Analogues with Dipeptide-like Side Chains That Act as Neuroinflammation Inhibitors. ACS Chem. Neurosci. 2016, 7, 305–315. [Google Scholar] [CrossRef] [PubMed]
- Harnedy, P.A.; FitzGerald, R.J. BIOACTIVE PROTEINS, PEPTIDES, AND AMINO ACIDS FROM MACROALGAE(1). J. Phycol. 2011, 47, 218–232. [Google Scholar] [CrossRef]
- Aydın, A.F.; Çoban, J.; Doğan-Ekici, I.; Betül-Kalaz, E.; Doğru-Abbasoğlu, S.; Uysal, M. Carnosine and taurine treatments diminished brain oxidative stress and apoptosis in D-galactose aging model. Metab Brain Dis. 2016, 31, 337–345. [Google Scholar] [CrossRef]
- Korhonen, H.; Pihlanto, A. Bioactive peptides: production and functionality. Int. Dairy J. 2006, 16, 945–960. [Google Scholar] [CrossRef]
- Vanderlei, E.S.; Patoilo, K.K.; Lima, N.A.; Lima, A.P.; Rodrigues, J.A.; Silva, L.M.; Lima, M.E.; Lima, V.; Benevides, N.M. Antinociceptive and anti-inflammatory activities of lectin from the marine green alga Caulerpa cupressoides. Int. Immunopharmacol. 2010, 10, 1113–1118. [Google Scholar] [CrossRef]
- Silva, L.M.; Lima, V.; Holanda, M.L.; Pinheiro, P.G.; Rodrigues, J.A.; Lima, M.E.; Benevides, N.M. Antinociceptive and anti-inflammatory activities of lectin from marine red alga Pterocladiella capillacea. Biol. Pharm Bull. 2010, 33, 830–835. [Google Scholar] [CrossRef]
- Coelho, L.C.; Silva, P.M.; Lima, V.L.; Pontual, E.V.; Paiva, P.M.; Napoleão, T.H.; Correia, M.T. Lectins, Interconnecting Proteins with Biotechnological/Pharmacological and Therapeutic Applications. Evid Based Complement. Altern. Med. 2017, 2017, 1594074. [Google Scholar] [CrossRef]
- Fan, X.; Bai, L.; Zhu, L.; Yang, L.; Zhang, X. Marine algae-derived bioactive peptides for human nutrition and health. J. Agric. Food Chem. 2014, 62, 9211–9222. [Google Scholar] [CrossRef] [PubMed]
- Eom, S.H.; Kim, Y.M.; Kim, S.K. Antimicrobial effect of phlorotannins from marine brown algae. Food Chem. Toxicol 2012, 50, 3251–3255. [Google Scholar] [CrossRef] [PubMed]
- Corona, G.; Coman, M.M.; Guo, Y.; Hotchkiss, S.; Gill, C.; Yaqoob, P.; Spencer, J.P.E.; Rowland, I. Effect of simulated gastrointestinal digestion and fermentation on polyphenolic content and bioactivity of brown seaweed phlorotannin-rich extracts. Mol. Nutr. Food Res. 2017, 61. [Google Scholar] [CrossRef] [PubMed]
- Montero, L.; del Pilar Sánchez-Camargo, A.; Ibáñez, E.; Gilbert-López, B. Phenolic Compounds from Edible Algae: Bioactivity and Health Benefits. Curr. Med. Chem. 2018, 25, 4808–4826. [Google Scholar] [CrossRef] [PubMed]
- Sivagnanam, S.P.; Yin, S.; Choi, J.H.; Park, Y.B.; Woo, H.C.; Chun, B.S. Biological Properties of Fucoxanthin in Oil Recovered from Two Brown Seaweeds Using Supercritical CO2 Extraction. Mar. Drugs 2015, 13, 3422–3442. [Google Scholar] [CrossRef] [PubMed]
- Mikami, K.; Hosokawa, M. Biosynthetic pathway and health benefits of fucoxanthin, an algae-specific xanthophyll in brown seaweeds. Int. J. Mol. Sci. 2013, 14, 13763–13781. [Google Scholar] [CrossRef] [PubMed]
- Peng, J.; Yuan, J.P.; Wu, C.F.; Wang, J.H. Fucoxanthin, a marine carotenoid present in brown seaweeds and diatoms: metabolism and bioactivities relevant to human health. Mar. Drugs 2011, 9, 1806–1828. [Google Scholar] [CrossRef] [PubMed]
- Gateau, H.; Solymosi, K.; Marchand, J.; Schoefs, B. Carotenoids of Microalgae Used in Food Industry and Medicine. Mini. Rev. Med. Chem. 2017, 17, 1140–1172. [Google Scholar] [CrossRef]
- Eggersdorfer, M.; Wyss, A. Carotenoids in human nutrition and health. Arch. Biochem. Biophys 2018, 652, 18–26. [Google Scholar] [CrossRef]
- Pangestuti, R.; Vo, T.S.; Ngo, D.H.; Kim, S.K. Fucoxanthin ameliorates inflammation and oxidative reponses in microglia. J. Agric. Food Chem. 2013, 61, 3876–3883. [Google Scholar] [CrossRef]
- Kim, Y.H.; Koh, H.K.; Kim, D.S. Down-regulation of IL-6 production by astaxanthin via ERK-, MSK-, and NF-κB-mediated signals in activated microglia. Int. Immunopharmacol. 2010, 10, 1560–1572. [Google Scholar] [CrossRef] [PubMed]
- Gaysinski, M.; Ortalo-Magné, A.; Thomas, O.P.; Culioli, G. Extraction, Purification, and NMR Analysis of Terpenes from Brown Algae. Methods Mol. Biol. 2015, 1308, 207–223. [Google Scholar] [CrossRef] [PubMed]
- Yang, P.; Liu, D.Q.; Liang, T.J.; Li, J.; Zhang, H.Y.; Liu, A.H.; Guo, Y.W.; Mao, S.C. Bioactive constituents from the green alga Caulerpa racemosa. Bioorg. Med. Chem. 2015, 23, 38–45. [Google Scholar] [CrossRef] [PubMed]
- Kirkley, K.S.; Popichak, K.A.; Afzali, M.F.; Legare, M.E.; Tjalkens, R.B. Microglia amplify inflammatory activation of astrocytes in manganese neurotoxicity. J. Neuroinflammation 2017, 14, 99. [Google Scholar] [CrossRef] [PubMed]
- Heneka, M.T.; Carson, M.J.; El Khoury, J.; Landreth, G.E.; Brosseron, F.; Feinstein, D.L.; Jacobs, A.H.; Wyss-Coray, T.; Vitorica, J.; Ransohoff, R.M.; et al. Neuroinflammation in Alzheimer’s disease. Lancet Neurol. 2015, 14, 388–405. [Google Scholar] [CrossRef]
- Pangestuti, R.; Kim, S.K. Neuroprotective effects of marine algae. Mar. Drugs 2011, 9, 803–818. [Google Scholar] [CrossRef]
- Lee, J.Y.; Lee, M.S.; Choi, H.J.; Choi, J.W.; Shin, T.; Woo, H.C.; Kim, J.I.; Kim, H.R. Hexane fraction from Laminaria japonica exerts anti-inflammatory effects on lipopolysaccharide-stimulated RAW 264.7 macrophages via inhibiting NF-kappaB pathway. Eur. J. Nutr. 2013, 52, 409–421. [Google Scholar] [CrossRef]
- Khan, M.N.; Lee, M.C.; Kang, J.Y.; Park, N.G.; Fujii, H.; Hong, Y.K. Effects of the brown seaweed Undaria pinnatifida on erythematous inflammation assessed using digital photo analysis. Phytother. Res. 2008, 22, 634–639. [Google Scholar] [CrossRef]
- Kang, J.Y.; Khan, M.N.; Park, N.H.; Cho, J.Y.; Lee, M.C.; Fujii, H.; Hong, Y.K. Antipyretic, analgesic, and anti-inflammatory activities of the seaweed Sargassum fulvellum and Sargassum thunbergii in mice. J. Ethnopharmacol. 2008, 116, 187–190. [Google Scholar] [CrossRef]
- Kim, S.; Kim, J.I.; Choi, J.W.; Kim, M.; Yoon, N.Y.; Choi, C.G.; Choi, J.S.; Kim, H.R. Anti-inflammatory effect of hexane fraction from Myagropsis myagroides ethanolic extract in lipopolysaccharide-stimulated BV-2 microglial cells. J. Pharm. Pharm. 2013, 65, 895–906. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.; Lee, M.S.; Lee, B.; Gwon, W.G.; Joung, E.J.; Yoon, N.Y.; Kim, H.R. Anti-inflammatory effects of sargachromenol-rich ethanolic extract of Myagropsis myagroides on lipopolysaccharide-stimulated BV-2 cells. Bmc Complement. Altern Med. 2014, 14, 231. [Google Scholar] [CrossRef]
- Aisya, G.S.; Ching, T.S.; Yee, G.S. Antioxidative, Anticholinesterase and Anti-Neuroinflammatory Properties of Malaysian Brown and Green Seaweeds. Int. J. Ind. Manuf. Eng. 2014, 8, 895–906. [Google Scholar]
- Craig, L.A.; Hong, N.S.; McDonald, R.J. Revisiting the cholinergic hypothesis in the development of Alzheimer’s disease. Neurosci. Biobehav. Rev. 2011, 35, 1397–1409. [Google Scholar] [CrossRef] [PubMed]
- Hoong, W.C.; John, I.; Yee, G.S.; Ling, C.E.W.; Ching, T.S.; Moi, P.S.; Aisya, G.S.; Tiong, Y.; Irvine, G.A. Fucosterol inhibits the cholinesterase activities and reduces the release of pro-inflammatory mediators in lipopolysaccharide and amyloid-induced microglial cells. J. Appl. Phycol. 2018, 30, 3261–3270. [Google Scholar]
- Jung, W.K.; Heo, S.J.; Jeon, Y.J.; Lee, C.M.; Park, Y.M.; Byun, H.G.; Choi, Y.H.; Park, S.G.; Choi, I.W. Inhibitory effects and molecular mechanism of dieckol isolated from marine brown alga on COX-2 and iNOS in microglial cells. J. Agric. Food Chem. 2009, 57, 4439–4446. [Google Scholar] [CrossRef] [PubMed]
- Lee, S.; Youn, K.; Kim, D.H.; Ahn, M.R.; Yoon, E.; Kim, O.Y.; Jun, M. Anti-Neuroinflammatory Property of Phlorotannins from Ecklonia cava on Aβ25-35-Induced Damage in PC12 Cells. Mar. Drugs 2019, 17, 7. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef]
- Heo, S.J.; Yoon, W.J.; Kim, K.N.; Ahn, G.N.; Kang, S.M.; Kang, D.H.; Affan, A.; Oh, C.; Jung, W.K.; Jeon, Y.J. Evaluation of anti-inflammatory effect of fucoxanthin isolated from brown algae in lipopolysaccharide-stimulated RAW 264.7 macrophages. Food Chem. Toxicol. 2010, 48, 2045–2051. [Google Scholar] [CrossRef]
- Lin, T.Y.; Lu, C.W.; Wang, S.J. Astaxanthin inhibits glutamate release in rat cerebral cortex nerve terminals via suppression of voltage-dependent Ca2+ entry and mitogen-activated protein kinase signaling pathway. J. Agric. Food Chem. 2010, 58, 8271–8278. [Google Scholar] [CrossRef]
- Wu, H.; Niu, H.; Shao, A.; Wu, C.; Dixon, B.J.; Zhang, J.; Yang, S.; Wang, Y. Astaxanthin as a Potential Neuroprotective Agent for Neurological Diseases. Mar. Drugs 2015, 13, 5750–5766. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wu, D.; Xu, H.; Chen, J.; Zhang, L. Effects of Astaxanthin Supplementation on Oxidative Stress. Int. J. Vitam. Nutr. Res. 2019, 1–16. [Google Scholar] [CrossRef] [PubMed]
- Balietti, M.; Giannubilo, S.R.; Giorgetti, B.; Solazzi, M.; Turi, A.; Casoli, T.; Ciavattini, A.; Fattorettia, P. The effect of astaxanthin on the aging rat brain: gender-related differences in modulating inflammation. J. Sci. Food Agric. 2016, 96, 4295. [Google Scholar] [CrossRef] [PubMed]
- Krogh, J.; Benros, M.E.; Jørgensen, M.B.; Vesterager, L.; Elfving, B.; Nordentoft, M. The association between depressive symptoms, cognitive function, and inflammation in major depression. Brain Behav. Immun. 2014, 35, 70–76. [Google Scholar] [CrossRef] [PubMed]
- Miller, A.H.; Raison, C.L. The role of inflammation in depression: from evolutionary imperative to modern treatment target. Nat. Rev. Immunol. 2015, 16, 22–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, X.S.; Zhang, X.; Wu, Q.; Li, W.; Wang, C.X.; Xie, G.B.; Zhou, X.M.; Shi, J.X.; Zhou, M.L. Astaxanthin offers neuroprotection and reduces neuroinflammation in experimental subarachnoid hemorrhage. J. Surg. Res. 2014, 192, 206–213. [Google Scholar] [CrossRef] [PubMed]
- Lee, J.C.; Hou, M.F.; Huang, H.W.; Chang, F.R.; Yeh, C.C.; Tang, J.Y.; Chang, H.W. Marine algal natural products with anti-oxidative, anti-inflammatory, and anti-cancer properties. Cancer Cell Int. 2013, 13, 55. [Google Scholar] [CrossRef]
- Choi, J.Y.; Hwang, C.J.; Lee, H.P.; Kim, H.S.; Han, S.B.; Hong, J.T. Inhibitory effect of ethanol extract of Nannochloropsis oceanica on lipopolysaccharide-induced neuroinflammation, oxidative stress, amyloidogenesis and memory impairment. Oncotarget 2017, 8, 45517–45530. [Google Scholar] [CrossRef] [Green Version]
- Xiang, Y.; Meng, S.; Wang, J.; Li, S.; Liu, J.; Li, H.; Li, T.; Song, W.; Zhou, W. Two novel DNA motifs are essential for BACE1 gene transcription. Sci Rep. 2014, 4, 6864. [Google Scholar] [CrossRef] [Green Version]
- Smith, A.M.; Gibbons, H.M.; Dragunow, M. Valproic acid enhances microglial phagocytosis of amyloid-beta(1-42). Neuroscience 2010, 169, 505–515. [Google Scholar] [CrossRef]
- Guo, X.; Xin, X.; Gan, L.; Nie, Q.; Geng, M. Determination of the accessibility of acidic oligosaccharide sugar chain to blood-brain barrier using surface plasmon resonance. Biol. Pharm. Bull. 2006, 29, 60–63. [Google Scholar] [CrossRef] [PubMed]
- 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. 2012, 18, 827–833. [Google Scholar] [CrossRef] [PubMed]
- Rafiquzzaman, S.M.; Kim, E.Y.; Lee, J.M.; Mohibbullah, M.; Alam, M.B.; Moon, I.S.; Kim, J.-M.; Kong, I.-S. Anti-Alzheimers and anti-inflammatory activities of a glycoprotein purified from the edible brown alga Undaria pinnatifida. Food Res. Int. 2015, 77, 118–124. [Google Scholar]
- Ryan, S.; O’Gorman, D.M.; Nolan, Y.M. Evidence that the marine-derived multi-mineral Aquamin has anti-inflammatory effects on cortical glial-enriched cultures. Phytother. Res. 2011, 25, 765–767. [Google Scholar] [CrossRef] [PubMed]
Carbohydrates | Brown Macroalgae | Red Macroalgae | Green Macroalgae |
---|---|---|---|
monosaccharides | glucose, galactose, xylose, fucose, uronic acid, glucuronic acid mannuronic acid, guluronic acid | glucose, galactose, mannose | glucose, mannose, xylose, rhamnose, glucuronic acid, uronic acid |
polysaccharides | laminarin alginate, fucoidan (sulphated), cellulose, mannitol | carrageenans (sulfated), agar (sulfated), floridean starch, cellulose, lignin, funoran | ulvan (sulfated), mannan, galactans (sulfated), xylans, floridean starch, cellulose, lignin |
Marine Algae Extract/Bioactive Compound | Treatment Conc. | Experimental Model | Key Findings |
---|---|---|---|
Ulva conglobata methanol extract | 10‒50 µg/mL | mouse hippocampal HT-22 cells; mouse microglial BV-2 cells | Restoration of cellular viability in HT-22 cells; downregulation of COX-2 and iNOS in BV-2 cells [22] |
Exane fraction of Myagropsis myagroides ethanolic extract | 5‒25 µg/mL | mouse microglial BV-2 cells | Decreased release of inflammatory cytokines, inactivation of NF-κB and reduced mRNA and protein levels of iNOS and COX-2 [124] |
Myagropsis myagroides ethanolic extract | 5–25 µg/mL | mouse microglial BV-2 cells | Reduction in NO, PGE2, IL-6, IL-1β and TNF-α release; inhibition of ERKs-JNKs/NF-κB axis [125] |
Padina australis, Sargassum polycystum and Caulerpa racemosa extracts | 0.05–0.4 mg/mL | mouse microglial C8-B4 cells | Decreased release of pro-inflammatory mediators (NO, PGE2, IL-6, IL-1β and TNF-α) [126] |
Fucosterol from Padina australis | 0.004–192 µM | mouse microglial C8-B4 and BV-2 cells | Inhibition of AChE and BChE; reduction in release of NO, PGE2, IL-6, IL-1β and TNF-α in LPS-stimulated C8-B4 cells; prevented production of NO, IL-6 and TNF-α in Aβ42-stimulated BV-2 cells [128] |
Eckol, dieckol and 8,8’-bieckol from Ecklonia cava | 1–50 µM | rat neuronal PC12 cells | Antioxidant activity; anti-apoptotic effects; decrease in key inflammatory proteins (COX-2, iNOS, IL-1β and TNF-α) [130] |
Dieckol from Ecklonia cava | 50–300 µg/mL | mouse microglial BV-2 cells | Inhibition of LPS-induced iNOS and COX-2 protein and mRNA expression; suppression of p-38/ NF-κB pathway; ROS scavenging activity [129] |
Phlorofucofuroeckol B from Ecklonia stolonifera | 10–40 µM | mouse microglial BV-2 cells | Inhibition of IκB-α/NF-κB and Akt/ERK/JNK pathways [23] |
Floridoside from Laurencia undulata | 1–50 µM | mouse microglial BV-2 cells | Inhibition of LPS-induced NO and ROS production; downregulation of COX-2 and iNOS mRNA and protein levels by reducing p38 and ERK phosphorylation [131] |
Fucoxanthin | 5–50 µM | mouse microglial BV-2 cells | Attenuation of Aβ42-induced cytokines release (NO, PGE2, IL-6, IL-1β and TNF-α) and enzymes upregulation (COX-2, iNOS) by suppressing MAPKs phosphorylation; protection from H2O2-induced ROS release and DNA damage by recovering antioxidant enzymes [114] |
Astaxanthin | 20–80 mg/Kg | male ICR mice | Reversed LPS-induced depressive-like behaviors; attenuation of cytokines level (IL-6, IL-1β and TNF-α) and antagonization of iNOS, nNOS and COX-2 expression in the hippocampus and prefrontal cortex [24] |
Astaxanthin | 75 mg/Kg | male Sprague‒Dawley rats | Amelioration in cerebral edema, blood‒brain barrier disruption, neurological dysfunction and neuronal degeneration after the induction of subarachnoid hemorrhage; downregulation of NF-κB activity, and intercellular adhesion molecule-1, IL-1β and TNF-α expression [139] |
AquaminTM | 0.05–2 mg/mL | cortical glial-enriched cultures from Sprague‒Dawley rat pups | Attenuation of LPS-induced IL-1β and TNF-α secretion [147] |
Nannochloropsis oceanica ethanol extract | 50–100 mg/Kg | male ICR mice | Decrease of ROS and malondialdehyde levels; improvement of LPS-induced memory impairment; suppression of Aβ42 generation by downregulating APP and BACE1 expression [141] |
Alginate-derived oligosaccharide | 50–500 µg/mL | mouse microglial BV-2 cells | Inhibition of LPS/ Aβ42-induced NO and PGE2 production, COX-2 and iNOS expression, and cytokines secretion; attenuation of TLR4 and NF-κB overexpression; promotion of Aβ phagocytosis [79] |
Seleno-polymannuronate | 0.5 mg/mL, 0.8 mg/mL | primary microglia and astrocytes from BALB/c mouse pups; female BALB/c mice | In LPS-activated primary cells, attenuation of NF-κB and MAPK signaling with the reduction of NO, PGE2 production, downregulation of COX-2 and iNOS expression, and IL-6, IL-1β and TNF-α secretion; decrease of Iba1- and GFAP-positive cells in the brain of a mouse model of LPS-induced inflammation [13] |
Fucoidan | 7.5 mg/Kg, 15 mg/Kg; 31.25–125 µg/mL | male Sprague‒Dawley rats; primary microglia from neonatal Sprague–Dawley rats | Improvement of behavioral deficits and prevention of dopaminergic neuron loss; inhibition of ROS and TNF-α release [145] |
Glycoprotein from Undaria pinnatifida | 5–45 µg/mL | primary hippocampal cells from embryonal Sprague–Dawley rats | Inhibition of AChE, BChE and BACE1; promotion of cell survival and neurite extension [146] |
© 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Barbalace, M.C.; Malaguti, M.; Giusti, L.; Lucacchini, A.; Hrelia, S.; Angeloni, C. Anti-Inflammatory Activities of Marine Algae in Neurodegenerative Diseases. Int. J. Mol. Sci. 2019, 20, 3061. https://doi.org/10.3390/ijms20123061
Barbalace MC, Malaguti M, Giusti L, Lucacchini A, Hrelia S, Angeloni C. Anti-Inflammatory Activities of Marine Algae in Neurodegenerative Diseases. International Journal of Molecular Sciences. 2019; 20(12):3061. https://doi.org/10.3390/ijms20123061
Chicago/Turabian StyleBarbalace, Maria Cristina, Marco Malaguti, Laura Giusti, Antonio Lucacchini, Silvana Hrelia, and Cristina Angeloni. 2019. "Anti-Inflammatory Activities of Marine Algae in Neurodegenerative Diseases" International Journal of Molecular Sciences 20, no. 12: 3061. https://doi.org/10.3390/ijms20123061
APA StyleBarbalace, M. C., Malaguti, M., Giusti, L., Lucacchini, A., Hrelia, S., & Angeloni, C. (2019). Anti-Inflammatory Activities of Marine Algae in Neurodegenerative Diseases. International Journal of Molecular Sciences, 20(12), 3061. https://doi.org/10.3390/ijms20123061