AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (749.1 KB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Review Article | Open Access

Potential of peptides and phytochemicals in attenuating different phases of islet amyloid polypeptide fibrillation for type 2 diabetes management

Raliat O. AbioyeaChibuike C. Udenigwea,b( )
Department of Chemistry and Biomolecular Sciences, Faculty of Science, University of Ottawa, Ottawa, Ontario, K1N 6N5, Canada
School of Nutrition Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario, K1H 8M5, Canada

Peer review under responsibility of KeAi Communications Co., Ltd

Show Author Information

Abstract

Islet amyloid polypeptide (IAPP), or amylin, has been identified as a key factor in the development of type 2 diabetes (T2D). IAPP aggregates, which form amyloid fibrils, contribute to cytotoxicity of the pancreatic β-cells, resulting in loss of function and subsequent reduction in insulin production. As a result, surviving β-cells overcompensate for this reduction of insulin production, further contributing to the loss of function because of increased stress, thus leading to insulin resistance. Endogenously, IAPP monomers function in a variety of roles; however, aggregation renders them non-functional. The use of naturally occurring compounds, including peptides and phytochemicals, has been explored as a way to mitigate or inhibit IAPP fibril formation. This review discusses the structure, endogenous roles and mechanism of IAPP fibril formation, recent advances on inhibitors of IAPP fibril formation, and new insights on the future development and application of food-derived inhibitors towards T2D management.

References

[1]

P.J. Marek, V. Patsalo, D.F. Green, et al., Ionic strength effects on amyloid formation by amylin are a complicated interplay among Debye screening, ion selectivity, and Hofmeister effects, Biochemistry 51 (2012) 8478-8490. https://doi.org/10.1021/bi300574r

[2]

M. Christensen, K.K. Skeby, B. Schiøtt, Identification of key interactions in the inital self-assembly of amylin in a membrane environment, Biochemistry 56 (2017) 4884-4894. https://doi.org/10.1021/acs.biochem.7b00344

[3]

Z. Qian, Y. Zou, Q. Zhang, et al., Atomistic-level study of the interactions between hIAPP protofibrils and membranes: influence of pH and lipid composition, BBA - Biomembr. 1860 (2018) 1818-1825. https://doi.org/10.1016/j.bbamem.2018.02.005

[4]

Y.H. Hsu, Y.W. Chen, M.H. Wu, et al., Protein glycation by glyoxal promotes amyloid formation by islet amyloid polypeptide, Biophys. J. 116 (2019) 2304-2313. https://doi.org/10.1016/j.bpj.2019.05.013

[5]

P. Cao, A. Abedini, D.P. Raleigh, Aggregation of islet amyloid polypeptide: from physical chemistry to cell biology, Curr. Opin. Struct. Biol. 23 (2013) 82-89. https://doi.org/10.1016/j.sbi.2012.11.003

[6]

S. Chakraborty, B. Mukherjee, S. Basu, Pinpointing proline substitution to be responsible for the loss of amyloidogenesis in IAPP, Chem. Biol. Drug Des. 82 (2013) 446-452. https://doi.org/10.1111/cbdd.12172

[7]

O. Bolarinwa, C. Li, N. Khadka, et al., γ-AApeptides-based small molecule ligands that disaggregate human islet amyloid polypeptide, Sci. Rep. 10 (2020) 1-10. https://doi.org/10.1038/s41598-019-56500-0

[8]

J. Yang, Y. Sun, F. Xu, et al., Silibinin ameliorates amylin-induced pancreatic β-cell apoptosis partly via upregulation of GLP-1R/PKA pathway, Mol. Cell. Biochem. 452 (2019) 83-94. https://doi.org/10.1007/s11010-018-3414-9

[9]

K. Sivanesam, I. Shu, K.N.L. Huggins, et al., Peptide inhibitors of the amyloidogenesis of IAPP: verification of the hairpin-binding geometry hypothesis, FEBS Lett. 590 (16) (2016) 2575-2583. https://doi.org/10.1002/1873-3468.12261

[10]

M. Olsson, M.K. Herrington, R.D. Reidelberger, et al., Food intake and meal pattern in IAPP knockout mice with and without infusion of exogenous IAPP, Scand. J. Gastroenterol. 47 (2012) 191-196. https://doi.org/10.3109/00365521.2011.638392

[11]

D.C. Bhowmick, A. Jeremic, Functional proteasome complex is required for turnover of islet amyloid polypeptide in pancreatic β-cells, J. Biol. Chem. 293 (2018) 14210-14223. https://doi.org/10.1074/jbc.RA118.002414

[12]

M. Press, T. Jung, J. König, et al., Protein aggregates and proteostasis in aging: amylin and β-cell function, Mech. Ageing Dev. 177 (2019) 46-54. https://doi.org/10.1016/j.mad.2018.03.010

[13]

P. Westermark, A. Andersson, G.T. Westermark, Islet amyloid polypeptide, islet amyloid, and diabetes mellitus, Physiol. Rev. 91 (2011) 795-826. https://doi.org/10.1152/physrev.00042.2009

[14]

P. Nedumpully-Govindan, F. Ding, Inhibition of IAPP aggregation by insulin depends on the insulin oligomeric state regulated by zinc ion concentration, Sci. Rep. 5 (2015) 1-7. https://doi.org/10.1038/srep08240

[15]

L.M. Palato, S. Pilcher, A. Oakes, et al., Amyloidogenicity of naturally occurring full-length animal IAPP variants, J. Pept. Sci. 25 (2019) 1-8. https://doi.org/10.1002/psc.3199

[16]

A. Chaari, M. Ladjimi, Human islet amyloid polypeptide (hIAPP) aggregation in type 2 diabetes: correlation between intrinsic physicochemical properties of hIAPP aggregates and their cytotoxicity, Int. J. Biol. Macromol. 136 (2019) 57-65. https://doi.org/10.1016/j.ijbiomac.2019.06.050

[17]

A.V. Matveyenko, T. Gurlo, M. Daval, et al., Successful versus failed adaptation to high-fat diet-induced insulin resistance the role of IAPP-induced-cell endoplasmic reticulum stress, Diabetes 58 (2009) 906-916. https://doi.org/10.2337/db08-1464

[18]

N.A. Alves, L.G. Dias, R.B. Frigori, Synergistic long-range effects of mutations underlie aggregation propensities of amylin analogues, J. Mol. Model. 25 (2019) 1-11. https://doi.org/10.1007/s00894-019-4137-x

[19]

S. Kumar, J. Walter, Phosphorylation of amyloid beta (Aβ) peptides- a trigger for formation of toxic aggregates in Alzheimer's disease, Aging 3 (8) (2011) 1-10. https://doi.org/10.18632/aging.100362

[20]

A.L. Serrano, J.P. Lomont, L.H. Tu, et al., A free energy barrier caused by the refolding of an oligomeric intermediate controls the lag time of amyloid formation by hIAPP, J. Am. Chem. Soc. 139 (2017) 16748-16758. https://doi.org/10.1021/jacs.7b08830

[21]

A. Kapurniotu, J. Bernhagen, N. Greenfield, et al., Contribution of advanced glycosylation to the amyloidogenicity of iselt amyloid polypeptide, Eur. J. Biochem. 251 (1998) 208-216. https://doi.org/10.1046/j.1432-1327.1998.2510208.x

[22]

A. Abedini, J. Derk, A.M. Schmidt, The receptor for advanced glycation endproducts is a mediator of toxicity by IAPP and other proteotoxic aggregates: establishing and exploiting common ground for novel amyloidosis therapies, Protein Sci. 27 (2018) 1166-1180. https://doi.org/10.1002/pro.3425

[23]

E.B. Dunkelberger, L.E. Buchanan, P. Marek, et al., Deamidation accelerates amyloid formatin and alters amylin fiber structure, J. Am. Chem. Soc. 134 (2012) 12658-12667. https://doi.org/10.1021/ja3039486

[24]

P.T. Nguyen, X. Zottig, M. Sebastiao, et al., Role of site-specific asparagine deamidation in islet amyloid polypeptide amyloidogenesis: key contributions of residues 14 and 21, Biochemistry 56 (2017) 3808-3817. https://doi.org/10.1021/acs.biochem.7b00209

[25]

A.R.F. Hoffmann, M.S. Saravanan, O. Lequin, et al., A single mutation on the human amyloid polypeptide modulates fibril growth and affects the mechanism of amyloid-induced membrane damage, BBA - Biomembr. 1860 (2018) 1783-1792. https://doi.org/10.1016/j.bbamem.2018.02.018

[26]

Y. Peretz, R. Malishev, S. Kolusheva, et al., Nanoparticles modulate membrane interactions of human islet amyloid polypeptide (hIAPP), BBA - Biomembr. 1860 (2018) 1810-1817. https://doi.org/10.1016/j.bbamem.2018.03.029

[27]

P. Arosio, M. Vendruscolo, C.M. Dobson, et al., Chemical kinetics for drug discovery to combat protein aggregation diseases, Cell Press 35 (2014) 127-135. https://doi.org/10.1016/j.tips.2013.12.005

[28]

M.F.M. Engel, L. Khemtémourian, C.C. Kleijer, et al., Membrane damage by human islet amyloid polypeptide through fibril growth at the membrane, PNAS 105 (2008) 6033-6038. https://doi.org/10.1073/pnas.0708354105

[29]

Y. Porat, A. Abramowitz, E. Gazit, Inhibition of amyloid fibril formation by polyphenols: structural similarity and aromatic interactions as a common inhibition mechanism, Chem. Biol. Drug Des. 67 (2006) 27-37. https://doi.org/10.1111/j.1747-0285.2005.00318.x

[30]

X. Dong, Q. Qiao, Z. Qian, et al., Recent computational studies of membrane interaction and disruption of human islet amyloid polypeptide: monomers, oligomers and protofibrils, Biochim. Biophys. Acta - Biomembr. 1860 (2018) 1826-1839. https://doi.org/10.1016/j.bbamem.2018.03.006

[31]

J.W.M. Höppener, B. Ahrén, C.J.M. Lips, Islet amyloid and type 2 diabetes mellitus, N. Engl. J. Med. 343 (2000) 411-419. https://doi.org/10.1056/NEJM200008103430607

[32]

E. Thormann, On understanding of the Hofmeister effect: how addition of salt alters the stability of temperature responsive polymers in aqueous solutions, RSC Adv. 2 (2012) 8297-8305. https://doi.org/10.1039/c2ra20164j

[33]

Y. Zhang, P.S. Cremer, Interactions between macromolecules and ions: the Hofmeister series, Curr. Opin. Chem. Biol. 10 (2006) 658-663. https://doi.org/10.1016/j.cbpa.2006.09.020

[34]

A.I. Ilitchev, M.J. Giammona, J.N. Schwarze, et al., Zinc-induced conformational transitions in human islet amyloid polypeptide and their role in the inhibition of amyloidosis, J. Phys. Chem. 122 (2018) 9852-9859. https://doi.org/10.1021/acs.jpcb.8b06206

[35]

S. Salamekh, J.R. Brender, S.J. Hyung, et al., A two-site mechanism for the inhibition of IAPP amyloidogenesis by zinc, J. Mol. Biol. 410 (2011) 294-306. https://doi.org/10.1016/j.jmb.2011.05.015

[36]

S.J.C. Lee, T.S. Choi, J.W. Lee, et al., Structure and assembly mechanisms of toxic human islet amyloid polypeptide oligomers associated with copper, Chem. Sci. 7 (2016) 5398-5406. https://doi.org/10.1039/C6SC00153J

[37]

M. Algrably, I. Czaban, Ł. Jaremko, et al., Interaction of amylin species with transition metals and membranes, J. Inorg. Biochem. 191 (2019) 69-76. https://doi.org/10.1016/j.jinorgbio.2018.11.004

[38]

D. Zhu, G. Gong, W. Wang, et al., Disaggregation of human islet amyloid polypeptide fibril formation by ruthenium polypyridyl complexes, J. Inorg. Biochem. 170 (2017) 109-116. https://doi.org/10.1016/j.jinorgbio.2017.02.008

[39]

X. Su, K. Wang, N. Liu, et al., All-atom structure ensembles of islet amyloid polypeptides determined by enhanced sampling and experiment data restraints, Proteins 87 (2019) 541-550. https://doi.org/10.1002/prot.25677

[40]

B.N. Ratha, R.K. Kar, S. Kalita, et al., Sequence specificity of amylin-insulin interaction: a fragment-based insulin fibrillation inhibition study, Biochim. Biophys. Acta Proteins Proteom. 1867 (2019) 405-415. https://doi.org/10.1016/j.bbapap.2019.01.007

[41]

Y. Porat, A. Stepensky, F.X. Ding, et al., Completely different amyloidogenic potential of nearly identical peptide fragments, Biopolymers 69 (2003) 161-164. https://doi.org/10.1002/bip.10386

[42]

P. Westermark, U. Engström, K.H. Johnson, et al., Islet amyloid polypeptide: pinpointing amino acid residues linked to amyloid fibril formation, PNAS 87 (1990) 5036-5040. https://doi.org/10.1073/pnas.87.13.5036

[43]

K.E. Marshall, K.L. Morris, D. Charlton, et al., Hydrophobic, aromatic, and electrostatic interactions play a central role in amyloid fibril formation and stability, Biochemistry 50 (2011) 2061-2071. https://doi.org/10.1021/bi101936c

[44]

A.A. Profit, V. Felsen, J. Chinwong, et al., Evidence of π-stacking interactions in the self-assembly of hIAPP22-29, Proteins 81 (2013) 690-703. https://doi.org/10.1002/prot.24229

[45]

K.N.L. Huggins, M. Bisaglia, L. Bubacco, et al., Designed hairpin peptides interfere with amyloidogenesis pathways: fibril formation and cytotoxicity inhibition, interception of the preamyloid state, Biochemistry 50 (2011) 8202-8212. https://doi.org/10.1021/bi200760h

[46]

Y. Shi, W. Lv, A. Jiao, et al., A novel pentapeptide inhibitor reduces amyloid deposit formation by direct interaction with hIAPP, Int. J. Endocrinol. 2019 (2019) 9062032. https://doi.org/10.1155/2019/9062032

[47]

Q. Xuan, J. He, R. Chai, et al., Monomer-targeting affinity peptide inhibitors of amyloid with no self-fibrillatin and low cytotoxicity, Chem. Commun. 56 (2020) 1633-1636. https://doi.org/10.1039/C9CC08671D

[48]

F. Meng, A. Abedini, A. Plesner, et al., The flavanol (-)-epigallocatechin 3-gallate inhibits amyloid formation by islet amyloid polypeptide, disaggregates amyloid fibrils and protects cultured cells against IAPP induced toxicity, Biochemistry 49 (2010) 8127-8133. https://doi.org/10.1021/bi100939a

[49]

A. Franko, D.C. Rodriguez Camargo, A. Böddrich, et al., Epigallocatechin gallate (EGCG) reduces the intensity of pancreatic amyloid fibrils in human islet amyloid polypeptide (hIAPP) transgenic mice, Sci. Rep. 8 (2018) 1-12. https://doi.org/10.1038/s41598-017-18807-8

[50]

Y. Wang, Y. Lv, L. Jin, et al., Revealing the mechanism of EGCG, Genistein, Rutin, Quercetin, and Silibinin against hIAPP aggregation via computational simulations, Interdiscip. Sci. Comput. Life Sci. 12 (2020) 59-68. https://doi.org/10.1007/s12539-019-00352-9

[51]

B. Ren, Y. Liu, Y. Zhang, et al., Genistein: a dual inhibitor of both amyloid β and human islet amylin peptides, ACS Chem. Neurosci. 9 (2018) 1215-1224. https://doi.org/10.1021/acschemneuro.8b00039

[52]

J. Sun, T. Murata, H. Shigemori, Inhibitory activities of phenylpropanoids from Lycopus lucidus on amyloid aggregation related to Alzheimer's disease and type 2 diabetes, J. Nat. Med. 74 (2020) 579-583. https://doi.org/10.1007/s11418-020-01398-6

[53]

J. Sun, G. Jiang, H. Shigemori, Inhibitory activity on amyloid aggregation of rosmarinic acid and its substructures from Isodon japonicus, Nat. Prod. Commun. 14 (2019) 1-5. https://doi.org/10.1177/1934578X19843039

[54]

G. Jiang, M. Takase, Y. Aihara, et al., Inhibitory activities of kukoamines A and B from Lycii Cortex on amyloid aggregation related to Alzheimer's disease and type 2 diabetes, J. Nat. Med. 74 (2020) 247-251. https://doi.org/10.1007/s11418-019-01337-0

[55]

A. Ben Hmidene, M. Hanaki, K. Murakami, et al., Inhibitory activities of antioxidant flavonoids from Tamarix gallica on amyloid aggregation related to Alzheimer's and type 2 diabetes diseases, Biol. Pharm. Bull. 40 (2017) 238-241. https://doi.org/10.1248/bpb.b16-00801

[56]

B. Ren, Y. Liu, Y. Zhang, et al., Tanshinones inhibit hIAPP aggregation, disaggregate preformed hIAPP fibrils, and protect cultured cells, J. Mater. Chem. B. 6 (2017) 56-67. https://doi.org/10.1039/c7tb02538f

[57]

R. Li, T. Liang, L. Xu, et al., Protective effect of cinnamon polyphenols against STZ-diabetic mice fed high-sugar, high-fat diet and its underlying mechanism, Food Chem. Toxicol. 51 (2013) 419-425. https://doi.org/10.1016/j.fct.2012.10.024

[58]

N. Auberval, S. Dal, W. Bietiger, et al., Oxidative stress type influences the properties of antioxidants containing polyphenols in RINm5F beta cells, Evid. Based Complement. Alternat. Med. 2015 (2015) 859048. https://doi.org/10.1155/2015/859048

[59]

S. Dragan, F. Andrica, M.C. Serban, et al., Polyphenols-rich natural products for treatment of diabetes, Curr. Med. Chem. 22 (2015) 14-22. https://doi.org/10.2174/0929867321666140826115422

[60]

S. Zraika, R.L. Hull, J. Udayasankar, et al., Oxidative stress is induced by islet amyloid formation and time-dependently mediates amyloid-induced beta cell apoptosis, Diabetologia 52 (2009) 626-635. https://doi.org/10.1007/s00125-008-1255-x

Food Science and Human Wellness
Pages 259-269
Cite this article:
Abioye RO, Udenigwe CC. Potential of peptides and phytochemicals in attenuating different phases of islet amyloid polypeptide fibrillation for type 2 diabetes management. Food Science and Human Wellness, 2021, 10(3): 259-269. https://doi.org/10.1016/j.fshw.2021.02.017

441

Views

32

Downloads

3

Crossref

3

Web of Science

4

Scopus

0

CSCD

Altmetrics

Received: 27 September 2020
Revised: 10 October 2020
Accepted: 12 October 2020
Published: 16 April 2021
© 2021 Beijing Academy of Food Sciences. Production and hosting by Elsevier B.V. on behalf of KeAi Communications Co., Ltd.

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return