Journal Home > Volume 13 , Issue 1

Milk fat globule membrane (MFGM), which contains abundant glycoproteins and phospholipids, exerts beneficial effects on intestinal health and immunomodulation. The aim of this study was to evaluate the protective effects and possible underlying mechanisms of MFGM on cow’s milk allergy (CMA) in a β-lactoglobulin (BLG)-induced allergic mice model. MFGM was supplemented to allergic mice induced by BLG at a dose of 400 mg/kg body weight. Results demonstrated that MFGM alleviated food allergy symptoms, decreased serum levels of lipopolysaccharide, pro-inflammatory cytokines, immunoglobulin (Ig) E, IgG1, and Th2 cytokines including interleukin (IL)-4, while increased serum levels of Th1 cytokines including interferon-γ and regulatory T cells (Tregs) cytokines including IL-10 and transforming growth factor-β. MFGM modulated gut microbiota and enhanced intestinal barrier of BLG-allergic mice, as evidenced by decreased relative abundance of Desulfobacterota, Rikenellaceae, Lachnospiraceae, and Desulfovibrionaceae, while increased relative abundance of Bacteroidetes, Lactobacillaceae and Muribaculaceae, and enhanced expressions of tight junction proteins including Occludin, Claudin-1 and zonula occludens-1. Furthermore, MFGM increased fecal short-chain fatty acids (SCFAs) levels, which elevated G protein-coupled receptor(GPR) 43 and GPR109A expressions. The increased expressions of GPR43 and GPR109A induced CD103+ dendritic cells accumulation and promoted Tregs differentiation in mesenteric lymph node to a certain extent. In summary, MFGM alleviated CMA in a BLG-induced allergic mice model through enhancing intestinal barrier and promoting Tregs differentiation, which may be correlated with SCFAs-mediated activation of GPRs. These findings suggest that MFGM may be useful as a promising functional ingredient against CMA.


menu
Abstract
Full text
Outline
Electronic supplementary material
About this article

Milk fat globule membrane supplementation protects against β-lactoglobulininduced food allergy in mice via upregulation of regulatory T cells and enhancement of intestinal barrier in a microbiota-derived short-chain fatty acids manner

Show Author's information Han Gonga,Tiange LibDong LiangcJingxin GaoaXiaohan LiuaXueying Maoa( )
Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering;China Agricultural University, Beijing 100083, China
Henan Engineering Technology Research Center of Food Processing and Circulation Safety Control, College of Food Science and Technology, Henan Agricultural University, Zhengzhou 450002, China
Applied Nutrition I, China National Center for Food Safety Risk Assessment, Beijing 100022, China

Peer review under responsibility of Tsinghua University Press.

Abstract

Milk fat globule membrane (MFGM), which contains abundant glycoproteins and phospholipids, exerts beneficial effects on intestinal health and immunomodulation. The aim of this study was to evaluate the protective effects and possible underlying mechanisms of MFGM on cow’s milk allergy (CMA) in a β-lactoglobulin (BLG)-induced allergic mice model. MFGM was supplemented to allergic mice induced by BLG at a dose of 400 mg/kg body weight. Results demonstrated that MFGM alleviated food allergy symptoms, decreased serum levels of lipopolysaccharide, pro-inflammatory cytokines, immunoglobulin (Ig) E, IgG1, and Th2 cytokines including interleukin (IL)-4, while increased serum levels of Th1 cytokines including interferon-γ and regulatory T cells (Tregs) cytokines including IL-10 and transforming growth factor-β. MFGM modulated gut microbiota and enhanced intestinal barrier of BLG-allergic mice, as evidenced by decreased relative abundance of Desulfobacterota, Rikenellaceae, Lachnospiraceae, and Desulfovibrionaceae, while increased relative abundance of Bacteroidetes, Lactobacillaceae and Muribaculaceae, and enhanced expressions of tight junction proteins including Occludin, Claudin-1 and zonula occludens-1. Furthermore, MFGM increased fecal short-chain fatty acids (SCFAs) levels, which elevated G protein-coupled receptor(GPR) 43 and GPR109A expressions. The increased expressions of GPR43 and GPR109A induced CD103+ dendritic cells accumulation and promoted Tregs differentiation in mesenteric lymph node to a certain extent. In summary, MFGM alleviated CMA in a BLG-induced allergic mice model through enhancing intestinal barrier and promoting Tregs differentiation, which may be correlated with SCFAs-mediated activation of GPRs. These findings suggest that MFGM may be useful as a promising functional ingredient against CMA.

Keywords: Gut microbiota, Short-chain fatty acid, Cow’s milk allergy, Milk fat globule membrane, G protein-coupled receptor, Regulatory T cell

References(68)

[1]

A. Golkar, J.M. Milani, T. Vasiljevic, Altering allergenicity of cow’s milk by food processing for applications in infant formula, Crit. Rev. Food Sci. Nutr. 59 (2019) 159-172. https://doi.org/10.1080/10408398.2017.1363156.

[2]

S. Kaminogawa, S. Hachimura, H. Nakajima-adachi, Food allergens and mucosal immune systems with special reference to recognition of food allergens by gut-associated lymphoid tissue, Allergol. Int. 48 (1999) 15-23. https://doi.org/10.1046/j.1440-1592.1999.00112.x.

[3]

L. Fu, J. Song, C. Wang, et al., Bifidobacterium infantis potentially alleviates shrimp tropomyosin-induced allergy by tolerogenic dendritic cell-dependent induction of regulatory T cells and alterations in gut microbiota, Front. Immunol. 8 (2017) 1-14. https://doi.org/10.3389/fimmu.2017.01536.

[4]

R.M. Steinman, Decisions about dendritic cells: past, present, and future, Annu. Rev. Immunol. 30 (2012) 1-22. https://doi.org/10.1146/annurev-immunol-100311-102839.

[5]

J. Tan, C. McKenzie, P.J. Vuillermin, G. Goverse, et al., Dietary fiber and bacterial SCFA enhance oral tolerance and protect against food allergy through diverse cellular pathways, Cell Rep. 15 (2016) 2809-2824. https://doi.org/10.1016/j.celrep.2016.05.047.

[6]

G. Matteoli, E. Mazzini, I.D. Iliev, et al., Gut CD103+ dendritic cells express indoleamine 2,3-dioxygenase which influences T regulatory/T effector cell balance and oral tolerance induction, Gut. 59 (2010) 595-604. https://doi.org/10.1136/gut.2009.185108.

[7]

C. Perrier, B. Corthésy, Gut permeability and food allergies, Clin. Exp. Allergy. 41 (2011) 20-28. https://doi.org/10.1111/j.1365-2222.2010.03639.x.

[8]

A. Koh, F. De Vadder, P. Kovatcheva-Datchary, et al., From dietary fiber to host physiology: short-chain fatty acids as key bacterial metabolites, Cell 165 (2016) 1332-1345. https://doi.org/10.1016/j.cell.2016.05.041.

[9]

F. De Filippis, L. Paparo, R. Nocerino, et al., Specific gut microbiome signatures and the associated pro-inflamatory functions are linked to pediatric allergy and acquisition of immune tolerance, Nat. Commun. 12 (2021). https://doi.org/10.1038/s41467-021-26266-z.

[10]

A. Cait, E. Cardenas, P.A. Dimitriu, et al., Reduced genetic potential for butyrate fermentation in the gut microbiome of infants who develop allergic sensitization, J. Allergy Clin. Immunol. 144 (2019) 1638-1647.e3. https://doi.org/10.1016/j.jaci.2019.06.029.

[11]

M.R. Goldberg, H. Mor, D. Magid Neriya, et al., Microbial signature in IgE-mediated food allergies, Genome Med. 12 (2020). https://doi.org/10.1186/s13073-020-00789-4.

[12]

X. Yan, J. Yan, Q. Xiang, et al., Fructooligosaccharides protect against OVA-induced food allergy in mice by regulating the Th17/Treg cell balance using tryptophan metabolites, Food Funct. 12 (2021) 3191-3205. https://doi.org/10.1039/D0FO03371E.

[13]

M.J. Bae, H.S. Shin, H.J. See, et al., Baicalein induces CD4+ Foxp3+ T cells and enhances intestinal barrier function in a mouse model of food allergy, Sci. Rep. 6 (2016) 1-11. https://doi.org/10.1038/srep32225.

[14]

D. Markovic, R.A.J. Challiss, Alternative splicing of G protein-coupled receptors: physiology and pathophysiology, Cell. Mol. Life Sci. 66 (2009) 3337-3352. https://doi.org/10.1007/s00018-009-0093-4.

[15]

L. Macia, J. Tan, A.T. Vieira, et al., Metabolite-sensing receptors GPR43 and GPR109A facilitate dietary fibre-induced gut homeostasis through regulation of the inflammasome, Nat. Commun. 6 (2015) 6734. https://doi.org/10.1038/ncomms7734.

[16]

L. Pan, Z. Ren, X. Tu, et al., GPR109A deficiency promotes IL-33 overproduction and type 2 immune response in food allergy in mice, Allergy 76 (2021) 2613-2616. https://doi.org/10.1111/all.14849.

[17]

R.S. Chinthrajah, J.D. Hernandez, S.D. Boyd, et al., Molecular and cellular mechanisms of food allergy and food tolerance, J. Allergy Clin. Immunol. 137 (2016) 984-997. https://doi.org/10.1016/j.jaci.2016.02.004.

[18]

D. Cheng, J.H. Xu, J.Y. Li, et al., Butyrate ameliorated-NLRC3 protects the intestinal barrier in a GPR43-dependent manner, Exp. Cell Res. 368 (2018) 101-110. https://doi.org/10.1016/j.yexcr.2018.04.018.

[19]

L. Paparo, R. Nocerino, E. Ciaglia, et al., Butyrate as a bioactive human milk protective component against food allergy, Allergy 76 (2021) 1398-1415. https://doi.org/10.1111/all.14625.

[20]

K. Dewettinck, R. Rombaut, N. Thienpont, et al., Nutritional and technological aspects of milk fat globule membrane material, Int. Dairy J. 18 (2008) 436-457. https://doi.org/10.1016/j.idairyj.2007.10.014.

[21]

Å. Nilsson, Role of sphingolipids in infant gut health and immunity, J. Pediatr. 173 (2016) S53-S59. https://doi.org/10.1016/j.jpeds.2016.02.076.

[22]

R. Zanabria, A.M. Tellez, M. Griffiths, et al., Modulation of immune function by milk fat globule membrane isolates, J. Dairy Sci. 97 (2014) 2017-2026. https://doi.org/10.3168/jds.2013-7563.

[23]

X. Zhang, Y. Wu, H. Ye, et al., Dietary milk fat globule membrane supplementation during late gestation increased the growth of neonatal piglets by improving their plasma parameters, intestinal barriers, and fecal microbiota, RSC Adv. 10 (2020) 16987-16998. https://doi.org/10.1039/d0ra02618b.

[24]

T. Li, J. Gao, M. Du, et al., Milk fat globule membrane supplementation modulates the gut microbiota and attenuates metabolic endotoxemia in high-fat diet-fed mice, J. Funct. Foods 47 (2018) 56-65. https://doi.org/10.1016/j.jff.2018.05.038.

[25]

H. Gong, Q. Yuan, J. Pang, et al., Dietary milk fat globule membrane restores decreased intestinal mucosal barrier development and alterations of intestinal flora in infant-formula-fed rat pups, Mol. Nutr. Food Res. 64 (2020) 1-12. https://doi.org/10.1002/mnfr.202000232.

[26]

S. Lamothe, G. Robitaille, D. St-Gelais, et al., Butter making from caprine creams: effect of washing treatment on phospholipids and milk fat globule membrane proteins distribution, J. Dairy Res. 75 (2008) 439-443. https://doi.org/10.1017/S002202990800349X.

[27]

G. Fu, K. Zhao, H. Chen, et al., Effect of 3 lactobacilli on immunoregulation and intestinal microbiota in a β-lactoglobulin–induced allergic mouse model, J. Dairy Sci. 102 (2019) 1943-1958. https://doi.org/10.3168/jds.2018-15683.

[28]

Z. Shu, Q. Liu, C. Xing, et al., Viridicatol isolated from deep-sea Penicillium griseofulvum alleviates anaphylaxis and repairs the intestinal barrier in mice by suppressing mast cell activation, Mar. Drugs. 18 (2020) 517. https://doi.org/10.3390/md18100517.

[29]

T. Liu, P. Chen, M. Munir, et al., HMOs modulate immunoregulation and gut microbiota in a β-lactoglobulin-induced allergic mice model, J. Funct. Foods 70 (2020) 103993. https://doi.org/10.1016/j.jff.2020.103993.

[30]

C. Chen, L. Lianhua, S. Nana, et al., Development of a BALB/c mouse model for food allergy: comparison of allergy-related responses to peanut agglutinin, β-lactoglobulin and potato acid phosphatase, Toxicol. Res. (Camb). 6 (2017) 251-261. https://doi.org/10.1039/c6tx00371k.

[31]

K. Adel-Patient, C. Créminon, H. Bernard, et al., Evaluation of a high IgE-responder mouse model of allergy to bovine β-lactoglobulin (BLG): development of sandwich immunoassays for total and allergen-specific IgE, IgG1 and IgG2a in BLG-sensitized mice, J. Immunol. Methods. 235 (2000) 21-32. https://doi.org/10.1016/S0022-1759(99)00210-0.

[32]

P. Chen, S. Lei, M. Tong, et al., Effect of polysaccharide fractions from Fortunella margarita on the fecal microbiota of mice and SCFA production in vitro, Food Sci. Hum. Wellness 11 (2022) 97-108. https://doi.org/10.1016/j.fshw.2021.07.011.

[33]

J. Ma, J. Zhang, Q. Li, et al., Oral administration of a mixture of probiotics protects against food allergy via induction of CD103+ dendritic cells and modulates the intestinal microbiota, J. Funct. Foods 55 (2019) 65-75. https://doi.org/10.1016/j.jff.2019.02.010.

[34]

J. Gao, J. Song, M. Du, et al., Bovine α-lactalbumin hydrolysates (α-LAH) attenuate high-fat diet induced nonalcoholic fatty liver disease by modulating hepatic lipid metabolism in C57BL/6J mice, J. Funct. Foods 54 (2019) 254-262. https://doi.org/10.1016/j.jff.2019.01.027.

[35]

Z. Cheng, J. Lin, N. Gao, et al., Blueberry malvidin-3-galactoside modulated gut microbial dysbiosis and microbial TCA cycle KEGG pathway disrupted in a liver cancer model induced by HepG2 cells, Food Sci. Hum. Wellness 9 (2020) 245-255. https://doi.org/10.1016/j.fshw.2020.04.006.

[36]

D. Mariat, O. Firmesse, F. Levenez, et al., The Firmicutes/Bacteroidetes ratio of the human microbiota changes with age, BMC Microbiol. 9 (2009) 123. https://doi.org/10.1186/1471-2180-9-123.

[37]

Y. Shao, Y. Zhang, L. Zhang, et al., Mechanism of reduction in allergenicity and altered human intestinal microbiota of digested β-lactoglobulin modified by ultrasonic pretreatment combined with glycation, J. Agric. Food Chem. 69 (2021) 14004-14012. https://doi.org/10.1021/acs.jafc.1c03501.

[38]

T. Li, Q. Yuan, H. Gong, et al., Gut microbiota mediates the alleviative effect of polar lipids-enriched milk fat globule membrane on obesity-induced glucose metabolism disorders in peripheral tissues in rat dams, Int. J. Obes. (2022). https://doi.org/10.1038/s41366-021-01029-4.

[39]
Y. Yang, G. Chen, Q. Yang, et al., Gut microbiota drives the attenuation of dextran sulphate sodium-induced colitis by Huangqin decoction, 8 (2017) 48863-48874.
DOI
[40]

Y. Liu, Y. Ma, Z. Chen, et al., Depolymerized sulfated galactans from Eucheuma serra ameliorate allergic response and intestinal flora in food allergic mouse model, Int. J. Biol. Macromol. (2020). https://doi.org/10.1016/j.ijbiomac.2020.10.254.

[41]

L. Fu, M. Xie, C. Wang, et al., Lactobacillus casei Zhang alleviates shrimp tropomyosin-induced food allergy by switching antibody isotypes through the NF-κB-dependent immune tolerance, Mol. Nutr. Food Res. 64 (2020). https://doi.org/10.1002/mnfr.201900496.

[42]

C. Huang, Y. Lin, T. Jan, Lactobacillus reuteri induces intestinal immune tolerance against food allergy in mice, J. Funct. Foods. 31 (2017) 44-51. https://doi.org/10.1016/j.jff.2017.01.034.

[43]

K.L. Ormerod, D.L.A. Wood, N. Lachner, et al., Genomic characterization of the uncultured Bacteroidales family S24-7 inhabiting the guts of homeothermic animals, Microbiome 4 (2016) 36. https://doi.org/10.1186/s40168-016-0181-2.

[44]

H. Zeng, S.L. Ishaq, F.Q. Zhao, et al., Colonic inflammation accompanies an increase of β-catenin signaling and Lachnospiraceae/Streptococcaceae bacteria in the hind gut of high-fat diet-fed mice, J. Nutr. Biochem. 35 (2016) 30-36. https://doi.org/10.1016/j.jnutbio.2016.05.015.

[45]

R. Berni Canani, N. Sangwan, A.T. Stefka, et al., Lactobacillus rhamnosus GG-supplemented formula expands butyrate-producing bacterial strains in food allergic infants, ISME J. 10 (2016) 742-750. https://doi.org/10.1038/ismej.2015.151.

[46]

M. Noval Rivas, O.T. Burton, P. Wise, et al., A microbiota signature associated with experimental food allergy promotes allergic sensitization and anaphylaxis, J. Allergy Clin. Immunol. 131 (2013) 201-212. https://doi.org/10.1016/j.jaci.2012.10.026.

[47]

M. Milard, F. Laugerette, A. Durand, et al., Milk polar lipids in a high-fat diet can prevent body weight gain: modulated abundance of gut bacteria in relation with fecal loss of specific fatty acids, Mol. Nutr. Food Res. 63 (2019) 1801078. https://doi.org/10.1002/mnfr.201801078.

[48]

G.H. Norris, M. Milard, M.C. Michalski, et al., Protective properties of milk sphingomyelin against dysfunctional lipid metabolism, gut dysbiosis, and inflammation, J. Nutr. Biochem. 73 (2019) 108224. https://doi.org/10.1016/j.jnutbio.2019.108224.

[49]

C. Stevens, T. Millar, J. Clinch, et al., Antibacterial properties of xanthine oxidase in human milk, Lancet. 356 (2000) 829-830. https://doi.org/10.1016/S0140-6736(00)02660-X.

[50]

S.K. Lindén, Y.H. Sheng, A.L. Every, et al., MUC1 limits Helicobacter pylori infection both by steric hindrance and by acting as a releasable decoy, PLoS Pathog. 5 (2009). https://doi.org/10.1371/journal.ppat.1000617.

[51]

M.F. Böttcher, E.K. Nordin, A. Sandin, et al., Microflora-associated characteristics in faeces from allergic and nonallergic infants, Clin. Exp. Allergy. 30 (2000) 1591-1596. https://doi.org/10.1046/j.1365-2222.2000.00982.x.

[52]

K. Struijs, T. Van De Wiele, T.T. Le, et al., Milk fat globule membrane glycoproteins prevent adhesion of the colonic microbiota and result in increased bacterial butyrate production, Int. Dairy J. 32 (2013) 99-109. https://doi.org/10.1016/j.idairyj.2013.05.001.

[53]

B. Tian, J. Zhao, M. Zhang, et al., Lycium ruthenicum anthocyanins attenuate high-fat diet-induced colonic barrier dysfunction and inflammation in mice by modulating the gut microbiota, Mol. Nutr. Food Res. 65 (2021) 1-16. https://doi.org/10.1002/mnfr.202000745.

[54]

P. Louis, H.J. Flint, Formation of propionate and butyrate by the human colonic microbiota, Environ. Microbiol. 19 (2017) 29-41. https://doi.org/10.1111/1462-2920.13589.

[55]

S. Wessler, S. Backert, Molecular mechanisms of epithelial-barrier disruption by Helicobacter pylori, Trends Microbiol. 16 (2008) 397-405. https://doi.org/10.1016/j.tim.2008.05.005.

[56]

X. Wang, S. Hirmo, R. Willen, et al., Inhibition of Helicobacter pylori infection by bovine milk glycoconjugates in a BALB/cA mouse model, J. Med. Microbiol. 50 (2001) 430-435. https://doi.org/10.1099/0022-1317-50-5-430.

[57]

T. Imaizumi, T. Aizawa-Yashiro, S. Watanabe, et al., TLR4 signaling induces retinoic acid-inducible gene-I and melanoma differentiation-associated gene 5 in mesangial cells, J. Nephrol. 26 (2013) 886-893. https://doi.org/10.5301/jn.5000254.

[58]

B. Funke, F. Lasitschka, W. Roth, et al., Selective downregulation of retinoic acid-inducible gene i within the intestinal epithelial compartment in Crohn’s disease, Inflamm. Bowel Dis. 17 (2011) 1943-1954. https://doi.org/10.1002/ibd.21572.

[59]

Y. Wang, H.X. Zhang, Y.P. Sun, et al., Rig-I-/- mice develop colitis associated with downregulation of Gαi2, Cell Res. 17 (2007) 858-868. https://doi.org/10.1038/cr.2007.81.

[60]

J.J. Wang, Q.M. Zhang, W.W. Ni, et al., Modulatory effect of Lactobacillus acidophilus KLDS 1.0738 on intestinal short-chain fatty acids metabolism and GPR41/43 expression in β-lactoglobulin–sensitized mice, Microbiol. Immunol. 63 (2019) 303-315. https://doi.org/10.1111/1348-0421.12723.

[61]

Y.J. Zhou, J. Gao, H.M. Yang, et al., The role of the lactadherin in promoting intestinal DCs development in vivo and vitro, Clin. Dev. Immunol. 2010 (2010) 1-9. https://doi.org/10.1155/2010/357541.

[62]

R. Sibilano, B. Frossi, C.E. Pucillo, Mast cell activation: a complex interplay of positive and negative signaling pathways, Eur. J. Immunol. 44 (2014) 2558-2566. https://doi.org/10.1002/eji.201444546.

[63]

N.Y. Lee, K.S. Chung, J.S. Jin, et al., The inhibitory effect of nodakenin on mast-cell-mediated allergic inflammation via downregulation of NF-κB and caspase-1 activation, J. Cell. Biochem. 118 (2017) 3993-4001. https://doi.org/10.1002/jcb.26055.

[64]

B.J. Wang, H.W. Chiu, Y.L. Lee, et al., Pterostilbene attenuates hexavalent chromium-induced allergic contact dermatitis by preventing cell apoptosis and inhibiting IL-1β-related NLRP3 inflammasome activation, J. Clin. Med. 7 (2018) 489. https://doi.org/10.3390/jcm7120489.

[65]

J. Tulyeu, H. Kumagai, E. Jimbo, et al., Probiotics prevents sensitization to oral antigen and subsequent increases in intestinal tight junction permeability in juvenile–young adult rats, Microorganisms 7 (2019) 1-19. https://doi.org/10.3390/microorganisms7100463.

[66]

N. Singh, A. Gurav, S. Sivaprakasam, et al., Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis, Immunity 40 (2014) 128-139. https://doi.org/10.1016/j.immuni.2013.12.007.

[67]

W. Feng, Y. Wu, G. Chen, et al., Sodium butyrate attenuates diarrhea in weaned piglets and promotes tight junction protein expression in colon in a GPR109A-dependent manner, Cell. Physiol. Biochem. 47 (2018) 1617-1629. https://doi.org/10.1159/000490981.

[68]

S. Huang, S. Hu, S. Liu, et al., Lithium carbonate alleviates colon inflammation through modulating gut microbiota and Treg cells in a GPR43-dependent manner, Pharmacol. Res. 175 (2022) 105992. https://doi.org/10.1016/j.phrs.2021.105992.

File
fshw-2024-9250010_ESM.pdf (119.2 KB)
Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 12 April 2022
Revised: 20 May 2022
Accepted: 22 July 2022
Published: 01 June 2023
Issue date: January 2024

Copyright

© 2024 Beijing Academy of Food Sciences. Publishing services by Tsinghua University Press.

Acknowledgements

Acknowledgements

This work was supported by the National Key Research and Development Program of China (Grant No. 2019YFC1605000), National Natural Science Foundation of China (Grant No. 31871806), and the Beijing Livestock Industry Innovation Team (BAIC05-2023).

Rights and permissions

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

Return