Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

ISSN (Print): 1381-6128
ISSN (Online): 1873-4286

Review Article

Targeting Nanotechnology and Nutraceuticals in Obesity: An Updated Approach

Author(s): Neelam Sharma, Tapan Behl*, Sukhbir Singh*, Parneet Kaur, Ishrat Zahoor, Syam Mohan, Mahesh Rachamalla, Hamed Ghaleb Dailah, Yosif Almoshari, Ahmad Salawi, Meshal Alshamrani and Lotfi Aleya

Volume 28, Issue 40, 2022

Published on: 11 November, 2022

Page: [3269 - 3288] Pages: 20

DOI: 10.2174/1381612828666221003105619

Price: $65

conference banner
Abstract

Hypothesis: This review article represents a brief layout of the risk factors and pathophysiology responsible for obesity, customary treatment strategies, and nanotechnology-based nutraceutical for the therapeutics of obesity.

Experiments: An exhaustive search of the literature was done for this purpose, using Google Scholar, PubMed, and ScienceDirect databases. A literature study was conducted using publications published in peer-reviewed journals between 2000 and 2022.

Findings: This was revealed that risk factors responsible for obesity were genetic abnormalities and environmental and socio-economic factors. Several research articles published between 2000 and 2022 were based on phytoconstituents-based nanoformulation for obesity therapeutics and, therefore, have been systematically compiled in this review. Various nutraceuticals like Garcinia cambogia, quercetin, resveratrol, capsaicin, Capsicum, Curcuma longa, Camella Sinensis, Zingiber officinalis, Citrus aurantium, Aegle marmelos, Coffea canephora, Asparagus officinalis, Gardenia jasminoides, Catha edulis, Clusia nemroisa, Rosmarinus officinalis, Cirsium setidens, Betula platyphylla, Tripterygium wilfordi possessing anti-obesity actions are discussed in this review along with their patents, clinical trials as well as their nanoformulation available.

Conclusion: This review illustrates that nanotechnology has a great propensity to impart a promising role in delivering phytochemicals and nutraceuticals in managing obesity conditions and other related disorders.

Keywords: Genetic Abnormalities, Nanoformulations, Nanotechnology, Nutraceuticals, Obesity, Phytoconstituents.

[1]
Bray GA. History of obesity 2016; 3-18. Available from: https://onlinelibrary.wiley.com/doi/abs/10.1002/9780470712221.ch1
[2]
Balaji M, Ganjayi MS, Hanuma Kumar GEN, Parim BN, Mopuri R, Dasari S. A review on possible therapeutic targets to contain obesity: The role of phytochemicals. Obes Res Clin Pract 2016; 10(4): 363-80.
[http://dx.doi.org/10.1016/j.orcp.2015.12.004] [PMID: 26740473]
[3]
Yun JW. Possible anti-obesity therapeutics from nature-A review. Phytochemistry 2010; 71(14-15): 1625-41.
[http://dx.doi.org/10.1016/j.phytochem.2010.07.011] [PMID: 20732701]
[4]
Seidell JC, Halberstadt J. The global burden of obesity and the challenges of prevention. Ann Nutr Metab 2015; 66 (Suppl. 2): 7-12.
[http://dx.doi.org/10.1159/000375143] [PMID: 26045323]
[5]
Pérez CM, Sánchez H, Ortiz AP. Prevalence of overweight and obesity and their cardiometabolic comorbidities in Hispanic adults living in Puerto Rico. J Community Health 2013; 38(6): 1140-6.
[http://dx.doi.org/10.1007/s10900-013-9726-5] [PMID: 23846388]
[6]
Sasson M, Lee M, Jan C, Fontes F, Motta J. Prevalence and associated factors of obesity among Panamanian adults. 1982-2010. PLoS One 2014; 9(3): e91689.
[http://dx.doi.org/10.1371/journal.pone.0091689] [PMID: 24621825]
[7]
Hu FB. Obesity Epidemiology. Oxford: Oxford University Press 2008; p. 498.
[http://dx.doi.org/10.1093/acprof:oso/9780195312911.001.0001]
[8]
Sun NN, Wu TY, Chau CF. Natural dietary and herbal products in anti-obesity treatment. Molecules 2016; 21(10): 1351.
[http://dx.doi.org/10.3390/molecules21101351] [PMID: 27727194]
[9]
Banjare J. Application of nanotechnology in food technology and targeted drug therapy for prevention of obesity: An overview. J Crit Rev 2016; 4(1): 7-11.
[http://dx.doi.org/10.22159/jcr.2017v4i1.14235]
[10]
Sibuyi NRS, Moabelo KL, Meyer M, Onani MO, Dube A, Madiehe AM. Nanotechnology advances towards development of targeted-treatment for obesity. J Nanobiotechnol 2019; 17(1): 122.
[http://dx.doi.org/10.1186/s12951-019-0554-3] [PMID: 31842876]
[11]
Shende P, Narvenker R. Herbal nanotherapy: A new paradigm over conventional obesity treatment. J Drug Deliv Sci Technol 2021; 61: 102291.
[http://dx.doi.org/10.1016/j.jddst.2020.102291]
[12]
Loos RJF, Bouchard C. FTO: The first gene contributing to common forms of human obesity. Obes Rev 2008; 9(3): 246-50.
[http://dx.doi.org/10.1111/j.1467-789X.2008.00481.x] [PMID: 18373508]
[13]
Speiser PW, Rudolf MCJ, Anhalt H, et al. Childhood obesity. J Clin Endocrinol Metab 2005; 90(3): 1871-87.
[http://dx.doi.org/10.1210/jc.2004-1389] [PMID: 15598688]
[14]
Lee EY, Yoon KH. Epidemic obesity in children and adolescents: Risk factors and prevention. Front Med 2018; 12(6): 658-66.
[http://dx.doi.org/10.1007/s11684-018-0640-1] [PMID: 30280308]
[15]
Gurka MJ, Filipp SL, DeBoer MD. Geographical variation in the prevalence of obesity, metabolic syndrome, and diabetes among US adults. Nutr Diabetes 2018; 8(1): 14.
[http://dx.doi.org/10.1038/s41387-018-0024-2] [PMID: 29549249]
[16]
Wen M, Fan JX, Kowaleski-Jones L, Wan N. Rural-Urban disparities in obesity prevalence among working age adults in the united states: Exploring the mechanisms. Am J Health Promot 2018; 32(2): 400-8.
[http://dx.doi.org/10.1177/0890117116689488] [PMID: 29214811]
[17]
Maddock J. The relationship between obesity and the prevalence of fast food restaurants: State-level analysis. Am J Health Promot 2004; 19(2): 137-43.
[http://dx.doi.org/10.4278/0890-1171-19.2.137] [PMID: 15559714]
[18]
Mazidi M, Speakman JR. Higher densities of fast-food and full-service restaurants are not associated with obesity prevalence. Am J Clin Nutr 2017; 106(2): 603-13.
[http://dx.doi.org/10.3945/ajcn.116.151407] [PMID: 28566310]
[19]
Hu FB. Resolved: There is sufficient scientific evidence that decreasing sugar-sweetened beverage consumption will reduce the prevalence of obesity and obesity-related diseases. Obes Rev 2013; 14(8): 606-19.
[http://dx.doi.org/10.1111/obr.12040] [PMID: 23763695]
[20]
Gebrie A, Alebel A, Zegeye A, Tesfaye B, Ferede A. Prevalence and associated factors of overweight/obesity among children and adolescents in Ethiopia: A systematic review and meta-analysis. BMC Obes 2018; 5(1): 19.
[http://dx.doi.org/10.1186/s40608-018-0198-0] [PMID: 30002860]
[21]
Mariapun J, Ng CW, Hairi NN. The gradual shift of overweight, obesity, and abdominal obesity towards the poor in a multi-ethnic developing country: Findings from the Malaysian National Health and Morbidity Surveys. J Epidemiol 2018; 28(6): 279-86.
[http://dx.doi.org/10.2188/jea.JE20170001] [PMID: 29657257]
[22]
Monteiro CA, Conde WL, Popkin BM. Income-specific trends in obesity in Brazil: 1975-2003. Am J Public Health 2007; 97(10): 1808-12.
[http://dx.doi.org/10.2105/AJPH.2006.099630] [PMID: 17761560]
[23]
Swinburn BA, Sacks G, Hall KD, et al. The global obesity pandemic: Shaped by global drivers and local environments. Lancet 2011; 378(9793): 804-14.
[http://dx.doi.org/10.1016/S0140-6736(11)60813-1] [PMID: 21872749]
[24]
Heymsfield SB, Wadden TA. Mechanisms, pathophysiology, and management of obesity. N Engl J Med 2017; 376(15): 1490-2.
[http://dx.doi.org/10.1056/NEJMc1701944] [PMID: 28402780]
[25]
Tschöp M, Weyer C, Tataranni PA, Devanarayan V, Ravussin E, Heiman ML. Circulating ghrelin levels are decreased in human obesity. Diabetes 2001; 50(4): 707-9.
[http://dx.doi.org/10.2337/diabetes.50.4.707] [PMID: 11289032]
[26]
Lowell BB, Spiegelman BM. Towards a molecular understanding of adaptive thermogenesis. Nature 2000; 404(6778): 652-60.
[http://dx.doi.org/10.1038/35007527] [PMID: 10766252]
[27]
Könner AC, Klöckener T, Brüning JC. Control of energy homeostasis by insulin and leptin: Targeting the arcuate nucleus and beyond. Physiol Behav 2009; 97(5): 632-8.
[http://dx.doi.org/10.1016/j.physbeh.2009.03.027] [PMID: 19351541]
[28]
Robertson SA, Leinninger GM, Myers MG Jr. Molecular and neural mediators of leptin action. Physiol Behav 2008; 94(5): 637-42.
[http://dx.doi.org/10.1016/j.physbeh.2008.04.005] [PMID: 18501391]
[29]
Garvey WT, Mechanick JI, Brett EM, et al. American association of clinical endocrinologists and American college of endocrinology comprehensive clinical practice guidelines formedical care of patients with obesity. Endocr Pract 2016; 22 (Suppl. 3): 1-203.
[http://dx.doi.org/10.4158/EP161365.GL] [PMID: 27219496]
[30]
Pusztai P, Sarman B, Ruzicska E, et al. Ghrelin: A new peptide regulating the neurohormonal system, energy homeostasis and glucose metabolism. Diabetes Metab Res Rev 2008; 24(5): 343-52.
[http://dx.doi.org/10.1002/dmrr.830] [PMID: 18350524]
[31]
Ritter S, Vetter ML, Sarwer DB. Lifestyle modifications and surgical options in the treatment of patients with obesity and type 2 diabetes mellitus. Postgrad Med 2012; 124(4): 168-80.
[http://dx.doi.org/10.3810/pgm.2012.07.2578] [PMID: 22913905]
[32]
Wadden TA, Tronieri JS, Butryn ML. Lifestyle modification approaches for the treatment of obesity in adults. Am Psychol 2020; 75(2): 235-51.
[http://dx.doi.org/10.1037/amp0000517] [PMID: 32052997]
[33]
Apovian CM, Aronne LJ, Bessesen DH, et al. Pharmacological management of obesity: An endocrine Society clinical practice guideline. J Clin Endocrinol Metab 2015; 100(2): 342-62.
[http://dx.doi.org/10.1210/jc.2014-3415] [PMID: 25590212]
[34]
Greenway FL, Fujioka K, Plodkowski RA, et al. Effect of naltrexone plus bupropion on weight loss in overweight and obese adults (COR-I): A multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2010; 376(9741): 595-605.
[http://dx.doi.org/10.1016/S0140-6736(10)60888-4] [PMID: 20673995]
[35]
Yanovski SZ, Yanovski JA. Long-term drug treatment for obesity: A systematic and clinical review. JAMA 2014; 311(1): 74-86.
[http://dx.doi.org/10.1001/jama.2013.281361] [PMID: 24231879]
[36]
Gadde KM, Allison DB, Ryan DH, et al. Effects of low-dose, controlled-release, phentermine plus topiramate combination on weight and associated comorbidities in overweight and obese adults (CONQUER): A randomised, placebo-controlled, phase 3 trial. Lancet 2011; 377(9774): 1341-52.
[http://dx.doi.org/10.1016/S0140-6736(11)60205-5] [PMID: 21481449]
[37]
James WPT, Astrup A, Finer N, et al. Effect of sibutramine on weight maintenance after weight loss: A randomised trial. Lancet 2000; 356(9248): 2119-25.
[http://dx.doi.org/10.1016/S0140-6736(00)03491-7] [PMID: 11191537]
[38]
Mun EC, Blackburn GL, Matthews JB. Current status of medical and surgical therapy for obesity. Gastroenterology 2001; 120(3): 669-81.
[http://dx.doi.org/10.1053/gast.2001.22430] [PMID: 11179243]
[39]
Astrup A, Toubro S. When, for whom and how to use sibutramine. Int J Obes 2001; 25(S4) (Suppl. 4): S2-7.
[http://dx.doi.org/10.1038/sj.ijo.0801930] [PMID: 11916101]
[40]
McDuffie JR, Calis KA, Booth SL, Uwaifo GI, Yanovski JA. Effects of orlistat on fat-soluble vitamins in obese adolescents. Pharmacotherapy 2002; 22(7): 814-22.
[http://dx.doi.org/10.1592/phco.22.11.814.33627] [PMID: 12126214]
[41]
Hanl K, Kimura Y, Okuda H. Anti-obesity effects of natural products. Stud Nat Prod Chem 2005; 30: 79-110.
[http://dx.doi.org/10.1016/S1572-5995(05)80031-6]
[42]
Kalra EK. Nutraceutical-definition and introduction. AAPS PharmSci 2003; 5(3): 27-8.
[http://dx.doi.org/10.1208/ps050325] [PMID: 14621960]
[43]
Mayer M, Höcht C, Puyó A, Taira C. Recent advances in obesity pharmacotherapy. Curr Clin Pharmacol 2009; 4(1): 53-61.
[http://dx.doi.org/10.2174/157488409787236128] [PMID: 19149502]
[44]
Madhu NBK. Potential functional foods for obesity. Int J Food Sci Nutr 2012; 1: 166-77.
[http://dx.doi.org/10.1007/s13197-014-1638-6]
[45]
Andlauer W, Fürst P. Nutraceuticals: A piece of history, present status and outlook. Food Res Int 2002; 35(2-3): 171-6.
[http://dx.doi.org/10.1016/S0963-9969(01)00179-X]
[46]
Lokhande SS. Role of nutraceuticals in various diseases: A comprehensive review. Asian J Pharm Res 2018; 8(4): 236-40.
[http://dx.doi.org/10.5958/2231-5691.2018.00040.0]
[47]
Sachdeva V, Roy A, Bharadvaja N. Current prospects of nutraceuticals: A review. Curr Pharm Biotechnol 2020; 21(10): 884-96.
[http://dx.doi.org/10.2174/1389201021666200130113441] [PMID: 32000642]
[48]
Fu C, Jiang Y, Guo J, Su Z. Natural products with anti-obesity effects and different mechanisms of action. J Agric Food Chem 2016; 64(51): 9571-85.
[http://dx.doi.org/10.1021/acs.jafc.6b04468] [PMID: 27931098]
[49]
Lu M, Cao Y, Xiao J, Song M, Ho CT. Molecular mechanisms of the anti-obesity effect of bioactive ingredients in common spices: A review. Food Funct 2018; 9(9): 4569-81.
[http://dx.doi.org/10.1039/C8FO01349G] [PMID: 30168574]
[50]
Turner-McGrievy G, Harris M. Key elements of plant-based diets associated with reduced risk of metabolic syndrome. Curr Diab Rep 2014; 14(9): 524.
[http://dx.doi.org/10.1007/s11892-014-0524-y] [PMID: 25084991]
[51]
Bahmani M, Eftekhari Z, Saki K, Fazeli-Moghadam E, Jelodari M, Rafieian-Kopaei M. Obesity phytotherapy. J Evid Based Complementary Altern Med 2016; 21(3): 228-34.
[http://dx.doi.org/10.1177/2156587215599105] [PMID: 26269377]
[52]
Ghani U, Naeem M, Rafeeq H, et al. A novel approach towards nutraceuticals and biomedical applications. Schol Int J Biochem 2019; 2(10): 245-52.
[http://dx.doi.org/10.36348/SIJB.2019.v02i10.001]
[53]
Daou C, Zhang H. Functional and physiological properties of total, soluble, and insoluble dietary fibres derived from defatted rice bran. J Food Sci Technol 2014; 51(12): 3878-85.
[http://dx.doi.org/10.1007/s13197-013-0925-y] [PMID: 25477656]
[54]
Gidley MJ, Yakubov GE. Functional categorisation of dietary fibre in foods: Beyond ‘soluble’ vs. ‘insoluble’. Trends Food Sci Technol 2019; 86: 563-8.
[http://dx.doi.org/10.1016/j.tifs.2018.12.006]
[55]
Gibson GR. Fibre and effects on probiotics (the prebiotic concept). Clin Nutr Suppl 2004; 1(2): 25-31.
[http://dx.doi.org/10.1016/j.clnu.2004.09.005]
[56]
Pedret A, Valls RM, Calderón-Pérez L, et al. Effects of daily consumption of the probiotic Bifidobacterium animalis subsp. lactis CECT 8145 on anthropometric adiposity biomarkers in abdominally obese subjects: A randomized controlled trial. Int J Obes 2019; 43(9): 1863-8.
[http://dx.doi.org/10.1038/s41366-018-0220-0] [PMID: 30262813]
[57]
Kuo SM. The interplay between fiber and the intestinal microbiome in the inflammatory response. Adv Nutr 2013; 4(1): 16-28.
[http://dx.doi.org/10.3945/an.112.003046] [PMID: 23319119]
[58]
Panda SK, Behera SK, Witness Qaku X, et al. Quality enhancement of prickly pears (Opuntia sp.) juice through probiotic fermentation using Lactobacillus fermentum - ATCC 9338. Lebensm Wiss Technol 2017; 75: 453-9.
[http://dx.doi.org/10.1016/j.lwt.2016.09.026]
[59]
Sarin R, Sharma M, Singh R, Kumar S. Nutraceuticals: A review. Int Res J Pharm 2012; 3: 95-9.
[http://dx.doi.org/10.20959/wjpr201911-15964]
[60]
Silvester AJ, Aseer KR, Yun JW. Dietary polyphenols and their roles in fat browning. J Nutr Biochem 2019; 64: 1-12.
[http://dx.doi.org/10.1016/j.jnutbio.2018.09.028] [PMID: 30414469]
[61]
Wang S, Moustaid-Moussa N, Chen L, et al. Novel insights of dietary polyphenols and obesity. J Nutr Biochem 2014; 25(1): 1-18.
[http://dx.doi.org/10.1016/j.jnutbio.2013.09.001] [PMID: 24314860]
[62]
Haber SL, Awwad O, Phillips A, Park AE, Pham TM. Garcinia cambogia for weight loss. Am J Health Syst Pharm 2018; 75(2): 17-22.
[http://dx.doi.org/10.2146/ajhp160915] [PMID: 29317394]
[63]
Pokusaeva K, Fitzgerald GF, van Sinderen D. Carbohydrate metabolism in Bifidobacteria. Genes Nutr 2011; 6(3): 285-306.
[http://dx.doi.org/10.1007/s12263-010-0206-6] [PMID: 21484167]
[64]
Márquez F, Babio N, Bulló M, Salas-Salvadó J. Evaluation of the safety and efficacy of hydroxycitric acid or Garcinia cambogia extracts in humans. Crit Rev Food Sci Nutr 2012; 52(7): 585-94.
[http://dx.doi.org/10.1080/10408398.2010.500551] [PMID: 22530711]
[65]
Han J, Li L, Wang D, Ma H. (−)-Hydroxycitric acid reduced fat deposition via regulating lipid metabolism-related gene expression in broiler chickens. Lipids Health Dis 2016; 15(1): 37.
[http://dx.doi.org/10.1186/s12944-016-0208-5] [PMID: 26912252]
[66]
Alappat L, Awad AB. Curcumin and obesity: Evidence and mechanisms. Nutr Rev 2010; 68(12): 729-38.
[http://dx.doi.org/10.1111/j.1753-4887.2010.00341.x] [PMID: 21091916]
[67]
Weisberg SP, Leibel R, Tortoriello DV. Dietary curcumin significantly improves obesity-associated inflammation and diabetes in mouse models of diabesity. Endocrinology 2008; 149(7): 3549-58.
[http://dx.doi.org/10.1210/en.2008-0262] [PMID: 18403477]
[68]
Shao W, Yu Z, Chiang Y, et al. Curcumin prevents high fat diet induced insulin resistance and obesity via attenuating lipogenesis in liver and inflammatory pathway in adipocytes. PLoS One 2012; 7(1): e28784.
[http://dx.doi.org/10.1371/journal.pone.0028784] [PMID: 22253696]
[69]
Hsu CL, Yen GC. Effects of capsaicin on induction of apoptosis and inhibition of adipogenesis in 3T3-L1 cells. J Agric Food Chem 2007; 55(5): 1730-6.
[http://dx.doi.org/10.1021/jf062912b] [PMID: 17295509]
[70]
Ohyama K, Nogusa Y, Suzuki K, Shinoda K, Kajimura S, Bannai M. A combination of exercise and capsinoid supplementation additively suppresses diet-induced obesity by increasing energy expenditure in mice. Am J Physiol Endocrinol Metab 2015; 308(4): E315-23.
[http://dx.doi.org/10.1152/ajpendo.00354.2014] [PMID: 25516550]
[71]
Rains TM, Agarwal S, Maki KC. Antiobesity effects of green tea catechins: A mechanistic review. J Nutr Biochem 2011; 22(1): 1-7.
[http://dx.doi.org/10.1016/j.jnutbio.2010.06.006] [PMID: 21115335]
[72]
Ebrahimzadeh Attari V, Asghari Jafarabadi M, Zemestani M, Ostadrahimi A. Effect of Zingiber officinale supplementation on obesity management with respect to the uncoupling protein 1-3826A> G and ß3-adrenergic receptor Trp64Arg polymorphism. Phytother Res 2015; 29(7): 1032-9.
[http://dx.doi.org/10.1002/ptr.5343] [PMID: 25899896]
[73]
Semwal RB, Semwal DK, Vermaak I, Viljoen A. A comprehensive scientific overview of Garcinia cambogia. Fitoterapia 2015; 102: 134-48.
[http://dx.doi.org/10.1016/j.fitote.2015.02.012] [PMID: 25732350]
[74]
Ademosun AO, Oboh G, Bello F, Ayeni PO. Antioxidative properties and effect of quercetin and its glycosylated form (Rutin) on acetylcholinesterase and butyrylcholinesterase activities. J Evid Based Complement Altern Med 2016; 21(4): NP11-7.
[http://dx.doi.org/10.1177/2156587215610032] [PMID: 26438716]
[75]
Wang W, Sun C, Mao L, et al. The biological activities, chemical stability, metabolism and delivery systems of quercetin: A review. Trends Food Sci Technol 2016; 56(1-2): 21-38.
[http://dx.doi.org/10.1016/j.tifs.2016.07.004]
[76]
Barceloux DG. Pepper and capsaicin (Capsicum and Piper species). Dis Mon 2009; 55(6): 380-90.
[http://dx.doi.org/10.1016/j.disamonth.2009.03.008] [PMID: 19446682]
[77]
Bradford PG. Curcumin and obesity. Biofactors 2013; 39(1): 78-87.
[http://dx.doi.org/10.1002/biof.1074] [PMID: 23339049]
[78]
Aggarwal BB. Targeting inflammation-induced obesity and metabolic diseases by curcumin and other nutraceuticals. Annu Rev Nutr 2010; 30(1): 173-99.
[http://dx.doi.org/10.1146/annurev.nutr.012809.104755] [PMID: 20420526]
[79]
Aziz HA, Peh KK, Tan YTF. Herbal delivery system for treatment of obesity administration of encapsulated khat-extracts on body weight of rats. Obes Res Clin Pract 2011; 5(4): e305-12.
[http://dx.doi.org/10.1016/j.orcp.2011.03.008] [PMID: 24331133]
[80]
Gamboa-Gómez CI, Rocha-Guzmán NE, Gallegos-Infante JA, Moreno-Jiménez MR, Vázquez-Cabral BD, González-Laredo RF. Plants with potential use on obesity and its complications. EXCLI J 2015; 14: 809-31.
[http://dx.doi.org/10.171792/Fexcli2015-186] [PMID: 26869866]
[81]
Rossato LG, Costa VM, Limberger RP, Bastos ML, Remião F. Synephrine: From trace concentrations to massive consumption in weight-loss. Food Chem Toxicol 2011; 49(1): 8-16.
[http://dx.doi.org/10.1016/j.fct.2010.11.007] [PMID: 21075161]
[82]
Ju EM, Lee SE, Hwang HJ, Kim JH. Antioxidant and anticancer activity of extract from Betula platyphylla var. japonica. Life Sci 2004; 74(8): 1013-26.
[http://dx.doi.org/10.1016/j.lfs.2003.07.025] [PMID: 14672757]
[83]
Baek SC, Choi E, Eom HJ, et al. LC/MS-based analysis of bioactive compounds from the bark of Betula platyphylla var. japonica and their effects on regulation of adipocyte and osteoblast differentiation. Nat Prod Sci 2018; 24(4): 235-40.
[http://dx.doi.org/10.20307/nps.2018.24.4.235]
[84]
Golzarand M, Omidian M, Toolabi K. Effect of Garcinia cambogia supplement on obesity indices: A systematic review and dose-response meta-analysis. Complement Ther Med 2020; 52: 102451.
[http://dx.doi.org/10.1016/j.ctim.2020.102451] [PMID: 32951714]
[85]
Sripradha R, Magadi SG. Efficacy of Garcinia cambogia on body weight, inflammation and glucose tolerance in high fat fed male wistar rats. J Clin Diagn Res 2015; 9(2): BF01-4.
[http://dx.doi.org/10.7860/JCDR/2015/12045.5577] [PMID: 25859449]
[86]
Tomar M, Rao RP, Dorairaj P, et al. Correction: A clinical and computational study on anti-obesity effects of hydroxycitric acid. RSC Advances 2019; 9(39): 22288.
[http://dx.doi.org/10.1039/C9RA90051A] [PMID: 35532504]
[87]
Ariyanto EF, Multom NO, Berbudi A, Rohmawaty E, Sujatmiko B. Efficacy and mechanism of action of Aloe vera, Cinnamomum Zeylanicum, Curcuma longa, Garcinia cambogia, and Garcinia mangostana extracts in lowering body weight in obesity: A literature review. Biomed Pharmacol J 2021; 14(4): 1791-7.
[http://dx.doi.org/10.13005/bpj/2278]
[88]
Lee JH, Kim BH, Yoon YC, et al. Effects against obesity and diabetes of red pepper (Capsicum annuum L.) fermented with lactic acid bacteria. J Life Sci 2019; 29(3): 354-61.
[http://dx.doi.org/10.5352/JLS.2019.29.3.354]
[89]
Lee MS, Jung S, Shin Y, et al. Lipolytic efficacy of alginate double-layer nanoemulsion containing oleoresin capsicum in differentiated 3T3-L1 adipocytes. Food Nutr Res 2017; 29.
[http://dx.doi.org/10.1080/16546628.2017.1339553]
[90]
Hwang JT, Park IJ, Shin JI, et al. Genistein, EGCG, and capsaicin inhibit adipocyte differentiation process via activating AMP-activated protein kinase. Biochem Biophys Res Commun 2005; 338(2): 694-9.
[http://dx.doi.org/10.1016/j.bbrc.2005.09.195] [PMID: 16236247]
[91]
Reza Ariamoghaddam A, Ebrahimi-Hosseinzadeh B, Hatamian-Zarmi A, Sahraeian R. In vivo anti-obesity efficacy of curcumin loaded nanofibers transdermal patches in high-fat diet induced obese rats. Mater Sci Eng C 92: 161-71.
[http://dx.doi.org/10.1016/j.msec.2018.06.030]
[92]
Kim JH, Kim OK, Yoon HG, et al. Anti-obesity effect of extract from fermented Curcuma longa L. through regulation of adipogenesis and lipolysis pathway in high-fat-diet-induced obese rats. Food Nutr Res 2016; 60(1): 30428.
[http://dx.doi.org/10.3402/fnr.v60.30428] [PMID: 26822962]
[93]
Ejaz A, Wu D, Kwan P, Meydani M. Curcumin inhibits adipogenesis in 3T3-L1 adipocytes and angiogenesis and obesity in C57/BL mice. J Nutr 2009; 139(5): 919-25.
[http://dx.doi.org/10.3945/jn.108.100966] [PMID: 19297423]
[94]
Dinh TC, Thi Phuong TN, Minh LB, et al. The effects of green tea on lipid metabolism and its potential applications for obesity and related metabolic disorders - An existing update. Diabetes Metab Syndr 2019; 13(2): 1667-73.
[http://dx.doi.org/10.1016/j.dsx.2019.03.021] [PMID: 31336539]
[95]
Neyrinck AM, Bindels LB, Geurts L, Van Hul M, Cani PD, Delzenne NM. A polyphenolic extract from green tea leaves activates fat browning in high-fat-diet-induced obese mice. J Nutr Biochem 2017; 49: 15-21.
[http://dx.doi.org/10.1016/j.jnutbio.2017.07.008] [PMID: 28863365]
[96]
Diepvens K, Westerterp KR, Westerterp-Plantenga MS. Obesity and thermogenesis related to the consumption of caffeine, ephedrine, capsaicin, and green tea. Am J Physiol Regul Integr Comp Physiol 2007; 292: 77-85.
[http://dx.doi.org/10.1152/ajpregu.00832.2005]
[97]
Ebrahimzadeh AV, Ostadrahimi A, Asghari JM, Mehralizadeh S, Mahluji S. Changes of serum adipocytokines and body weight following Zingiber officinale supplementation in obese women: A RCT. Eur J Nutr 2016; 55(6): 2129-36.
[http://dx.doi.org/10.1007/s00394-015-1027-6] [PMID: 26318445]
[98]
Suk S, Kwon GT, Lee E, et al. Gingerenone A, a polyphenol present in ginger, suppresses obesity and adipose tissue inflammation in high-fat diet-fed mice. Mol Nutr Food Res 2017; 61(10): 1700139.
[http://dx.doi.org/10.1002/mnfr.201700139] [PMID: 28556482]
[99]
Seo SH, Fang F, Kang I. Ginger (Zingiber officinale) attenuates obesity and adipose tissue remodeling in high-fat diet-fed C57BL/6 Mice. Int J Environ Res Public Health 2021; 18(2): 631.
[http://dx.doi.org/10.3390/ijerph18020631] [PMID: 33451038]
[100]
Lee D, Kwak HJ, Kim BH, Kim SH, Kim DW, Kang KS. Combined anti-adipogenic effects of Hispidulin and p-Synephrine on 3T3-L1 adipocytes. Biomolecules 2021; 11(12): 1764.
[http://dx.doi.org/10.3390/biom11121764] [PMID: 34944408]
[101]
Onakpoya I, Davies L, Ernst E. Efficacy of herbal supplements containing Citrus aurantium and synephrine alkaloids for the management of overweight and obesity: A systematic review. Focus Altern Complement Ther 2011; 16(4): 254-60.
[http://dx.doi.org/10.1111/j.2042-7166.2011.01115.x]
[102]
Zhao L, Zhang Q, Ma W, Tian F, Shen H, Zhou M. A combination of quercetin and resveratrol reduces obesity in high-fat diet-fed rats by modulation of gut microbiota. Food Funct 2017; 8(12): 4644-56.
[http://dx.doi.org/10.1039/C7FO01383C] [PMID: 29152632]
[103]
Jeon BT, Jeong EA, Shin HJ, et al. Resveratrol attenuates obesity-associated peripheral and central inflammation and improves memory deficit in mice fed a high-fat diet. Diabetes 2012; 61(6): 1444-54.
[http://dx.doi.org/10.2337/db11-1498] [PMID: 22362175]
[104]
Yang JY, Della-Fera MA, Rayalam S, et al. Enhanced inhibition of adipogenesis and induction of apoptosis in 3T3-L1 adipocytes with combinations of resveratrol and quercetin. Life Sci 2008; 82(19-20): 1032-9.
[http://dx.doi.org/10.1016/j.lfs.2008.03.003] [PMID: 18433793]
[105]
Park SJ, Ahmad F, Philp A, et al. Resveratrol ameliorates aging-related metabolic phenotypes by inhibiting cAMP phosphodiesterases. Cell 2012; 148(3): 421-33.
[http://dx.doi.org/10.1016/j.cell.2012.01.017] [PMID: 22304913]
[106]
Zhao H, Wang Y, Peng J, et al. Biodegradable self-assembled micelles based on MPEG-PTMC copolymers: An ideal drug delivery system for vincristine. J Biomed Nanotechnol 2017; 13(4): 427-36.
[http://dx.doi.org/10.1166/jbn.2017.2357] [PMID: 29384619]
[107]
Saravanan M, Pandikumar P, Saravanan S, Toppo E, Pazhanivel N, Ignacimuthu S. Lipolytic and antiadipogenic effects of (3,3-dimethylallyl) halfordinol on 3T3-L1 adipocytes and high fat and fructose diet induced obese C57/BL6J mice. Eur J Pharmacol 2014; 740: 714-21.
[http://dx.doi.org/10.1016/j.ejphar.2014.06.004] [PMID: 24952133]
[108]
Karmase A, Jagtap S, Bhutani KK. Anti adipogenic activity of Aegle marmelos Correa. Phytomedicine 2013; 20(14): 1267-71.
[http://dx.doi.org/10.1016/j.phymed.2013.07.011] [PMID: 23972792]
[109]
Cho AS, Jeon SM, Kim MJ, et al. Chlorogenic acid exhibits anti-obesity property and improves lipid metabolism in high-fat diet-induced-obese mice. Food Chem Toxicol 2010; 48(3): 937-43.
[http://dx.doi.org/10.1016/j.fct.2010.01.003] [PMID: 20064576]
[110]
Wang W, Pan Y, Wang L, et al. Optimal dietary ferulic acid for suppressing the obesity-related disorders in leptin-deficient obese C57BL/6J-ob/ob mice. J Agric Food Chem 2019; 67(15): 4250-8.
[http://dx.doi.org/10.1021/acs.jafc.8b06760] [PMID: 30907082]
[111]
He X, Zheng S, Sheng Y, et al. Chlorogenic acid ameliorates obesity by preventing energy balance shift in high‐fat diet induced obese mice. J Sci Food Agric 2021; 101(2): 631-7.
[http://dx.doi.org/10.1002/jsfa.10675] [PMID: 32683698]
[112]
Wang W, Pan Y, Zhou H, et al. Ferulic acid suppresses obesity and obesity-related metabolic syndromes in high fat diet-induced obese C57BL/6J mice. Food Agric Immunol 2018; 29(1): 1116-25.
[http://dx.doi.org/10.1080/09540105.2018.1516739]
[113]
Wang Z, Lam KL, Hu J, et al. Chlorogenic acid alleviates obesity and modulates gut microbiota in high‐fat‐fed mice. Food Sci Nutr 2019; 7(2): 579-88.
[http://dx.doi.org/10.1002/fsn3.868] [PMID: 30847137]
[114]
Seo CR, Yi BR, Oh S, et al. Aqueous extracts of hulled barley containing coumaric acid and ferulic acid inhibit adipogenesis in vitro and obesity in vivo. J Funct Foods 2015; 12: 208-18.
[http://dx.doi.org/10.1016/j.jff.2014.11.022]
[115]
Sheng L, Qian Z, Zheng S, Xi L. Mechanism of hypolipidemic effect of crocin in rats: Crocin inhibits pancreatic lipase. Eur J Pharmacol 2006; 543(1-3): 116-22.
[http://dx.doi.org/10.1016/j.ejphar.2006.05.038] [PMID: 16828739]
[116]
Mashmoul M, Azlan A, Yusof BNM, Khaza’ai H, Mohtarrudin N, Boroushaki MT. Effects of saffron extract and crocin on anthropometrical, nutritional and lipid profile parameters of rats fed a high fat diet. J Funct Foods 2014; 8: 180-7.
[http://dx.doi.org/10.1016/j.jff.2014.03.017]
[117]
Gul T, Balkhi HM, Haq E. Inhibition of adipocyte differentiation by crocin in in vitro model of obesity. Saudi J Life Sci 2017; 2: 306-11.
[118]
Alshagga MA, Mohamed Z, Seyedan A, Ebling FJP, Alshawsh MA. Khat (Catha edulis) upregulates lipolytic genes in white adipose tissue of male obese mice (C57BL/6J). J Ethnopharmacol 2020; 262: 113187.
[http://dx.doi.org/10.1016/j.jep.2020.113187] [PMID: 32730892]
[119]
de Melo CL, Queiroz MGR, Arruda Filho ACV, et al. Betulinic acid, a natural pentacyclic triterpenoid, prevents abdominal fat accumulation in mice fed a high-fat diet. J Agric Food Chem 2009; 57(19): 8776-81.
[http://dx.doi.org/10.1021/jf900768w] [PMID: 19754196]
[120]
Kim J, Lee YS, Kim CS, Kim JS. Betulinic acid has an inhibitory effect on pancreatic lipase and induces adipocyte lipolysis. Phytother Res 2012; 26(7): 1103-6.
[http://dx.doi.org/10.1002/ptr.3672] [PMID: 22114077]
[121]
Kim KD, Jung HY, Ryu HG, et al. Betulinic acid inhibits high-fat diet-induced obesity and improves energy balance by activating AMPK. Nutr Metab Cardiovasc Dis 2019; 29(4): 409-20.
[http://dx.doi.org/10.1016/j.numecd.2018.12.001] [PMID: 30799179]
[122]
Choi YJ, Park SY, Kim JY, et al. Combined treatment of betulinic acid, a PTP1B inhibitor, with Orthosiphon stamineus extract decreases body weight in high-fat-fed mice. J Med Food 2013; 16(1): 2-8.
[http://dx.doi.org/10.1089/jmf.2012.2384] [PMID: 23256448]
[123]
Ibarra A, Cases J, Roller M, Chiralt-Boix A, Coussaert A, Ripoll C. Carnosic acid-rich rosemary (Rosmarinus officinalis L.) leaf extract limits weight gain and improves cholesterol levels and glycaemia in mice on a high-fat diet. Br J Nutr 2011; 106(8): 1182-9.
[http://dx.doi.org/10.1017/S0007114511001620] [PMID: 21676274]
[124]
Cho BY, Park MR, Lee JH, et al. Standardized Cirsium setidens Nakai ethanolic extract suppresses adipogenesis and regulates lipid metabolisms in 3T3-L1 adipocytes and C57BL/6J mice fed high-fat diets. J Med Food 2017; 20(8): 763-76.
[http://dx.doi.org/10.1089/jmf.2017.3965] [PMID: 28686516]
[125]
Yoo YM, Nam JH, Kim MY, Choi J, Park HJ. Pectolinarin and pectolinarigenin of cirsium setidens prevent the hepatic injury in rats caused by d-galactosamine via an antioxidant mechanism. Biol Pharm Bull 2008; 31(4): 760-4.
[http://dx.doi.org/10.1248/bpb.31.760] [PMID: 18379079]
[126]
Lee M, Sung SH. Platyphylloside isolated from Betula platyphylla inhibit adipocyte differentiation and induce lipolysis via regulating adipokines including PPARγ in 3T3-L1 Cells. Pharmacogn Mag 2016; 12(48): 276-81.
[http://dx.doi.org/10.4103/2F0973-1296.192208] [PMID: 27867269]
[127]
Huh JY, Lee S, Ma EB, et al. The effects of phenolic glycosides from Betula platyphylla var. japonica on adipocyte differentiation and mature adipocyte metabolism. J Enzyme Inhib Med Chem 2018; 33(1): 1167-73.
[http://dx.doi.org/10.1080/14756366.2018.1491846] [PMID: 30126307]
[128]
De Angelis M, Schriever SC, Kyriakou E, et al. Detection and quantification of the anti-obesity drug celastrol in murine liver and brain. Neurochem Int 2020; 136: 104713.
[http://dx.doi.org/10.1016/j.neuint.2020.104713] [PMID: 32151623]
[129]
Wen C, Wang D, Li X, Huang T, Huang C, Hu K. Targeted isolation and identification of bioactive compounds lowering cholesterol in the crude extracts of crabapples using UPLC-DAD-MS-SPE/NMR based on pharmacology-guided PLS-DA. J Pharm Biomed Anal 2018; 150: 144-51.
[http://dx.doi.org/10.1016/j.jpba.2017.11.061] [PMID: 29232626]
[130]
Ding Y, Gu Z, Wang Y, et al. Clove extract functions as a natural fatty acid synthesis inhibitor and prevents obesity in a mouse model. Food Funct 2017; 8(8): 2847-56.
[http://dx.doi.org/10.1039/C7FO00096K] [PMID: 28726934]
[131]
Nepali S, Cha JY, Ki HH, et al. Chrysanthemum indicum inhibits adipogenesis and activates the AMPK pathway in high-fat-diet-induced obese mice. Am J Chin Med 2018; 46(1): 119-36.
[http://dx.doi.org/10.1142/S0192415X18500076] [PMID: 29298511]
[132]
Nepali S, Kim DK, Lee HY, et al. Euphorbia supina extract results in inhibition of high-fat-diet-induced obesity in mice. Int J Mol Med 2018; 41(5): 2952-60.
[http://dx.doi.org/10.3892/ijmm.2018.3495] [PMID: 29484428]
[133]
Vitalone A, Di Sotto A, Mammola CL, et al. Phytochemical analysis and effects on ingestive behaviour of a Caralluma fimbriata extract. Food Chem Toxicol 2017; 108(Pt A): 63-73.
[http://dx.doi.org/10.1016/j.fct.2017.07.027] [PMID: 28713048]
[134]
Yimam M, Jiao P, Hong M, et al. A botanical composition from Morus alba, Ilex paraguariensis, and Rosmarinus officinalis for body weight management. J Med Food 2017; 20(11): 1100-12.
[http://dx.doi.org/10.1089/jmf.2017.0002] [PMID: 28708468]
[135]
Kim GN, Shin MR, Shin SH, et al. Study of antiobesity effect through inhibition of pancreatic lipase activity of Diospyros kaki fruit and Citrus unshiu peel. BioMed Res Int 2016; 2016: 1-7.
[http://dx.doi.org/10.1155/2016/1723042] [PMID: 27529064]
[136]
Rahman HA, Sahib NG, Saari N, et al. Anti-obesity effect of ethanolic extract from Cosmos caudatus Kunth leaf in lean rats fed a high fat diet. BMC Complement Altern Med 2017; 17(1): 122.
[http://dx.doi.org/10.1186/s12906-017-1640-4] [PMID: 28228098]
[137]
Lim HH, Lee SO, Kim SY, Yang SJ, Lim Y. Anti-inflammatory and antiobesity effects of mulberry leaf and fruit extract on high fat diet-induced obesity. Exp Biol Med (Maywood) 2013; 238(10): 1160-9.
[http://dx.doi.org/10.1177/1535370213498982] [PMID: 24000381]
[138]
Du H, You JS, Zhao X, Park JY, Kim SH, Chang KJ. Antiobesity and hypolipidemic effects of lotus leaf hot water extract with taurine supplementation in rats fed a high fat diet. J Biomed Sci 2010; 17(1) (Suppl. 1): S42.
[http://dx.doi.org/10.1186/1423-0127-17-S1-S42] [PMID: 20804619]
[139]
Monsef R, Salavati-Niasari M. Hydrothermal architecture of Cu5V2O10 nanostructures as new electro-sensing catalysts for voltammetric quantification of mefenamic acid in pharmaceuticals and biological samples. Biosens Bioelectron 2021; 178: 113017.
[http://dx.doi.org/10.1016/j.bios.2021.113017] [PMID: 33493895]
[140]
Salavati-Niasari M, Davar F. In situ one-pot template synthesis (IOPTS) and characterization of copper(II) complexes of 14-membered hexaaza macrocyclic ligand “3,10-dialkyl-dibenzo-1,3,5,8,10,12-hexaazacyclotetradecane”. Inorg Chem Commun 2006; 9(2): 175-9.
[http://dx.doi.org/10.1016/j.inoche.2005.10.028]
[141]
Ahmadian-Fard-Fini S, Ghanbari D, Amiri O, Salavati-Niasari M. Electro-spinning of cellulose acetate nanofibers/Fe/carbon dot as photoluminescence sensor for mercury (II) and lead (II) ions. Carbohydr Polym 2020; 229: 115428.
[http://dx.doi.org/10.1016/j.carbpol.2019.115428] [PMID: 31826498]
[142]
Kumar S, Kelly AS. Review of childhood obesity: From epidemiology, etiology, and comorbidities to clinical assessment and treatment. Mayo Clin Proc 2017; 92(2): 251-65.
[http://dx.doi.org/10.1016/j.mayocp.2016.09.017] [PMID: 28065514]
[143]
Farokhzad OC, Langer R. Impact of nanotechnology on drug delivery. ACS Nano 2009; 3(1): 16-20.
[http://dx.doi.org/10.1021/nn900002m] [PMID: 19206243]
[144]
Ting Y, Jiang Y, Ho CT, Huang Q. Common delivery systems for enhancing in vivo bioavailability and biological efficacy of nutraceuticals. J Funct Foods 2014; 7: 112-28.
[http://dx.doi.org/10.1016/j.jff.2013.12.010]
[145]
Etheridge ML, Campbell SA, Erdman AG, Haynes CL, Wolf SM, McCullough J. The big picture on nanomedicine: The state of investigational and approved nanomedicine products. Nanomedicine 2013; 9(1): 1-14.
[http://dx.doi.org/10.1016/j.nano.2012.05.013] [PMID: 22684017]
[146]
Rezvani Z, Venugopal JR, Urbanska AM, Mills DK, Ramakrishna S, Mozafari M. A bird’s eye view on the use of electrospun nanofibrous scaffolds for bone tissue engineering: Current state‐of‐the‐art, emerging directions and future trends. Nanomedicine 2016; 12(7): 2181-200.
[http://dx.doi.org/10.1016/j.nano.2016.05.014] [PMID: 27247186]
[147]
Kumar Teli M, Mutalik S, Rajanikant GK. Nanotechnology and nanomedicine: Going small means aiming big. Curr Pharm Des 2010; 16(16): 1882-92.
[http://dx.doi.org/10.2174/138161210791208992] [PMID: 20222866]
[148]
Kargozar S, Mozafari M. Nanotechnology and Nanomedicine: Start small, think big. Mater Today Proc 2018; 5(7): 15492-500.
[http://dx.doi.org/10.1016/j.matpr.2018.04.155]
[149]
Lingayat VJ, Zarekar NS, Shendge RS. Solid lipid nanoparticles: A review. J Nanosci Nanotechnol 2017; 2: 67-72.
[http://dx.doi.org/10.12691/nnr-4-2-5]
[150]
Singh N, Khullar N, Kakkar V, Kaur IP. Sesamol loaded solid lipid nanoparticles: A promising intervention for control of carbon tetrachloride induced hepatotoxicity. BMC Complement Altern Med 2015; 15(1): 142.
[http://dx.doi.org/10.1186/s12906-015-0655-y] [PMID: 25935744]
[151]
Singh N, Khullar N, Kakkar V, Kaur IP. Hepatoprotective effects of sesamol loaded solid lipid nanoparticles in carbon tetrachloride induced sub-chronic hepatotoxicity in rats. Environ Toxicol 2014; 31(5) n/a.
[http://dx.doi.org/10.1002/tox.22064] [PMID: 25410024]
[152]
Kakkar V, Muppu SK, Chopra K, Kaur IP. Curcumin loaded solid lipid nanoparticles: An efficient formulation approach for cerebral ischemic reperfusion injury in rats. Eur J Pharm Biopharm 2013; 85(3): 339-45.
[http://dx.doi.org/10.1016/j.ejpb.2013.02.005] [PMID: 23454202]
[153]
Eloy JO, Claro de Souza M, Petrilli R, Barcellos JPA, Lee RJ, Marchetti JM. Liposomes as carriers of hydrophilic small molecule drugs: Strategies to enhance encapsulation and delivery. Colloids Surf B Biointerfaces 2014; 123: 345-63.
[http://dx.doi.org/10.1016/j.colsurfb.2014.09.029] [PMID: 25280609]
[154]
Vanić Ž, Hafner A, Bego M, Škalko-Basnet N. Characterization of various deformable liposomes with metronidazole. Drug Dev Ind Pharm 2013; 39(3): 481-8.
[http://dx.doi.org/10.3109/03639045.2012.670247] [PMID: 22455377]
[155]
Vrignaud S, Benoit JP, Saulnier P. Strategies for the nanoencapsulation of hydrophilic molecules in polymer-based nanoparticles. Biomaterials 2011; 32(33): 8593-604.
[http://dx.doi.org/10.1016/j.biomaterials.2011.07.057] [PMID: 21831421]
[156]
Zhou X, Hao Y, Yuan L, et al. Nano-formulations for transdermal drug delivery: A review. Chin Chem Lett 2018; 29(12): 1713-24.
[http://dx.doi.org/10.1016/j.cclet.2018.10.037]
[157]
Santos P, Watkinson AC, Hadgraft J, Lane ME. Application of microemulsions in dermal and transdermal drug delivery. Skin Pharmacol Physiol 2008; 21(5): 246-59.
[http://dx.doi.org/10.1159/000140228] [PMID: 18562799]
[158]
Voon SH, Tiew SX, Kue CS, et al. Chitosan-coated poly (lactic-co-glycolic acid)-diiodinated boron-dipyrromethene nanoparticles improve tumor selectivity and stealth properties in photodynamic cancer therapy. J Biomed Nanotechnol 2016; 12(7): 1431-52.
[http://dx.doi.org/10.1166/jbn.2016.2263] [PMID: 29336539]
[159]
Singh Y, Meher JG, Raval K, et al. Nanoemulsion: Concepts, development and applications in drug delivery. J Control Release 2017; 252: 28-49.
[http://dx.doi.org/10.1016/j.jconrel.2017.03.008] [PMID: 28279798]
[160]
Li Y, Ai L, Yokoyama W, et al. Properties of chitosan-microencapsulated orange oil prepared by spray-drying and its stability to detergents. J Agric Food Chem 2013; 61(13): 3311-9.
[http://dx.doi.org/10.1021/jf305074q] [PMID: 23473289]
[161]
Bouchemal K, Briançon S, Perrier E, Fessi H. Nano-emulsion formulation using spontaneous emulsification: Solvent, oil and surfactant optimisation. Int J Pharm 2004; 280(1-2): 241-51.
[http://dx.doi.org/10.1016/j.ijpharm.2004.05.016] [PMID: 15265563]
[162]
Anton N, Benoit JP, Saulnier P. Design and production of nanoparticles formulated from nano-emulsion templates-A review. J Control Release 2008; 128(3): 185-99.
[http://dx.doi.org/10.1016/j.jconrel.2008.02.007] [PMID: 18374443]
[163]
Blaiszik BJ, Sottos NR, White SR. Nanocapsules for self-healing materials. Compos Sci Technol 2008; 68(3-4): 978-86.
[http://dx.doi.org/10.1016/j.compscitech.2007.07.021]
[164]
Mora-Huertas CE, Fessi H, Elaissari A. Polymer-based nanocapsules for drug delivery. Int J Pharm 2010; 385(1-2): 113-42.
[http://dx.doi.org/10.1016/j.ijpharm.2009.10.018] [PMID: 19825408]
[165]
Hassanzadeh P, Kharaziha M, Nikkhah M, et al. Chitin nanofiber micropatterned flexible substrates for tissue engineering. J Mater Chem B Mater Biol Med 2013; 1(34): 4217-24.
[http://dx.doi.org/10.1039/c3tb20782j] [PMID: 24179675]
[166]
Shah S, Yin PT, Uehara TM, Chueng STD, Yang L, Lee KB. Guiding stem cell differentiation into oligodendrocytes using graphene-nanofiber hybrid scaffolds. Adv Mater 2014; 26(22): 3673-80.
[http://dx.doi.org/10.1002/adma.201400523] [PMID: 24668911]
[167]
Yang X, Zou W, Su Y, et al. Activated nitrogen-doped carbon nanofibers with hierarchical pore as efficient oxygen reduction reaction catalyst for microbial fuel cells. J Power Sources 2014; 266: 36-42.
[http://dx.doi.org/10.1016/j.jpowsour.2014.04.126]
[168]
Cheng L, Ma SY, Wang TT, et al. Synthesis and characterization of SnO2 hollow nanofibers by electrospinning for ethanol sensing properties. Mater Lett 2014; 131: 23-6.
[http://dx.doi.org/10.1016/j.matlet.2014.05.151]
[169]
Shang M, Wang W, Sun S, et al. The design and realization of a large-area flexible nanofiber-based mat for pollutant degradation: An application in photocatalysis. Nanoscale 2013; 5(11): 5036-42.
[http://dx.doi.org/10.1039/c3nr00503h] [PMID: 23640283]
[170]
Liu Y, Zhao L, Li M, Guo L. TiO2/CdSe core-shell nanofiber film for photoelectrochemical hydrogen generation. Nanoscale 2014; 6(13): 7397-404.
[http://dx.doi.org/10.1039/C4NR00856A] [PMID: 24876070]
[171]
Shi H, Zhou M, Song D, et al. Highly porous SnO2/TiO2 electrospun nanofibers with high photocatalytic activities. Ceram Int 2014; 40(7): 10383-93.
[http://dx.doi.org/10.1016/j.ceramint.2014.02.124]
[172]
Wu Q, Tran T, Lu W, Wu J. Electrospun silicon/carbon/titanium oxide composite nanofibers for lithium ion batteries. J Power Sources 2014; 258: 39-45.
[http://dx.doi.org/10.1016/j.jpowsour.2014.02.047]
[173]
du Preez R, Pahl J, Arora M, Ravi Kumar MNV, Brown L, Panchal SK. Low-dose curcumin nanoparticles normalise blood pressure in male wistar rats with diet-induced metabolic syndrome. Nutrients 2019; 11(7): 1542.
[http://dx.doi.org/10.3390/nu11071542] [PMID: 31288419]
[174]
Li X, Liu Y, Yu Y, Chen W, Liu Y, Yu H. Nanoformulations of quercetin and cellulose nanofibers as healthcare supplements with sustained antioxidant activity. Carbohydr Polym 2019; 207: 160-8.
[http://dx.doi.org/10.1016/j.carbpol.2018.11.084] [PMID: 30599995]
[175]
Kim Y, Kim J-Y, Lee M-S, et al. Anti-obesity efficacy of nanoemulsion oleoresin capsicum in obese rats fed a high-fat diet. Int J Nanomedicine 2014; 9: 301-10.
[http://dx.doi.org/10.2147/IJN.S52414] [PMID: 24403834]
[176]
Sheng F, Chow PS, Hu J, Cheng S, Guo L, Dong Y. Preparation of quercetin nanorod/microcrystalline cellulose formulation via fluid bed coating crystallization for dissolution enhancement. Int J Pharm 2020; 576: 118983.
[http://dx.doi.org/10.1016/j.ijpharm.2019.118983] [PMID: 31874195]
[177]
Singh G, Pai RS. Optimized PLGA nanoparticle platform for orally dosed trans -resveratrol with enhanced bioavailability potential. Expert Opin Drug Deliv 2014; 11(5): 647-59.
[http://dx.doi.org/10.1517/17425247.2014.890588] [PMID: 24661109]
[178]
Pandita D, Kumar S, Poonia N, Lather V. Solid lipid nanoparticles enhance oral bioavailability of resveratrol, a natural polyphenol. Food Res Int 2014; 62: 1165-74.
[http://dx.doi.org/10.1016/j.foodres.2014.05.059]
[179]
Ezhilarasi PN, Muthukumar SP, Anandharamakrishnan C. Solid lipid nanoparticle enhances bioavailability of hydroxycitric acid compared to a microparticle delivery system. RSC Advances 2016; 6(59): 53784-93.
[http://dx.doi.org/10.1039/C6RA04312G]
[180]
Shankar SB, Arivarasu L, Rajeshkumar S. Biosynthesis of hydroxy citric acid mediated Zinc nanoparticles and its antioxidant and cytotoxic activity. J Pharm Res Int 2020; 32: 108-12.
[http://dx.doi.org/10.9734/jpri/2020/v32i2630845]
[181]
Chen CH, Chen CJ, Elzoghby AO, Yeh TS, Fang JY. Self-assembly and directed assembly of lipid nanocarriers for prevention of liver fibrosis in obese rats: A comparison with the therapy of bariatric surgery. Nanomedicine (Lond) 2018; 13(13): 1551-66.
[http://dx.doi.org/10.2217/nnm-2018-0001] [PMID: 29998778]
[182]
Cerri GC, Lima LCF, Lelis DF, et al. Sclareol-loaded lipid nanoparticles improved metabolic profile in obese mice. Life Sci 2019; 218: 292-9.
[http://dx.doi.org/10.1016/j.lfs.2018.12.063] [PMID: 30610871]
[183]
El-Menshawe S, Ali A, Rabeh M, Khalil N. Nanosized soy phytosome-based thermogel as topical anti-obesity formulation: An approach for acceptable level of evidence of an effective novel herbal weight loss product. Int J Nanomedicine 2018; 13: 307-18.
[http://dx.doi.org/10.2147/IJN.S153429] [PMID: 29391791]
[184]
Nallamuthu I, Devi A, Khanum F. Chlorogenic acid loaded chitosan nanoparticles with sustained release property, retained antioxidant activity and enhanced bioavailability. Asian J Pharm Sci 2015; 10(3): 203-11.
[http://dx.doi.org/10.1016/j.ajps.2014.09.005]
[185]
National Library of Medicine (NLM) at the National Institutes of Health (NIH).. Available from: https://clinicaltrials.gov
[186]
Hwang I, Park S. Method for reducing off-flavor of Garcinia cambogia extract and anti-obesity composition comprising Garcinia cambogia extract prepared therefrom. KR1020200089541 2020.
[187]
Wang J, Liang Q, Liang L, Hou L, Li J. Composition with auxiliary hypoglycemic effect and preparation method thereof. CN110934923 2020.
[188]
Wang B. Anti-obesity composition, and preparation method and application thereof. CN111317138 2020.
[189]
Yoon Hyo T, Moon C, Kim Y, Son J. Method for manufacturing functional composition for alleviating obesity using extracts of Gynura procumbens and mori cortex as main ingredients. KR102019008204 2019.
[190]
Kim , Seo GL, Sang BK, Young D. Composition having excellent anti-obesity and body fat reduction effects and comprising extract of peanut sprout. KR1020180098883 2018.
[191]
Kim , Yang HA, Kim Chong S, Kim L, Kim Joo Y, Jung Sun Y. Composition for body fat lipolysis using alginate double-layer nanoemulsions containing oleoresin capsicum. KR1020130103101 2013.
[192]
Shen Junhui Yang, Changqing Qi Xiaolong. Weight-reducing composition with obesity preventing effect and weight-reducing solution. CN102743547 2012.
[193]
Alvarez F, Mariana G. Composition with enhanced thermogenic activity and the use thereof in the prevention and treatment of obesity. US20120219621 2010.
[194]
Dhruva KS. Fat burner herbal composition - external use cream. IN909/DEL/2012 2012.
[195]
Ramanpreet W, Maryam S. A method for preparation of herbal composition for the treatment of obesity. IN201911014033 2021.
[196]
Kajal C. A process to prepare anti-dyslipidemic concentrate marine macroalgae and coastal sea crustacean. IN201911017799 2021.
[197]
Patanjali S, Sundaram C, Jayashree M, et al. Herbal composition. US20200085904 2020.
[198]
Davariar V, Vinoth R, Dinesh KV, Selvaraj A. Novel health promoting functional during formulation. IN533/CHE/2014 2014.
[199]
Jong DK. Pharmaceutical composition for preventing and treating obesity, containing green-tea see husk extract as active ingredient. US20160151439 2016.
[200]
Kim Jong D. Pharmaceutical composition for preventing and treating obesity, containing green-tea seed husk extract as active ingredient. WO2015020489 2015.
[201]
Syed TA, Gurmukh SG. A novel composition for weight management. IN2283/DEL/2013 2015.
[202]
Ho Si H. Formulation, composition or foodstuff additives for the modification of glycemic response methods of manufacturing and using the same. WO2021126078 2021.
[203]
Cichello SA. Oral nicotinamide adenine dinucleotide (NAD+) precursor oral supplementation formulation to support physiological functions associated with aging in humans. CN112022874 2020.
[204]
Zemel Michael B. Compositions, methods and kits for altering adipocytes. WO2021096882 2021.
[205]
Junco B, Liang H, Sara R, Thomas J. Methods and compositions comprising ursolic acid and/or resveratrol for treating obesity, diabetes, or cancer. JP2020147590 2020.
[206]
Chen Y, Jiao J, Wang Z, et al. Weight-reducing medicine for dietinduced obesity as well as preparation method thereof. CN110548039 2019.
[207]
Zhang D. High-efficiency weight-reducing composition and application thereof in weight reduction. CN107822129 2017.
[208]
Zhang L, Gao P, Pan D, Xu Y, Xiao Q. Traditional chinese medicine composition for treating obesity and application thereof. CN108324913 2018.
[209]
Rina HG, Manish AR, Roochi RA. Novel antiobesity herbal composition method of preparation of the same and use thereof. IN1681/MUM/2012 2013.
[210]
Dhruva KS. Fat burner herbal composition-intrnal use medicine. IN1153/DEL/2012 2012.
[211]
Saito C. Crude drug-containing composition and use thereof. JP2010202634 2010.
[212]
Pushpangadan Palpu, Sreedevi Padmavathi, Mehrotra Shanta, Rao Chndana Venkateswara, Ojha Sanjeev Kumar, Govind Raghavan. Antihyperlipidemic and slimming herbal composition(s). IN579/DELNP/2004 2007.
[213]
Neethirajan S, Jayas DS. Nanotechnology for the food and bioprocessing industries. Food Bioprocess Technol 2011; 4(1): 39-47.
[http://dx.doi.org/10.1007/s11947-010-0328-2] [PMID: 32215165]
[214]
Razavi R, Kenari RE, Farmani J, Jahanshahi M. Fabrication of zein/alginate delivery system for nanofood model based on pumpkin. Int J Biol Macromol 2020; 165(Pt B): 3123-34.
[http://dx.doi.org/10.1016/j.ijbiomac.2020.10.176] [PMID: 33127546]
[215]
Low LE, Wong SK, Chew CL, Tang SY. Principles and potential applications of cavitation technology for nano-foods. In: Hebbar U, Ranjan S, Dasgupta N, Kumar Mishra R, Eds. Nano-food Engineering Food Engineering Series. Cham: Springer 2020; pp. 125-52.
[http://dx.doi.org/10.1007/978-3-030-44552-2_5]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy