Acessibilidade / Reportar erro

Impacts of chitosan and its nanoformulations on the metabolic syndromes: a review

Impactos da quitosana e suas nanoformulações nas síndromes metabólicas: uma revisão

Abstract

A significant public health issue worldwide is metabolic syndrome, a cluster of metabolic illnesses that comprises insulin resistance, obesity, dyslipidemia, hyperglycemia, and hypertension. The creation of natural treatments and preventions for metabolic syndrome is crucial. Chitosan, along with its nanoformulations, is an oligomer of chitin, the second-most prevalent polymer in nature, which is created via deacetylation. Due to its plentiful biological actions in recent years, chitosan and its nanoformulations have drawn much interest. Recently, the chitosan nanoparticle-based delivery of CRISPR-Cas9 has been applied in treating metabolic syndromes. The benefits of chitosan and its nanoformulations on insulin resistance, obesity, diabetes mellitus, dyslipidemia, hyperglycemia, and hypertension will be outlined in the present review, highlighting potential mechanisms for the avoidance and medication of the metabolic syndromes by chitosan and its nanoformulations.

Keywords:
chitosan; nano chitosan; metabolic syndrome; obesity; diabetes

Resumo

Uma questão significativa de saúde pública em todo o mundo é a síndrome metabólica, um conjunto de doenças metabólicas que compreende resistência à insulina, obesidade, dislipidemia, hiperglicemia e hipertensão. A criação de tratamentos e prevenções naturais para a síndrome metabólica é crucial. A quitosana, juntamente com suas nanoformulações, é um oligômero de quitina, o segundo polímero mais prevalente na natureza, criado por desacetilação. Devido às suas abundantes ações biológicas nos últimos anos, a quitosana e suas nanoformulações têm despertado muito interesse. Recentemente, a entrega de CRISPR-Cas9 baseada em nanopartículas de quitosana tem sido aplicada no tratamento de síndromes metabólicas. Por isto, os benefícios da quitosana e suas nanoformulações na resistência à insulina, obesidade, diabetes mellitus, dislipidemia, hiperglicemia e hipertensão serão delineados na presente revisão, destacando potenciais mecanismos para evitar e medicação das síndromes metabólicas pela quitosana e suas nanoformulações.

Palavras-chave:
quitosana; nano quitosana; síndrome metabólica; obesidade; diabetes

1. Introduction

Chitosan has numerous health benefits, including its potent antioxidant and antibacterial properties and it's being a non-antigenic, biocompatible, non-toxic, and eco-friendly natural polymer formed from chitin (Chou et al., 2015CHOU, C.-K., CHEN, S.-M., LI, Y.-C., HUANG, T.-C. and LEE, J.-A., 2015. Low-molecular-weight chitosan scavenges methylglyoxal and N ε-(carboxyethyl)lysine, the major factors contributing to the pathogenesis of nephropathy. SpringerPlus, vol. 4, no. 1, pp. 312. http://dx.doi.org/10.1186/s40064-015-1106-4. PMid:25674489.
http://dx.doi.org/10.1186/s40064-015-110...
; Muxika et al., 2017MUXIKA, A., ETXABIDE, A., URANGA, J., GUERRERO, P. and DE LA CABA, K., 2017. Chitosan as a bioactive polymer: processing, properties and applications. International Journal of Biological Macromolecules, vol. 105, no. Pt 2, pp. 1358-1368. http://dx.doi.org/10.1016/j.ijbiomac.2017.07.087. PMid:28735006.
http://dx.doi.org/10.1016/j.ijbiomac.201...
; Guan et al., 2019GUAN, G., AZAD, M.A.K., LIN, Y., KIM, S.W., TIAN, Y., LIU, G. and WANG, H., 2019. Biological effects and applications of chitosan and chito-oligosaccharides. Frontiers in Physiology, vol. 10, pp. 516. http://dx.doi.org/10.3389/fphys.2019.00516. PMid:31133871.
http://dx.doi.org/10.3389/fphys.2019.005...
). While many polysaccharides are neutral or anionic in charge, chitosan is a naturally occurring cationic polymer. Using further synthetic polymers or naturally negatively charged natural materials, this chitosan characteristic enables the construction of multilayer structures or electrostatic complexes (Venkatesan and Kim, 2010VENKATESAN, J. and KIM, S.K., 2010. Chitosan composites for bone tissue engineering: an overview. Marine Drugs, vol. 8, no. 8, pp. 2252-2266. http://dx.doi.org/10.3390/md8082252. PMid:20948907.
http://dx.doi.org/10.3390/md8082252...
). Additionally, chitosan has many biological abilities, such as antibacterial (Amato et al., 2018AMATO, A., MIGNECO, L.M., MARTINELLI, A., PIETRELLI, L., PIOZZI, A. and FRANCOLINI, I., 2018. Antimicrobial activity of catechol functionalized-chitosan versus Staphylococcus epidermidis. Carbohydrate Polymers, vol. 179, pp. 273-281. http://dx.doi.org/10.1016/j.carbpol.2017.09.073. PMid:29111051.
http://dx.doi.org/10.1016/j.carbpol.2017...
; Wei et al., 2019WEI, L., TAN, W., WANG, G., LI, Q., DONG, F. and GUO, Z., 2019. The antioxidant and antifungal activity of chitosan derivatives bearing Schiff bases and quaternary ammonium salts. Carbohydrate Polymers, vol. 226, pp. 115256. http://dx.doi.org/10.1016/j.carbpol.2019.115256. PMid:31582056.
http://dx.doi.org/10.1016/j.carbpol.2019...
), anticancer, and antioxidant properties (Karagozlu and Kim, 2014KARAGOZLU, M.Z. and KIM, S.K., 2014. Anticancer effects of chitin and chitosan derivatives. Advances in Food and Nutrition Research, vol. 72, pp. 215-225. http://dx.doi.org/10.1016/B978-0-12-800269-8.00012-9. PMid:25081085.
http://dx.doi.org/10.1016/B978-0-12-8002...
; Ngo and Kim, 2014NGO, D.H. and KIM, S.K., 2014. Antioxidant effects of chitin, chitosan, and their derivatives. Advances in Food and Nutrition Research, vol. 73, pp. 15-31. http://dx.doi.org/10.1016/B978-0-12-800268-1.00002-0. PMid:25300540.
http://dx.doi.org/10.1016/B978-0-12-8002...
). Chitosan is widely employed in a diversity of biomedical and biological uses, such as a platform for genetic manipulation (Islam et al., 2020ISLAM, M.M., SHAHRUZZAMAN, M., BISWAS, S., SAKIB, M.N. and RASHID, T.U., 2020. Chitosan based bioactive materials in tissue engineering applications-a review. Bioactive Materials, vol. 5, no. 1, pp. 164-183. http://dx.doi.org/10.1016/j.bioactmat.2020.01.012. PMid:32083230.
http://dx.doi.org/10.1016/j.bioactmat.20...
), a drug carrier (Peers et al., 2020PEERS, S., MONTEMBAULT, A. and LADAVIÈRE, C., 2020. Chitosan hydrogels for sustained drug delivery. Journal of Controlled Release, vol. 326, pp. 150-163. http://dx.doi.org/10.1016/j.jconrel.2020.06.012. PMid:32562854.
http://dx.doi.org/10.1016/j.jconrel.2020...
), and water purification (Das et al., 2020DAS, L., DAS, P., BHOWAL, A. and BHATTACHARIEE, C., 2020. Synthesis of hybrid hydrogel nano-polymer composite using Graphene oxide, Chitosan and PVA and its application in waste water treatment. Environmental Technology & Innovation, vol. 18, pp. 100664. http://dx.doi.org/10.1016/j.eti.2020.100664.
http://dx.doi.org/10.1016/j.eti.2020.100...
).

Moreover, chitosan has special potential that makes it safe for usage in biomedical, therapeutics, and wastewater remediation, where chitosan and its derivatives are considered viable sources for creating efficient and secure medication delivery systems because of their unique physical and chemical features (Abd El-Hack et al., 2020ABD EL-HACK, M.E., EL-SAADONY, M.T., SHAFI, M.E., ZABERMAWI, N.M., ARIF, M., BATIHA, G.E., KHAFAGA, A.F., ABD EL-HAKIM, Y.M. and AL-SAGHEER, A.A., 2020. Antimicrobial and antioxidant properties of chitosan and its derivatives and their applications: a review. International Journal of Biological Macromolecules, vol. 164, pp. 2726-2744. http://dx.doi.org/10.1016/j.ijbiomac.2020.08.153. PMid:32841671.
http://dx.doi.org/10.1016/j.ijbiomac.202...
). Likewise, in the utmost available investigation, many previous investigational data disguised that the growth-enhancing properties of chitosan are equivalent to those of dietary antibiotics (Kamal et al., 2023bKAMAL, M., YOUSSEF, I.M., KHALIL, H.A., AYOUB, M.A. and HASHEM, N.M., 2023b. Multifunctional role of chitosan in farm animals: a comprehensive review. Annals of Animal Science, vol. 23, no. 1, pp. 69-86. http://dx.doi.org/10.2478/aoas-2022-0054.
http://dx.doi.org/10.2478/aoas-2022-0054...
, aKAMAL, M., KISHK, W.H., KHALIL, H.A., ABDEL-KHALEK, A.M., AYOUB, M.A., SWELUM, A.A., ALQHTANI, A.H., BA-AWADH, H.A. and ABD EL-HACK, M.E., 2023a. Effect of dietary chitosan supplementation on productive and physiological performance parameters of growing New Zealand white rabbits. International Journal of Biological Macromolecules, vol. 230, pp. 123166. http://dx.doi.org/10.1016/j.ijbiomac.2023.123166. PMid:36623627.
http://dx.doi.org/10.1016/j.ijbiomac.202...
) indicating that chitosan is a practical and operative antibiotic additional.

Nano chitosan is a natural molecule with brilliant physicochemical and biological actions, constructing it a greater naturally friendly substance, and it holds bio-competence action that levels safe on a human being. In addition, it’s commonly utilized as a controlled-release drug transporter for genetic editing in synthetic tissues and immune syndromes. Besides, nano chitosan has been utilized to impart antimicrobial benefits and increase the forte and washability of textiles (Ting and Shen, 2005TING, D.R. and SHEN, Y., 2005. Antibacterial finishing with chitosan derivatives and their nano-particles. Dyeing Finishing, vol. 14, no. 4, pp. 12-14.). Nanosized materials are good, but their impacts on natural organisms and human organs have been investigated. Polysaccharide-coated nanoparticles are recognized to be ecologically kind, much less related to physiological stability, and concerns over toxicity and biodegradability. For instance, chitosan, a natural polysaccharide, is widely used in medical preparations (Swierczewska et al., 2016SWIERCZEWSKA, M., HAN, H.S., KIM, K., PARK, J.H. and LEE, S., 2016. Polysaccharide-based nanoparticles for theranostic nanomedicine. Advanced Drug Delivery Reviews, vol. 99, no. Pt A, pp. 70-84. http://dx.doi.org/10.1016/j.addr.2015.11.015. PMid:26639578.
http://dx.doi.org/10.1016/j.addr.2015.11...
; Shariatinia, 2019SHARIATINIA, Z., 2019. Pharmaceutical applications of chitosan. Advances in Colloid and Interface Science, vol. 263, pp. 131-194. http://dx.doi.org/10.1016/j.cis.2018.11.008. PMid:30530176.
http://dx.doi.org/10.1016/j.cis.2018.11....
).

Chitosan has been generally applied for protein encapsulation, therapeutically enzymes (Koyani et al., 2018KOYANI, R.D., ANDRADE, M., QUESTER, K., GAYTÁN, P., HUERTA-SAQUERO, A. and VAZQUEZ-DUHALT, R., 2018. Surface modification of protein enhances encapsulation in chitosan nanoparticles. Applied Nanoscience, vol. 8, no. 5, pp. 1197-1203. http://dx.doi.org/10.1007/s13204-018-0779-8.
http://dx.doi.org/10.1007/s13204-018-077...
), or as biocatalytic nanoparticles (NPs) (Alarcón-Payán et al., 2017ALARCÓN-PAYÁN, D.A., KOYANI, R.D. and VAZQUEZ-DUHALT, R., 2017. Chitosan-based biocatalytic nanoparticles for pollutant removal from wastewater. Enzyme and Microbial Technology, vol. 100, pp. 71-78. http://dx.doi.org/10.1016/j.enzmictec.2017.02.008. PMid:28284314.
http://dx.doi.org/10.1016/j.enzmictec.20...
). Although non-toxic and biodegradable, chitosan nanoparticles are recognized to accomplish prolonged gradual emancipation of the capacity, escalation bioavailability, and enhance therapeutic effectiveness (Safdar et al., 2019SAFDAR, R., OMAR, A.A., ARUNAGIRI, A., REGUPATHI, I. and THANABALAN, M., 2019. Potential of chitosan and its derivatives for controlled drug release applications: a review. Journal of Drug Delivery Science and Technology, vol. 49, pp. 642-659. http://dx.doi.org/10.1016/j.jddst.2018.10.020.
http://dx.doi.org/10.1016/j.jddst.2018.1...
). To benefit from the affluence usefulness of manufacturing, great revenue at squat cost, and greater loading capability, the manufacturing procedure for nanoparticles such as chitosan NPs might be enhanced (Gallego et al., 2019GALLEGO, I., VILLATE-BEITIA, I., MARTÍNEZ-NAVARRETE, G., MENÉNDEZ, M., LÓPEZ-MÉNDEZ, T., SOTO-SÁNCHEZ, C., ZÁRATE, J., PURAS, G., FERNÁNDEZ, E. and PEDRAZ, J.L., 2019. Non-viral vectors based on cationic niosomes and minicircle DNA technology enhance gene delivery efficiency for biomedical applications in retinal disorders. Nanomedicine: Nanotechnology, Biology, and Medicine, vol. 17, pp. 308-318. http://dx.doi.org/10.1016/j.nano.2018.12.018. PMid:30790710.
http://dx.doi.org/10.1016/j.nano.2018.12...
; Kamel and El-Sayed, 2019KAMEL, M. and EL-SAYED, A., 2019. Utilization of herpesviridae as recombinant viral vectors in vaccine development against animal pathogens. Virus Research, vol. 270, pp. 197648. http://dx.doi.org/10.1016/j.virusres.2019.197648. PMid:31279828.
http://dx.doi.org/10.1016/j.virusres.201...
). According to (Quester et al., 2022QUESTER, K., RODRÍGUEZ-GONZÁLEZ, S., GONZÁLEZ-DÁVALOS, L., LOZANO-FLORES, C., GONZÁLEZ-GALLARDO, A., ZAPIAIN-MERINO, S.J., SHIMADA, A., MORA, O. and VAZQUEZ-DUHALT, R., 2022. Chitosan nanoparticles containing lipoic acid with antioxidant properties as a potential nutritional supplement. Animals, vol. 12, no. 4, pp. 417. http://dx.doi.org/10.3390/ani12040417. PMid:35203125.
http://dx.doi.org/10.3390/ani12040417...
), chitosan-based nanoparticles comprising α-lipoic acid could pass the gastrointestinal barrier and emancipate their antioxidant consignment while remaining stable in stomach-like circumstances.

The incidence of metabolic disorders, a conglomeration of metabolic illnesses comprising obesity, insulin resistance, hyperglycemia, hypertension, and dyslipidemia, is estimated to be 25% worldwide (Eckel et al., 2005ECKEL, R.H., GRUNDY, S.M. and ZIMMET, P.Z., 2005. The metabolic syndrome. Lancet, vol. 365, no. 9468, pp. 1415-1428. http://dx.doi.org/10.1016/S0140-6736(05)66378-7. PMid:15836891.
http://dx.doi.org/10.1016/S0140-6736(05)...
; Saklayen , 2018SAKLAYEN, M.G., 2018. The global epidemic of the metabolic syndrome. Current Hypertension Reports, vol. 20, no. 2, pp. 12. http://dx.doi.org/10.1007/s11906-018-0812-z. PMid:29480368.
http://dx.doi.org/10.1007/s11906-018-081...
). Over time, many administrations, comprising the World Health Organization, the International Diabetes Federation, and the National Cholesterol Education Program Adult Treatment Panel III, have created varied clinical criteria for various metabolic syndromes (Huang, 2009HUANG, P.L., 2009. A comprehensive definition for metabolic syndrome. Disease Models & Mechanisms, vol. 2, no. 5-6, pp. 231-237. http://dx.doi.org/10.1242/dmm.001180. PMid:19407331.
http://dx.doi.org/10.1242/dmm.001180...
). Thus, the danger of metabolic disease for cardiac ailment and type 2 diabetes is becoming more widely acknowledged (Hudish et al., 2019HUDISH, L.I., REUSCH, J.E. and SUSSEL, L., 2019. β Cell dysfunction during progression of metabolic syndrome to type 2 diabetes. The Journal of Clinical Investigation, vol. 129, no. 10, pp. 4001-4008. http://dx.doi.org/10.1172/JCI129188. PMid:31424428.
http://dx.doi.org/10.1172/JCI129188...
). Pharmacological treatments and lifestyle changes are the predominant approaches for metabolic disorders (Saboya et al., 2017SABOYA, P.P., BODANESE, L.C., ZIMMERMANN, P.R., GUSTAVO, A.D.S., MACAGNAN, F.E., FEOLI, A.P. and OLIVEIRA, M.D.S., 2017. Lifestyle intervention on metabolic syndrome and its impact on quality of life: a randomized controlled trial. Arquivos Brasileiros de Cardiologia, vol. 108, no. 1, pp. 60-69. http://dx.doi.org/10.5935/abc.20160186. PMid:27982160.
http://dx.doi.org/10.5935/abc.20160186...
; Larsen et al., 2018LARSEN, J.R., DIMA, L., CORRELL, C.U. and MANU, P., 2018. The pharmacological management of metabolic syndrome. Expert Review of Clinical Pharmacology, vol. 11, no. 4, pp. 397-410. http://dx.doi.org/10.1080/17512433.2018.1429910. PMid:29345505.
http://dx.doi.org/10.1080/17512433.2018....
). Drugs are frequently employed since, in many situations, changing one's lifestyle may not yield the optimum results. Therefore, these medications have various adverse effects, including myalgia, hypoglycemia, and gastrointestinal discomfort. Their restricted use is also due to their high price (Ramkumar et al., 2016RAMKUMAR, S., RAGHUNATH, A. and RAGHUNATH, S., 2016. Statin therapy: review of safety and potential side effects. Acta Cardiologica Sinica, vol. 32, no. 6, pp. 631-639. http://dx.doi.org/10.6515/ACS20160611A. PMid:27899849.
http://dx.doi.org/10.6515/ACS20160611A...
; Wang and Hoyte, 2019WANG, G.S. and HOYTE, C., 2019. Review of biguanide (metformin) toxicity. Journal of Intensive Care Medicine, vol. 34, no. 11-12, pp. 863-876. http://dx.doi.org/10.1177/0885066618793385. PMid:30126348.
http://dx.doi.org/10.1177/08850666187933...
). Therefore, the search for natural remedies to lower the jeopardy and development of metabolic disorders has gained more and more attention. The antibacterial, immunostimulatory, hypoglycemic, anti-inflammatory, anti-obesity, hypolipidemic, anti-oxidative, and anti-hypertensive abilities of chitosan and its nanoformulations have been the substance of numerous research (Naveed et al., 2019NAVEED, M., PHIL, L., SOHAIL, M., HASNAT, M., BAIG, M.M.F.A., IHSAN, A.U., SHUMZAID, M., KAKAR, M.U., KHAN, T.M., AKABAR, M. HUSSAIN, M.I. and ZHOU Q.-G., 2019. Chitosan oligosaccharide (COS): an overview. International Journal of Biological Macromolecules, vol. 129, pp. 827-843. http://dx.doi.org/10.1016/j.ijbiomac.2019.01.192. PMid:30708011.
http://dx.doi.org/10.1016/j.ijbiomac.201...
; Khalaf et al., 2023KHALAF, E.M., ABOOD, N.A., ATTA, R.Z., RAMÍREZ-CORONEL, A.A., ALAZRAGI, R., PARRA, R.M.R., ABED, O.H., ABOSAOODA, M., JALIL, A.T., MUSTAFA, Y.F., NARMANI, A. and FARHOOD, B., 2023. Recent progressions in biomedical and pharmaceutical applications of chitosan nanoparticles: a comprehensive review. International Journal of Biological Macromolecules, vol. 231, pp. 123354. http://dx.doi.org/10.1016/j.ijbiomac.2023.123354. PMid:36681228.
http://dx.doi.org/10.1016/j.ijbiomac.202...
). Chitosan and its nano formulations may be a potential natural product to prevent delicacy metabolic disorders.

According to Herdiana et al. (2023)HERDIANA, Y., HUSNI, P., NURHASANAH, S., SHAMSUDDIN, S. and WATHONI, N., 2023. Chitosan-based nano systems for natural antioxidants in breast cancer therapy. Polymers, vol. 15, no. 13, pp. 2953. http://dx.doi.org/10.3390/polym15132953. PMid:37447598.
http://dx.doi.org/10.3390/polym15132953...
, one of the primary causes of cancer worldwide, breast cancer, frequently results in the creation of reactive oxygen species (ROS) and oxidative stress. This emphasizes the need for antioxidants to maintain the immune system and cell health. Natural antioxidants are crucial in lowering oxidative stress and restoring the body's internal environment equilibrium. Due to their weak solubility, 80 percent have limited effectiveness when taken orally. Increasing solubility in water is one tactic. Systems of chitosan-based nanoparticles are investigated because of their consistency and ease of manufacture. They can serve as a paradigm for developing natural antioxidant oral dosage forms that are effective and enhance the efficacy of cancer drugs. Cancer is one of the top causes of death, according to ALaqeel (2024)ALAQEEL, N.K., 2024. Antioxidants from different citrus peels provide protection against cancer. Brazilian Journal of Biology = Revista Brasileira de Biologia, vol. 84, e271619. http://dx.doi.org/10.1590/1519-6984.271619. PMid:37436265.
http://dx.doi.org/10.1590/1519-6984.2716...
, but patients are not always fit for current therapies, and they frequently have negative effects. The field of functional foods has witnessed a surge in the creation of natural anti-cancer medications, as several molecules have demonstrated both effectiveness and low toxicity. Due to their high flavonoid content, citrus peels may be able to prevent cancer. These antioxidants enhance apoptosis, prevent metastatic chain reactions, reduce the motility of cancer cells, and inhibit vasculature.

Body mass index (BMI) values are markedly elevated in obese individuals, although a slightly elevated BMI can boost overall survival and therapeutic responses (Berger, 2014BERGER, N.A., 2014. Obesity and cancer pathogenesis. Annals of the New York Academy of Sciences, vol. 1311, no. 1, pp. 57-76. http://dx.doi.org/10.1111/nyas.12416. PMid:24725147.
http://dx.doi.org/10.1111/nyas.12416...
). The “obesity paradox” states that when a person's BMI reaches the threshold of morbid obesity, the preventive benefits of a moderately elevated BMI disappear. It is still unclear how a person's degree of morbid obesity affects how their body reacts to cancer treatment (Lennon et al., 2016LENNON, H., SPERRIN, M., BADRICK, E. and RENEHAN, A.G., 2016. The obesity paradox in cancer: a review. Current Oncology Reports, vol. 18, no. 9, pp. 56. http://dx.doi.org/10.1007/s11912-016-0539-4. PMid:27475805.
http://dx.doi.org/10.1007/s11912-016-053...
). The increased bulk of adipose tissue could be a sign of energy reserves that help certain patients withstand the damaging effects of chemotherapy for a longer period. Longer lifetimes could be made possible by increased adipose storage, which could act as an energy reserve (Sánchez-Jiménez et al., 2019SÁNCHEZ-JIMÉNEZ, F., PÉREZ-PÉREZ, A., DE LA CRUZ-MERINO, L. and SÁNCHEZ-MARGALET, V., 2019. Obesity and breast cancer: role of leptin. Frontiers in Oncology, vol. 9, pp. 596. http://dx.doi.org/10.3389/fonc.2019.00596. PMid:31380268.
http://dx.doi.org/10.3389/fonc.2019.0059...
).

In this criticism, we will discuss the indication for the favorable abilities of chitosan and its nanoformulations on obesity, hyperglycemia, diabetes mellitus, dyslipidemia, and hypertension based on the in vivo and in vitro tests. We will also discuss the promising mechanisms of chitosan and its nanoformulations' avoidance and management of the metabolic syndrome.

2. Chitosan Structure

Chitosan is a polysaccharide molecule derivative from the chitin compound. It’s a common naturalistic polymer present in the cell walls of fungi and many exoskeletons such as arthropods, shellfish, and some insects (Nwe et al., 2009NWE, N., FURUIKE, T. and TAMURA, H., 2009. The mechanical and biological properties of chitosan scaffolds for tissue regeneration templates are significantly enhanced by chitosan from Gongronella butleri. Materials, vol. 2, no. 2, pp. 374-398. http://dx.doi.org/10.3390/ma2020374.
http://dx.doi.org/10.3390/ma2020374...
; Ahsan et al., 2018AHSAN, S.M., THOMAS, M., REDDY, K.K., SOORAPARAJU, S.G., ASTHANA, A. and BHATNAGAR, I., 2018. Chitosan as biomaterial in drug delivery and tissue engineering. International Journal of Biological Macromolecules, vol. 110, pp. 97-109. http://dx.doi.org/10.1016/j.ijbiomac.2017.08.140. PMid:28866015.
http://dx.doi.org/10.1016/j.ijbiomac.201...
). Different sources of chitosan and chitin are depicted in Figure 1. The unique structure of its free amino group, chitosan, is more capable of chemical changes and has better water solubility and hydrophilicity. Chitosan is manufactured by handling chitin with a determined NaOH solution, which fulfills N-deacetylation (Dutta et al., 2004DUTTA, P.K., DUTTA, J. and TRIPATHI, V.S., 2004 [viewed 15 November 2023]. Chitin and chitosan: chemistry, properties and applications. Journal of Scientific & Industrial Research [online], vol. 63, pp. 20-31. Available from: http://nopr.niscpr.res.in/handle/123456789/5397
http://nopr.niscpr.res.in/handle/1234567...
; Negm et al., 2020NEGM, N.A., HEFNI, H.H., ABD-ELAAL, A.A., BADR, E.A. and ABOU KANA, M.T., 2020. Advancement on modification of chitosan biopolymer and its potential applications. International Journal of Biological Macromolecules, vol. 152, pp. 681-702. http://dx.doi.org/10.1016/j.ijbiomac.2020.02.196. PMid:32084486.
http://dx.doi.org/10.1016/j.ijbiomac.202...
).

Figure 1
Different sources of chitosan and chitin.

Because it permits unique biological roles and the utilization of alteration responses, the attendance of amine clusters in chitosan and chitin is a significant benefit (Kumar, 2000KUMAR, M.N.V.R., 2000. A review of chitin and chitosan applications. Reactive & Functional Polymers, vol. 46, no. 1, pp. 1-27. http://dx.doi.org/10.1016/S1381-5148(00)00038-9.
http://dx.doi.org/10.1016/S1381-5148(00)...
). These polysaccharides' exceptional qualities, including their biocompatibility, non-toxicity, biodegradability, bioresorptivity, bioactivity, and worthy adsorption actions, make them ideal biomaterials and attract a lot of industrial interest as potential substitutes for synthetic polymers (Tan et al., 2009TAN, H., CHU, C.R., PAYNE, K.A. and MARRA, K.G., 2009. Injectable in situ forming biodegradable chitosan-hyaluronic acid-based hydrogels for cartilage tissue engineering. Biomaterials, vol. 30, no. 13, pp. 2499-2506. http://dx.doi.org/10.1016/j.biomaterials.2008.12.080. PMid:19167750.
http://dx.doi.org/10.1016/j.biomaterials...
; Croisier and Jérôme, 2013CROISIER, F. and JÉRÔME, C., 2013. Chitosan-based biomaterials for tissue engineering. European Polymer Journal, vol. 49, no. 4, pp. 780-792. http://dx.doi.org/10.1016/j.eurpolymj.2012.12.009.
http://dx.doi.org/10.1016/j.eurpolymj.20...
). The physicochemical possessions of chitosan are restricted by the molecular weight (MW) and further the N-deacetylation proportion. The more frequently employed expression is DA, which characterizes the percentage of N-acetylgluchitosanamine monomers to the whole numeral of polymer elements. As recounted by (Benediktsdóttir et al., 2014BENEDIKTSDÓTTIR, B.E., BALDURSSON, Ó. and MÁSSON, M., 2014. Challenges in evaluation of chitosan and trimethylated chitosan (TMC) as mucosal permeation enhancers: from synthesis to in vitro application. Journal of Controlled Release, vol. 173, pp. 18-31. http://dx.doi.org/10.1016/j.jconrel.2013.10.022. PMid:24511609.
http://dx.doi.org/10.1016/j.jconrel.2013...
), the D-A of a polymer such as chitosan can differ, but it is frequently arranged more than <50%

3. Chitosan Derivatives

The biological and physicochemical possessions of chitosan can be improved chemically. Chitosan has limited water solubility, and another prevalent organic solvent is one of its principal problems; however, this can be fixed chemically. Additionally, adding additional moieties to the polymer chain can enhance chitosan's other features, increasing its employment for medicinal requests (Negm et al., 2020NEGM, N.A., HEFNI, H.H., ABD-ELAAL, A.A., BADR, E.A. and ABOU KANA, M.T., 2020. Advancement on modification of chitosan biopolymer and its potential applications. International Journal of Biological Macromolecules, vol. 152, pp. 681-702. http://dx.doi.org/10.1016/j.ijbiomac.2020.02.196. PMid:32084486.
http://dx.doi.org/10.1016/j.ijbiomac.202...
). Chitosan derivatives can be produced using diverse synthetic approaches comprising direct modification, enzymatic processes, and chemical grafting (Nagy, 2018NAGY, V., 2018 [viewed 15 November 2023]. Chitosan-natural antioxidant conjugates: synthesis, antimicrobial and antioxidant properties [online]. Reykjavík: University of Iceland. Doctoral dissertation. Available from: http://hdl.handle.net/1946/32021
http://hdl.handle.net/1946/32021...
). The focal worry is that some functional clusters might obstruct the reaction and produce undesirable byproducts. So, they use shielding groups that briefly cover the functional clusters that would otherwise conflict (Carey and Sundberg, 2007CAREY, F.A. and SUNDBERG, R.J., 2007. Advanced organic chemistry: part B: reactions and synthesis. Boston: Springer, vol. 3.).

Wuts and Greene (2006)WUTS, P.G. and GREENE, T.W., 2006. Greene’s protective groups in organic synthesis. New York: John Wiley & Sons. http://dx.doi.org/10.1002/0470053488.
http://dx.doi.org/10.1002/0470053488...
reported that the molecule can add protective groups and withdraw without affecting the result. To provide a stable and sheltered substrate, a worthy defensive group must, among other things, react selectively with the required efficient cluster to produce an invention with a satisfactory yield. Chitosan has three nucleophilic functional clusters—the main\\O. H. group at the C-6 position, a secondary\\O. H. group at the C-3 position and an -NH2 group at the C-2 position.

Applying protecting clusters on the extremely reactive hydroxyl moieties is sought to create novel moieties on the amino responsibility of chitosan abilities. Since organic chemical reactions frequently involve hydroxyl protecting groups, there are many hydroxyls protective techniques accessible. The secondary - O.H. group of chitosan cannot be protected because the triphenylmethyl group can only be added to the primary\\O. H. group. Three synthesis steps are also included in the process, which is done at 100°C (Benediktsdóttir et al., 2011BENEDIKTSDÓTTIR, B.E., GAWARE, V.S., RÚNARSSON, Ö.V., JÓNSDÓTTIR, S., JENSEN, K.J. and MÁSSON, M., 2011. Synthesis of N,N,N-trimethyl chitosan homopolymer and highly substituted N-alkyl-N,N-dimethyl chitosan derivatives with the aid of di-tert-butyldimethylsilyl chitosan. Carbohydrate Polymers, vol. 86, no. 4, pp. 1451-1460. http://dx.doi.org/10.1016/j.carbpol.2011.06.007.
http://dx.doi.org/10.1016/j.carbpol.2011...
).

Wuts and Greene (2006)WUTS, P.G. and GREENE, T.W., 2006. Greene’s protective groups in organic synthesis. New York: John Wiley & Sons. http://dx.doi.org/10.1002/0470053488.
http://dx.doi.org/10.1002/0470053488...
indicated that TMS (trimethylsilyl) and TBDMS (tert-butyldimethylsilyl) ethers of silyl are simply formed from hydroxyl groups. In contrast, TBDMS ethers are stable and satirically hindered protective groups that show promise. Using TBDMS protection groups, (Kurita et al., 2002KURITA, K., IKEDA, H., YOSHIDA, Y., SHIMOJOH, M. and HARATA, M., 2002. Chemoselective protection of the amino groups of chitosan by controlled phthaloylation: facile preparation of a precursor useful for chemical modifications. Biomacromolecules, vol. 3, no. 1, pp. 1-4. http://dx.doi.org/10.1021/bm0101163. PMid:11866549.
http://dx.doi.org/10.1021/bm0101163...
; Nagy, 2018NAGY, V., 2018 [viewed 15 November 2023]. Chitosan-natural antioxidant conjugates: synthesis, antimicrobial and antioxidant properties [online]. Reykjavík: University of Iceland. Doctoral dissertation. Available from: http://hdl.handle.net/1946/32021
http://hdl.handle.net/1946/32021...
) created an artificial method to shield the hydroxyl clusters of chitosan. To create a completely 3,6-O-TBDMS protected chitosan, the TBDMS moiety is added to the mesylate salt of chitosan in one phase of the technique. Applying protecting groups to the amino group of chitosan is anticipated to create novel moieties on the hydroxyl responsibility of the substance where chitosan is created when polyamines precipitate in alkaline liquids. Even though it offers medicinal qualities like ulcer-fighting (Fini and Orienti ,2003FINI, A. and ORIENTI, I., 2003. The role of chitosan in drug delivery: current and potential applications. American Journal of Drug Delivery, vol. 1, no. 1, pp. 43-59. http://dx.doi.org/10.2165/00137696-200301010-00004.
http://dx.doi.org/10.2165/00137696-20030...
), wound-healing (Azad et al., 2004AZAD, A.K., SERMSINTHAM, N., CHANDRKRACHANG, S. and STEVENS, W.F., 2004. Chitosan membrane as a wound‐healing dressing: characterization and clinical application. Journal of Biomedical Materials Research Part B: Applied Biomaterials, vol. 69B, no. 2, pp. 216-222. http://dx.doi.org/10.1002/jbm.b.30000. PMid:15116411.
http://dx.doi.org/10.1002/jbm.b.30000...
), and antibacterial capabilities as well as the capacity to lower cholesterol (Sugano et al., 1988SUGANO, M., WATANABE, S., KISHI, A., IZUME, M. and OHTAKARA, A., 1988. Hypocholesterolemic action of chitosans with different viscosity in rats. Lipids, vol. 23, no. 3, pp. 187-191. http://dx.doi.org/10.1007/BF02535456. PMid:2836688.
http://dx.doi.org/10.1007/BF02535456...
). Chitosan's R-NH3 + group, which are cationic, have mucoadhesive properties when it interacts with the negatively charged groups on mucosal surfaces (Kockisch et al., 2003KOCKISCH, S., REES, G.D., YOUNG, S.A., TSIBOUKLIS, J. and SMART, J.D., 2003. Polymeric microspheres for drug delivery to the oral cavity: an in vitro evaluation of mucoadhesive potential. Journal of Pharmaceutical Sciences, vol. 92, no. 8, pp. 1614-1623. http://dx.doi.org/10.1002/jps.10423. PMid:12884248.
http://dx.doi.org/10.1002/jps.10423...
). Also, protein-associated tight junctions endure revocable structural remodeling in response to interactions with protonated amine groups, which are followed by tight junction openings. The simplicity structure of chitosan may be chemically altered, notably in the C-2 position, causing derivatives with numerous possessions and possible uses, which is another feature that sets chitosan apart from other polysaccharide polymers (Huo et al., 2010HUO, M., ZHANG, Y., ZHOU, J., ZOU, A., YU, D., WU, Y., LI, J. and LI, H., 2010. Synthesis and characterization of low-toxic amphiphilic chitosan derivatives and their application as micelle carrier for antitumor drug. International Journal of Pharmaceutics, vol. 394, no. 1-2, pp. 162-173. http://dx.doi.org/10.1016/j.ijpharm.2010.05.001. PMid:20457237.
http://dx.doi.org/10.1016/j.ijpharm.2010...
). Likewise, Bashir et al. (2022)BASHIR, S.M., AHMED RATHER, G., PATRÍCIO, A., HAQ, Z., SHEIKH, A.A., SHAH, M.Z.U.H., SINGH, H., KHAN, A.A., IMTIYAZ, S., AHMAD, S.B., NABI, S., RAKHSHAN, R., HASSAN, S. and FONTE, P., 2022. Chitosan nanoparticles: a versatile platform for biomedical applications. Materials, vol. 15, no. 19, pp. 6521. http://dx.doi.org/10.3390/ma15196521. PMid:36233864.
http://dx.doi.org/10.3390/ma15196521...
showed that owing to their exquisite biological characteristics, extensibility, and efficient consumption by intranasal mucosal cells to tumor cells.

Furthermore, chitosan and its nano derivatives could participate significantly in metabolic syndromes drug delivery. Developing anticancer medications, catalysis, gene delivery, sensor requests, packaging and wrapping supplies, cosmetic fabrics, and bioimaging is also progressing industrially for chitosan and its derivatives. The different possessions of chitosan and its derivatives make it a brilliant biomolecule for countless biomedical uses as revealed in Figure 2.

Figure 2
Biomedical uses of chitosan and its derivatives.

4. The Production of Chitosan Nanoparticles

Chitosan nanoparticles were initially stated in 1994 when (Ohya et al., 1994OHYA, Y., SHIRATANI, M., KOBAYASHI, H. and OUCHI, T., 1994. Release behavior of 5-fluorouracil from chitosan-gel nanospheres immobilizing 5-fluorouracil coated with polysaccharides and their cell specific cytotoxicity. Journal of Macromolecular Science, Part A: Pure and Applied Chemistry, vol. 31, no. 5, pp. 629-642. http://dx.doi.org/10.1080/10601329409349743.
http://dx.doi.org/10.1080/10601329409349...
) anticipated consuming them to deliver the anticancer prescription 5-fluorouracil intravenously. These synthesized nanoparticles were formed by cross-linking and emulsifying chitosan. Meanwhile, these schemes have experienced considerable exploration for drug delivery tenacities. The innovative formularization has either been modified by utilizing various preparation procedures or for other uses, such as integrating active ingredients in toothpaste (Calvo et al., 1997CALVO, P., REMUÑÁN‐LÓPEZ, C., VILA‐JATO, J.L. and ALONSO, M.J., 1997. Novel hydrophilic chitosan‐polyethylene oxide nanoparticles as protein carriers. Journal of Applied Polymer Science, vol. 63, no. 1, pp. 125-132. http://dx.doi.org/10.1002/(SICI)1097-4628(19970103)63:1<125::AID-APP13>3.0.CO;2-4.
http://dx.doi.org/10.1002/(SICI)1097-462...
; Erbacher et al., 1998ERBACHER, P., ZOU, S., BETTINGER, T., STEFFAN, A.M. and REMY, J.S., 1998. Chitosan-based vector/DNA complexes for gene delivery: biophysical characteristics and transfection ability. Pharmaceutical Research, vol. 15, no. 9, pp. 1332-1339. http://dx.doi.org/10.1023/A:1011981000671. PMid:9755882.
http://dx.doi.org/10.1023/A:101198100067...
; El-Shabouri, 2002EL-SHABOURI, M.H., 2002. Positively charged nanoparticles for improving the oral bioavailability of cyclosporin-A. International Journal of Pharmaceutics, vol. 249, no. 1-2, pp. 101-108. http://dx.doi.org/10.1016/S0378-5173(02)00461-1. PMid:12433438.
http://dx.doi.org/10.1016/S0378-5173(02)...
; Liu et al., 2007aLIU, B., LIU, W.S., HAN, B.Q. and SUN, Y.Y., 2007a. Antidiabetic effects of chitooligosaccharides on pancreatic islet cells in streptozotocin-induced diabetic rats. World Journal of Gastroenterology, vol. 13, no. 5, pp. 725-731. http://dx.doi.org/10.3748/wjg.v13.i5.725. PMid:17278195.
http://dx.doi.org/10.3748/wjg.v13.i5.725...
). Furthermore, numerous teams have formed fresh chitosan nanoparticle inventions with accompanying matrix-shaping machinery (Sarmento et al., 2006SARMENTO, B., MARTINS, S., RIBEIRO, A., VEIGA, F., NEUFELD, R. and FERREIRA, D., 2006. Development and comparison of different nanoparticulate polyelectrolyte complexes as insulin carriers. International Journal of Peptide Research and Therapeutics, vol. 12, no. 2, pp. 131-138. http://dx.doi.org/10.1007/s10989-005-9010-3.
http://dx.doi.org/10.1007/s10989-005-901...
; Grenha et al., 2010GRENHA, A., GOMES, M.E., RODRIGUES, M., SANTO, V.E., MANO, J.F., NEVES, N.M. and REIS, R.L., 2010. Development of new chitosan/carrageenan nanoparticles for drug delivery applications. Journal of Biomedical Materials Research Part A, vol. 92A, no. 4, pp. 1265-1272. http://dx.doi.org/10.1002/jbm.a.32466. PMid:19322874.
http://dx.doi.org/10.1002/jbm.a.32466...
).

Frequent approaches have been generated, primarily connecting emulsification, numerous coacervations, or even tiny variations. More specifically, the devices comprise desolvation (Tian and Groves, 1999TIAN, X.-X. and GROVES, M.J., 1999. Formulation and biological activity of antineoplastic proteoglycans derived from Mycobacterium vaccae in chitosan nanoparticles. The Journal of Pharmacy and Pharmacology, vol. 51, no. 2, pp. 151-157. http://dx.doi.org/10.1211/0022357991772268. PMid:10217313.
http://dx.doi.org/10.1211/00223579917722...
), reverse micellar method (Orellano et al., 2017ORELLANO, M.S., PORPORATTO, C., SILBER, J.J., FALCONE, R.D. and CORREA, N.M., 2017. AOT reverse micelles as versatile reaction media for chitosan nanoparticles synthesis. Carbohydrate Polymers, vol. 171, pp. 85-93. http://dx.doi.org/10.1016/j.carbpol.2017.04.074. PMid:28578974.
http://dx.doi.org/10.1016/j.carbpol.2017...
), ionic gelation, polyelectrolyte complexation (Sarmento et al., 2006SARMENTO, B., MARTINS, S., RIBEIRO, A., VEIGA, F., NEUFELD, R. and FERREIRA, D., 2006. Development and comparison of different nanoparticulate polyelectrolyte complexes as insulin carriers. International Journal of Peptide Research and Therapeutics, vol. 12, no. 2, pp. 131-138. http://dx.doi.org/10.1007/s10989-005-9010-3.
http://dx.doi.org/10.1007/s10989-005-901...
), emulsion solvent diffusion (El-Shabouri, 2002EL-SHABOURI, M.H., 2002. Positively charged nanoparticles for improving the oral bioavailability of cyclosporin-A. International Journal of Pharmaceutics, vol. 249, no. 1-2, pp. 101-108. http://dx.doi.org/10.1016/S0378-5173(02)00461-1. PMid:12433438.
http://dx.doi.org/10.1016/S0378-5173(02)...
), emulsion droplet coalescence (Tokumitsu et al., 1999TOKUMITSU, H., ICHIKAWA, H., FUKUMORI, Y. and BLOCK, L.H., 1999. Preparation of gadopentetic acid-loaded chitosan microparticles for gadolinium neutron-capture therapy of cancer by a novel emulsion-droplet coalescence technique. Chemical & Pharmaceutical Bulletin, vol. 47, no. 6, pp. 838-842. http://dx.doi.org/10.1248/cpb.47.838. PMid:10399838.
http://dx.doi.org/10.1248/cpb.47.838...
), and All of these techniques fall under the category of bottom-up manufacture manners, which entail the gathering of compounds in solution to create specific structures, in this instance, nanoparticles (Chan and Kwok, 2011CHAN, H.K. and KWOK, P.C.L., 2011. Production methods for nanodrug particles using the bottom-up approach. Advanced Drug Delivery Reviews, vol. 63, no. 6, pp. 406-416. http://dx.doi.org/10.1016/j.addr.2011.03.011. PMid:21457742.
http://dx.doi.org/10.1016/j.addr.2011.03...
).

Bottom-up technologies frequently exhibit size polydispersity in their delivery systems, which might occasionally limit the effectiveness of nanoparticles (Wang et al., 2011WANG, J., BYRNE, J.D., NAPIER, M.E. and DESIMONE, J.M., 2011. More effective nanomedicines through particle design. Small, vol. 7, no. 14, pp. 1919-1931. http://dx.doi.org/10.1002/smll.201100442. PMid:21695781.
http://dx.doi.org/10.1002/smll.201100442...
). Chitosan, or one of its derivatives, is employed to make chitosan NPs. Because of chitosan's special non-toxicity, polymeric cationic nature, biodegradability, mucoadhesive chitosan, great biocompatibility, and absorption-enhancing properties, the N-deacetylated derivative of chitin is a desirable biopolymer for making nanoparticles (Kunjachan et al., 2010KUNJACHAN, S., JOSE, S. and LAMMERS, T., 2010. Understanding the mechanism of ionic gelation for synthesis of chitosan nanoparticles using qualitative techniques. Asian Journal of Pharmaceutics, vol. 4, no. 2, pp. 148. http://dx.doi.org/10.4103/0973-8398.68467.
http://dx.doi.org/10.4103/0973-8398.6846...
). Chitosan is advantageous in creating nanoparticles due to its cationic character, which permits ionic cross-related with multivalent anions (Agnihotri et al., 2004AGNIHOTRI, S.A., MALLIKARJUNA, N.N. and AMINABHAVI, T.M., 2004. Recent advances on chitosan-based micro-and nanoparticles in drug delivery. Journal of Controlled Release, vol. 100, no. 1, pp. 5-28. http://dx.doi.org/10.1016/j.jconrel.2004.08.010. PMid:15491807.
http://dx.doi.org/10.1016/j.jconrel.2004...
), and its linear polyamine structure, which has a diversity of free amine clusters that are reachable for cross-linking.

Chitosan NPs have distinct properties that enable in vivo site-specific targeting and increased affinity for negatively charged biological membranes (Qi et al., 2004QI, L., XU, Z., JIANG, X., HU, C. and ZOU, X., 2004. Preparation and antibacterial activity of chitosan nanoparticles. Carbohydrate Research, vol. 339, no. 16, pp. 2693-2700. http://dx.doi.org/10.1016/j.carres.2004.09.007. PMid:15519328.
http://dx.doi.org/10.1016/j.carres.2004....
). As a result, they can be employed for an assortment of requests in different industries to load medicines efficiently, enzymes, and nucleic acids (Colonna et al., 2007COLONNA, C., CONTI, B., PERUGINI, P., PAVANETTO, F., MODENA, T., DORATI, R. and GENTA, I., 2007. Chitosan glutamate nanoparticles for protein delivery: development and effect on prolidase stability. Journal of Microencapsulation, vol. 24, no. 6, pp. 553-564. http://dx.doi.org/10.1080/02652040701449608. PMid:17654175.
http://dx.doi.org/10.1080/02652040701449...
) using a controlled release (Corradini et al., 2010CORRADINI, E., MOURA, M. and MATTOSO, L., 2010. A preliminary study of the incorparation of NPK fertilizer into chitosan nanoparticles. Express Polymer Letters, vol. 4, no. 8, pp. 509-515. http://dx.doi.org/10.3144/expresspolymlett.2010.64.
http://dx.doi.org/10.3144/expresspolymle...
). Because of the characteristics of the material and the manufacturing process, chitosan nanoparticles exhibit excellent chemical, morphological, and physical capabilities. Chitosan is soluble in acidic solutions like citric, tartaric, and acetic acids but insoluble in water (Furuike et al., 2017FURUIKE, T., KOMOTO, D., HASHIMOTO, H. and TAMURA, H., 2017. Preparation of chitosan hydrogel and its solubility in organic acids. International Journal of Biological Macromolecules, vol. 104, no. Pt B, pp. 1620-1625. http://dx.doi.org/10.1016/j.ijbiomac.2017.02.099. PMid:28258006.
http://dx.doi.org/10.1016/j.ijbiomac.201...
). It comes in low- and high-molecular-weight varieties with weights stretching from 3800 to 20,000 Da. Chitosan's characteristics are substantially prejudiced by its molecular weight and level of deacetylation, especially when it comes to the creation of nanoparticles. Chitosan-based polymeric drug carriers, growth factors, anticancer medications, anti-inflammatories, antimicrobials, peptides, and other therapeutics have all been efficaciously administered (Sun et al., 2007SUN, T., ZHOU, D., XIE, J. and MAO, F., 2007. Preparation of chitosan oligomers and their antioxidant activity. European Food Research and Technology, vol. 225, no. 3-4, pp. 451-456. http://dx.doi.org/10.1007/s00217-006-0439-1.
http://dx.doi.org/10.1007/s00217-006-043...
).

Othman et al. (2018)OTHMAN, N., MASARUDIN, M.J., KUEN, C.Y., DASUAN, N.A., ABDULLAH, L.C. and JAMIL, S.N.A.M., 2018. Synthesis and optimization of chitosan nanoparticles loaded with L-ascorbic acid and thymoquinone. Nanomaterials, vol. 8, no. 11, pp. 920. http://dx.doi.org/10.3390/nano8110920. PMid:30405074.
http://dx.doi.org/10.3390/nano8110920...
indicated that the hydrophilic L-ascorbic acid and hydrophobic thymoquinone, a myriad of greatly effective multifactorial with inferior systemic intake, could be encapsulated collected in chitosan NPs schemes to escalate their therapeutic competence by indirectly participating to the improvement of pharmaceutical and medical areas. Also, pharmaceuticals can be delivered orally, transdermal, or intravenously using NPs as carriers. According to studies, chitosan NPs have been extensively employed in the medical and biological fields to remedy conditions like cancer (Nayak et al., 2016NAYAK, D., MINZ, A.P., ASHE, S., RAUTA, P.R., KUMARI, M., CHOPRA, P. and NAYAK, B., 2016. Synergistic combination of antioxidants, silver nanoparticles and chitosan in a nanoparticle based formulation: characterization and cytotoxic effect on MCF-7 breast cancer cell lines. Journal of Colloid and Interface Science, vol. 470, pp. 142-152. http://dx.doi.org/10.1016/j.jcis.2016.02.043. PMid:26939078.
http://dx.doi.org/10.1016/j.jcis.2016.02...
) and diabetes (Wong et al., 2017WONG, C.Y., AL-SALAMI, H. and DASS, C.R., 2017. Potential of insulin nanoparticle formulations for oral delivery and diabetes treatment. Journal of Controlled Release, vol. 264, pp. 247-275. http://dx.doi.org/10.1016/j.jconrel.2017.09.003. PMid:28887133.
http://dx.doi.org/10.1016/j.jconrel.2017...
).

5. Metabolic Syndromes

With modernization and globalization, people's lifestyles have been meaningfully rehabilitated, comprising less leisure and more employed hours. Furthermore, the persistent usage of electronic gadgets has prepared the lifestyle softly and increased the generation of diseases. One of the prevalent ailments is metabolic syndrome (Azad et al., 2004AZAD, A.K., SERMSINTHAM, N., CHANDRKRACHANG, S. and STEVENS, W.F., 2004. Chitosan membrane as a wound‐healing dressing: characterization and clinical application. Journal of Biomedical Materials Research Part B: Applied Biomaterials, vol. 69B, no. 2, pp. 216-222. http://dx.doi.org/10.1002/jbm.b.30000. PMid:15116411.
http://dx.doi.org/10.1002/jbm.b.30000...
), which is a cluster of pathologies such as insulin resistance, obesity, dyslipidemia, hyperglycemia, and hypertension, that make susceptible to cardiovascular diseases (Nakhaei et al., 2019NAKHAEI, H., MOGHARNASI, M. and FANAEI, H., 2019. Effect of swimming training on levels of asprosin, lipid profile, glucose and insulin resistance in rats with metabolic syndrome. Obesity Medicine, vol. 15, pp. 100111. http://dx.doi.org/10.1016/j.obmed.2019.100111.
http://dx.doi.org/10.1016/j.obmed.2019.1...
; Rossi et al., 2022ROSSI, J.L.S., BARBALHO, S.M., ARAUJO, R.R., BECHARA, M.D., SLOAN, K.P. and SLOAN, L.A., 2022. Metabolic syndrome and cardiovascular diseases: going beyond traditional risk factors. Diabetes Metabolism Research and Reviews, vol. 38, no. 3, e3502. http://dx.doi.org/10.1002/dmrr.3502. PMid:34614543.
http://dx.doi.org/10.1002/dmrr.3502...
). Even though prevailing international clusters have assembled to elucidate a consent characterization of “metabolic syndrome,” the identical has not been fulfilled for describing “metabolic dysfunction,” a purport demonstrating disordered metabolism on a continuum rather than a definitive diagnosis. According to the pathophysiology reports of MS, augmented insulin resistance, plasma-free fatty acids, inflammation indices, and oxidative stress are the main underlying features of MS (Rossi et al., 2022ROSSI, J.L.S., BARBALHO, S.M., ARAUJO, R.R., BECHARA, M.D., SLOAN, K.P. and SLOAN, L.A., 2022. Metabolic syndrome and cardiovascular diseases: going beyond traditional risk factors. Diabetes Metabolism Research and Reviews, vol. 38, no. 3, e3502. http://dx.doi.org/10.1002/dmrr.3502. PMid:34614543.
http://dx.doi.org/10.1002/dmrr.3502...
). Insulin is an indispensable element for tissue uptake of glucose, deterring lipolysis and hepatic gluconeogenesis. Higher circulatory free fatty acids (FFAs) can conquer insulin consent, which is connected with insulin resistance in obese persons (Fahed et al., 2022FAHED, G., AOUN, L., BOU ZERDAN, M., ALLAM, S., BOU ZERDAN, M., BOUFERRAA, Y. and ASSI, H.I., 2022. Metabolic syndrome: updates on pathophysiology and management in 2021. International Journal of Molecular Sciences, vol. 23, no. 2, pp. 786. http://dx.doi.org/10.3390/ijms23020786. PMid:35054972.
http://dx.doi.org/10.3390/ijms23020786...
).

Moreover, protein kinase activity is repressed with circulating FFAs; this feature can lower muscle glucose consumed. Contrariwise, greater hepatic protein kinase levels boost the assembly of atherogenic ingredients, counting glucose, LDL, and TGs. Furthermore, the resultant hyperglycemia activates more insulin releasing, thus causing hyperinsulinemia. Oxidative stress is also involved with insulin resistance and can prevent adipocytes from producing adiponectin (Furukawa et al., 2004FURUKAWA, S., FUJITA, T., SHIMABUKURO, M., IWAKI, M., YAMADA, Y., NAKAJIMA, Y., NAKAYAMA, O., MAKISHIMA, M., MATSUDA, M. and SHIMOMURA, I., 2004. Increased oxidative stress in obesity and its impact on metabolic syndrome. The Journal of Clinical Investigation, vol. 114, no. 12, pp. 1752-1761. http://dx.doi.org/10.1172/JCI21625. PMid:15599400.
http://dx.doi.org/10.1172/JCI21625...
). The connection between belly fat in insulin resistance is considerable as lipolysis of belly fat leads to boosted circulation of FFAs to the hepatic, triggering the amplified synthesis of TGs and LDL (Nakhaei et al., 2019NAKHAEI, H., MOGHARNASI, M. and FANAEI, H., 2019. Effect of swimming training on levels of asprosin, lipid profile, glucose and insulin resistance in rats with metabolic syndrome. Obesity Medicine, vol. 15, pp. 100111. http://dx.doi.org/10.1016/j.obmed.2019.100111.
http://dx.doi.org/10.1016/j.obmed.2019.1...
). Besides, visceral adipose tissue triggers greater levels of plasminogen activator inhibitor, augmenting heparin-binding epidermal growth factor- and prothrombotic state that vascular modeling and encourages smooth muscle cells (Slate-Romano et al., 2022SLATE-ROMANO, J.J., YANO, N. and ZHAO, T.C., 2022. Irisin reduces inflammatory signaling pathways in inflammation-mediated metabolic syndrome. Molecular and Cellular Endocrinology, vol. 552, pp. 111676. http://dx.doi.org/10.1016/j.mce.2022.111676. PMid:35569582.
http://dx.doi.org/10.1016/j.mce.2022.111...
).

Previously, the association between MS and inflammation has been well documented through visceral obesity, which exaggerates insulin resistance. In this regard, the adipose tissue macrophages release TNF-α, which encourages the inactivation of insulin receptors in the thesis’s tissues, instigating lipolysis with the synthesis of FFAs, thus preventing the emancipating of adiponectin (Nakhaei et al., 2019NAKHAEI, H., MOGHARNASI, M. and FANAEI, H., 2019. Effect of swimming training on levels of asprosin, lipid profile, glucose and insulin resistance in rats with metabolic syndrome. Obesity Medicine, vol. 15, pp. 100111. http://dx.doi.org/10.1016/j.obmed.2019.100111.
http://dx.doi.org/10.1016/j.obmed.2019.1...
). Based on literature and clinical reports, there is a substantial connection between elevated amounts of TNF-α, obesity insulin and resistance (Wisse, 2004WISSE, B.E., 2004. The inflammatory syndrome: the role of adipose tissue cytokines in metabolic disorders linked to obesity. Journal of the American Society of Nephrology, vol. 15, no. 11, pp. 2792-2800. http://dx.doi.org/10.1097/01.ASN.0000141966.69934.21. PMid:15504932.
http://dx.doi.org/10.1097/01.ASN.0000141...
). The immune and adipocyte cells release IL-6 (interleukin-6), and its synthesis escalations with adipose tissue mass (Rocha et al., 2022ROCHA, A.R.F., MORAIS, N.S., PRIORE, S.E. and FRANCESCHINI, S.C.C., 2022. Inflammatory biomarkers and components of metabolic syndrome in adolescents: a systematic review. Inflammation, vol. 45, no. 1, pp. 14-30. http://dx.doi.org/10.1007/s10753-021-01549-1. PMid:34546513.
http://dx.doi.org/10.1007/s10753-021-015...
), which further encourages the hepatocytes to create C-reactive proteins, whose raised level has been involved with the etiology of MS (Devaraj et al., 2009DEVARAJ, S., SINGH, U. and JIALAL, I., 2009. Human C-reactive protein and the metabolic syndrome. Current Opinion in Lipidology, vol. 20, no. 3, pp. 182-189. http://dx.doi.org/10.1097/MOL.0b013e32832ac03e. PMid:19369869.
http://dx.doi.org/10.1097/MOL.0b013e3283...
). Moreover, it is also involved in stimulating RSA pathways (Wisse, 2004WISSE, B.E., 2004. The inflammatory syndrome: the role of adipose tissue cytokines in metabolic disorders linked to obesity. Journal of the American Society of Nephrology, vol. 15, no. 11, pp. 2792-2800. http://dx.doi.org/10.1097/01.ASN.0000141966.69934.21. PMid:15504932.
http://dx.doi.org/10.1097/01.ASN.0000141...
).

6. Impacts of Chitosan and Its Nanoformulations on Obesity and Dyslipidemia

Obesity is a persistently sustained metabolic condition described by an extreme buildup of body fat brought on by an unbalanced energy intake. Additionally, dyslipidemia describes unhealthily high levels of one or more lipid types in the blood, including higher levels of triglycerides and LDL (low-density lipoprotein) and reduced levels of HDL (high-density lipoprotein cholesterol), resulting from several abnormalities in structure, metabolism, antiatherogenic lipoproteins and biology of atherogenic (Srikanth and Deedwania, 2016SRIKANTH, S. and DEEDWANIA, P., 2016. Management of dyslipidemia in patients with hypertension, diabetes, and metabolic syndrome. Current Hypertension Reports, vol. 18, no. 10, pp. 76. http://dx.doi.org/10.1007/s11906-016-0683-0. PMid:27730495.
http://dx.doi.org/10.1007/s11906-016-068...
). Chitosan has been established in numerous kinds of research to have effective anti-obesity and hypolipidemic properties. Furthermore, it has been shown that chitosan successfully suppressed hypertrophy and adipocyte hyperplasia in HFD (high-fat diet)-stimulated obese rat models (Bai et al., 2018BAI, Y., ZHENG, J., YUAN, X., JIAO, S., FENG, C., DU, Y., LIU, H. and ZHENG, L., 2018. Chitosan oligosaccharides improve glucolipid metabolism disorder in liver by suppression of obesity-related inflammation and restoration of peroxisome proliferator-activated receptor gamma (PPARγ). Marine Drugs, vol. 16, no. 11, pp. 455. http://dx.doi.org/10.3390/md16110455. PMid:30463189.
http://dx.doi.org/10.3390/md16110455...
; Pan et al., 2018PAN, H., FU, C., HUANG, L., JIANG, Y., DENG, X., GUO, J. and SU, Z., 2018. Anti-obesity effect of chitosan oligosaccharide capsules (COSCs) in obese rats by ameliorating leptin resistance and adipogenesis. Marine Drugs, vol. 16, no. 6, pp. 198. http://dx.doi.org/10.3390/md16060198. PMid:29874843.
http://dx.doi.org/10.3390/md16060198...
; He et al., 2020HE, N., WANG, S., LV, Z., ZHAO, W. and LI, S., 2020. Low molecular weight chitosan oligosaccharides (LMW-COSs) prevent obesity-related metabolic abnormalities in association with the modification of gut microbiota in high-fat diet (HFD)-fed mice. Food & Function, vol. 11, no. 11, pp. 9947-9959. http://dx.doi.org/10.1039/D0FO01871F. PMid:33108433.
http://dx.doi.org/10.1039/D0FO01871F...
; Lee et al., 2021LEE, J.Y., KIM, T.Y., KANG, H., OH, J., PARK, J.W., KIM, S.C., KIM, M., APOSTOLIDIS, E., KIM, Y.C. and KWON, Y.I., 2021. Anti-obesity and anti-adipogenic effects of chitosan oligosaccharide (GO2KA1) in SD rats and in 3T3-L1 preadipocytes models. Molecules, vol. 26, no. 2, pp. 331. http://dx.doi.org/10.3390/molecules26020331. PMid:33440605.
http://dx.doi.org/10.3390/molecules26020...
). It also decreases body weight growth, hepatic fat gathering, blood lipid levels, hypertrophy, and adipocyte hyperplasia.

One of the major health issues associated with modern wealthy society is obesity. In 2015, 2 billion adults worldwide—or 38-40% of the global population—were classified as overweight or obese (GBD, 2017GLOBAL BURDEN OF DISEASE COLLABORATIVE NETWORK - GBD, 2017. Global Burden of Disease Study 2015 (GBD 2015) obesity and overweight prevalence 1980-2015. Seattle: Institute for Health Metrics and Evaluation.). Several persistent illnesses, including breast cancer (BC), have greater rates of death and morbidity when an individual is obese. The elderly population has a rising rate of obesity (Shekhar et al., 2021SHEKHAR, S., LIU, Y., WANG, S., ZHANG, H., FANG, X., ZHANG, J., FAN, L., ZHENG, B., ROMAN, R.J., WANG, Z., FAN, F. and BOOZ, G.W., 2021. Novel mechanistic insights and potential therapeutic impact of TRPC6 in neurovascular coupling and ischemic stroke. International Journal of Molecular Sciences, vol. 22, no. 4, pp. 2074. http://dx.doi.org/10.3390/ijms22042074. PMid:33669830.
http://dx.doi.org/10.3390/ijms22042074...
; Fang et al., 2022FANG, X., ZHANG, J., ROMAN, R.J. and FAN, F., 2022. From 1901 to 2022, how far are we from truly understanding the pathogenesis of age-related dementia? GeroScience, vol. 44, no. 3, pp. 1879-1883. http://dx.doi.org/10.1007/s11357-022-00591-7. PMid:35585301.
http://dx.doi.org/10.1007/s11357-022-005...
). According to Chen et al. (2022)CHEN, K., ZHANG, J., BEERAKA, N.M., TANG, C., BABAYEVA, Y.V., SINELNIKOV, M.Y., ZHANG, X., ZHANG, J., LIU, J., RESHETOV, I.V., SUKOCHEVA, O.A., LU, P. and FAN, R., 2022. Advances in the prevention and treatment of obesity-driven effects in breast cancers. Frontiers in Oncology, vol. 12, pp. 820968. http://dx.doi.org/10.3389/fonc.2022.820968. PMid:35814391.
http://dx.doi.org/10.3389/fonc.2022.8209...
, estrogen and inflammatory are associated with fat accumulation and hypertrophy, which may contribute to the occurrence of BC in postmenopausal women. Uncertainty surrounds the function of adipose tissue in cancer patients, though. Patients who are somewhat overweight fare better from treatment, illuminating the “obesity paradox.” Weight control and prevention programs should be added to current treatments, and a tailored medical strategy should consider adiposity reduction. Liu et al. (2023)LIU, Y., SUN, Y., GUO, Y., SHI, X., CHEN, X., FENG, W., WU, L.L., ZHANG, J., YU, S., WANG, Y. and SHI, Y., 2023. An overview: the diversified role of mitochondria in cancer metabolism. International Journal of Biological Sciences, vol. 19, no. 3, pp. 897-915. http://dx.doi.org/10.7150/ijbs.81609. PMid:36778129.
http://dx.doi.org/10.7150/ijbs.81609...
state that mitochondria are essential organelles for synthesizing energy, cell metabolism, and signaling. They also play a role in the growth and spread of tumors. Biology and synthesis in cancer cells can be enhanced by mutations in mtDNA and the tricarboxylic acid cycle (TCA) enzymes. Because mitochondria rely on glycolysis and oxidative phosphorylation for energy, they are the focus of cancer treatment. Targeting these pathways and metabolism may be a useful treatment approach for several malignancies.

The importance of mitochondria in carcinogenesis was highlighted by Kaelin Junior and McKnight (2013)KAELIN JUNIOR, W.G. and MCKNIGHT, S.L., 2013. Influence of metabolism on epigenetics and disease. Cell, vol. 153, no. 1, pp. 56-69. http://dx.doi.org/10.1016/j.cell.2013.03.004. PMid:23540690.
http://dx.doi.org/10.1016/j.cell.2013.03...
, who also discussed how their metabolites affect gene expression and cell signaling through epigenetic controls. The basic function of mitochondria is energy production. Furthermore, metabolites in the TCA have been shown to promote epigenetic alterations such as DNA methylation and post-translational modification of histones (Liu et al., 2022LIU, Y., CHEN, C., WANG, X., SUN, Y., ZHANG, J., CHEN, J. and SHI, Y., 2022. An epigenetic role of mitochondria in cancer. Cells, vol. 11, no. 16, pp. 2518. http://dx.doi.org/10.3390/cells11162518. PMid:36010594.
http://dx.doi.org/10.3390/cells11162518...
). These alterations control how histones interact with chromatin remodeling complexes and DNA. The tumor microenvironment can alter cell fate through epigenetic control. Tumors can be targeted by mutations in important TCA cycle metabolic enzymes, and cell proliferation can be inhibited by inhibitors that target cancer stemness. Designing treatments for the diagnosis and treatment of cancer can be aided by knowledge of carcinogenesis, mitochondrial metabolites, and epigenetics.

According to Abdullah et al. (2024)ABDULLAH, S.S., MASOOD, S., ZANEB, H., RABBANI, I., AKBAR, J., KUTHU, Z.H., MASOOD, A. and VARGAS-BELLO-PÉREZ, E., 2024. Effects of copper nanoparticles on performance, muscle and bone characteristics and serum metabolites in broilers. Brazilian Journal of Biology = Revista Brasileira de Biologia, vol. 84, e261578. http://dx.doi.org/10.1590/1519-6984.261578. PMid:35730812.
http://dx.doi.org/10.1590/1519-6984.2615...
, broiler weight gain and feed conversion ratio (FCR) were enhanced by feeding copper nanoparticles at a dose of 15 mg/kg. Furthermore, it has enhanced the bone and muscular features of broilers.

Chitosan has also been shown to lessen plasma lipids and increase body weight in mice (Kumar et al., 2009KUMAR, S.G., RAHMAN, M.A., LEE, S.H., HWANG, H.S., KIM, H.A. and YUN, J.W., 2009. Plasma proteome analysis for anti‐obesity and anti‐diabetic potentials of chitosan oligosaccharides in ob/ob mice. Proteomics, vol. 9, no. 8, pp. 2149-2162. http://dx.doi.org/10.1002/pmic.200800571. PMid:19296549.
http://dx.doi.org/10.1002/pmic.200800571...
). Furthermore, Kamal et al. (2023a)KAMAL, M., KISHK, W.H., KHALIL, H.A., ABDEL-KHALEK, A.M., AYOUB, M.A., SWELUM, A.A., ALQHTANI, A.H., BA-AWADH, H.A. and ABD EL-HACK, M.E., 2023a. Effect of dietary chitosan supplementation on productive and physiological performance parameters of growing New Zealand white rabbits. International Journal of Biological Macromolecules, vol. 230, pp. 123166. http://dx.doi.org/10.1016/j.ijbiomac.2023.123166. PMid:36623627.
http://dx.doi.org/10.1016/j.ijbiomac.202...
described that the chitosan 0.2 g/kg diet enhanced the serum triglyceride and HDL, improving the health profile of NZW rabbits in Egyptian environments. The suppression of adipogenesis machinery is one method for avoiding and managing obesity.

In vitro trials demonstrated that chitosan could constrain the differentiation of 3T3-L1 preadipocytes and lessen fat gathering by decreasing the transcripts of PPAR-γ (peroxisome proliferator-activated receptor γ) and CCAAT enhancer-binding proteins α (C/EBP α), which are the main adipogenesis-associated transcription features (Cho et al., 2008CHO, E.J., RAHMAN, A., KIM, S.W., BAEK, Y.M., HWANG, H.J., OH, J.Y., HWANG, H.S., LEE, S.H. and YUN, J.W., 2008. Chitosan oligosaccharides inhibit adipogenesis in 3T3-L1 adipocytes. Journal of Microbiology and Biotechnology, vol. 18, no. 1, pp. 80-87. PMid:18239421.; Kong et al., 2017KONG, S., DING, C., HUANG, L., BAI, Y., XIAO, T., GUO, J. and SU, Z., 2017. The effects of COST on the differentiation of 3T3-L1 preadipocytes and the mechanism of action. Saudi Journal of Biological Sciences, vol. 24, no. 2, pp. 251-255. http://dx.doi.org/10.1016/j.sjbs.2016.09.008. PMid:28149159.
http://dx.doi.org/10.1016/j.sjbs.2016.09...
). Additionally, chitosan reduced the transcript of associated elements in 3T3-L1 adipocytes, comprising leptin, adiponectin, resisting, FAS (fatty acid synthase), FABP (fatty acid binding protein), and GLUT4 (glucose transporter 4); (Rahman et al., 2008RAHMAN, M.A., KUMAR, S.G., KIM, S.W., HWANG, H.J., BAEK, Y.M., LEE, S.H., HWANG, H.S., SHON, Y.H., NAM, K.S. and YUN, J.W., 2008. Proteomic analysis for inhibitory effect of chitosan oligosaccharides on 3T3‐L1 adipocyte differentiation. Proteomics, vol. 8, no. 3, pp. 569-581. http://dx.doi.org/10.1002/pmic.200700888. PMid:18175373.
http://dx.doi.org/10.1002/pmic.200700888...
; Lee et al., 2021LEE, J.Y., KIM, T.Y., KANG, H., OH, J., PARK, J.W., KIM, S.C., KIM, M., APOSTOLIDIS, E., KIM, Y.C. and KWON, Y.I., 2021. Anti-obesity and anti-adipogenic effects of chitosan oligosaccharide (GO2KA1) in SD rats and in 3T3-L1 preadipocytes models. Molecules, vol. 26, no. 2, pp. 331. http://dx.doi.org/10.3390/molecules26020331. PMid:33440605.
http://dx.doi.org/10.3390/molecules26020...
).

Remarkably, Bahar et al. (2013)BAHAR, B., O’DOHERTY, J.V., O’DOHERTY, A.M. and SWEENEY, T., 2013. Chito-oligosaccharide inhibits the de-methylation of a ‘CpG’ Island within the leptin (LEP) promoter during adipogenesis of 3T3-L1 cells. PLoS One, vol. 8, no. 3, e60011. http://dx.doi.org/10.1371/journal.pone.0060011. PMid:23544120.
http://dx.doi.org/10.1371/journal.pone.0...
described that chitosan repressed the de-methylation of leptin gene promoter in 3T3-L1 adipocytes, representing that chitosan decreased the differential of adipocytes via epigenetic machinery. Additionally, suppression of the PPAR-signaling way was one of the methods by which chitosan prevented hypertrophy and adipocyte hyperplasia in HFD-fed rats by controlling the transcriptomic related to lipogenesis in the adipose tissue (Huang et al., 2015HUANG, L., CHEN, J., CAO, P., PAN, H., DING, C., XIAO, T., ZHANG, P., GUO, J. and SU, Z., 2015. Anti-obese effect of glucosamine and chitosan oligosaccharide in high-fat diet-induced obese rats. Marine Drugs, vol. 13, no. 5, pp. 2732-2756. http://dx.doi.org/10.3390/md13052732. PMid:25942093.
http://dx.doi.org/10.3390/md13052732...
; Pan et al., 2018PAN, H., FU, C., HUANG, L., JIANG, Y., DENG, X., GUO, J. and SU, Z., 2018. Anti-obesity effect of chitosan oligosaccharide capsules (COSCs) in obese rats by ameliorating leptin resistance and adipogenesis. Marine Drugs, vol. 16, no. 6, pp. 198. http://dx.doi.org/10.3390/md16060198. PMid:29874843.
http://dx.doi.org/10.3390/md16060198...
).

According to an in vitro investigation, chitosan stimulated the PPAR γ signaling pathway to reduce fat formation in HepG2 cells exposed to palmitic acid (Bai et al., 2018BAI, Y., ZHENG, J., YUAN, X., JIAO, S., FENG, C., DU, Y., LIU, H. and ZHENG, L., 2018. Chitosan oligosaccharides improve glucolipid metabolism disorder in liver by suppression of obesity-related inflammation and restoration of peroxisome proliferator-activated receptor gamma (PPARγ). Marine Drugs, vol. 16, no. 11, pp. 455. http://dx.doi.org/10.3390/md16110455. PMid:30463189.
http://dx.doi.org/10.3390/md16110455...
). Also, chitosan has been shown to be efficient in reducing hepatic lipid accumulation, hepatic steatosis, and serum activities of both aspartate and alanine aminotransferases in obese rats or mice generated by HFD (Liu et al., 2018LIU, S.H., CHIU, C.Y., SHI, C.M. and CHIANG, M.T., 2018. Functional comparison of high and low molecular weight chitosan on lipid metabolism and signals in high-fat diet-fed rats. Marine Drugs, vol. 16, no. 8, pp. 251. http://dx.doi.org/10.3390/md16080251. PMid:30060615.
http://dx.doi.org/10.3390/md16080251...
; Tao et al., 2019TAO, W., SUN, W., LIU, L., WANG, G., XIAO, Z., PEI, X. and WANG, M., 2019. Chitosan oligosaccharide attenuates nonalcoholic fatty liver disease induced by high fat diet through reducing lipid accumulation, inflammation and oxidative stress in C57BL/6 mice. Marine Drugs, vol. 17, no. 11, pp. 645. http://dx.doi.org/10.3390/md17110645. PMid:31744059.
http://dx.doi.org/10.3390/md17110645...
). Chitosan enhanced intestinal barrier anomalies and dysbiosis of the gastro-microbiota in mice on an HFD, according to research by (He et al., 2020HE, N., WANG, S., LV, Z., ZHAO, W. and LI, S., 2020. Low molecular weight chitosan oligosaccharides (LMW-COSs) prevent obesity-related metabolic abnormalities in association with the modification of gut microbiota in high-fat diet (HFD)-fed mice. Food & Function, vol. 11, no. 11, pp. 9947-9959. http://dx.doi.org/10.1039/D0FO01871F. PMid:33108433.
http://dx.doi.org/10.1039/D0FO01871F...
). Notably, the release of LPS (lipopolysaccharide), a constituent of the cellular structure of Gram-negative bacteria, into the blood to promote inflammation resulted from intestinal epithelium malfunction and dysbiosis of microbiota (Cani and Jordan, 2018CANI, P.D. and JORDAN, B.F., 2018. Gut microbiota-mediated inflammation in obesity: a link with gastrointestinal cancer. Nature Reviews. Gastroenterology & Hepatology, vol. 15, no. 11, pp. 671-682. http://dx.doi.org/10.1038/s41575-018-0025-6. PMid:29844585.
http://dx.doi.org/10.1038/s41575-018-002...
). Furthermore, chitosan was found to reduce inflammatory blood markers, hepatic, fat tissues, and colon of HFD-triggered obese rats or mice, according to several research (Bai et al., 2018BAI, Y., ZHENG, J., YUAN, X., JIAO, S., FENG, C., DU, Y., LIU, H. and ZHENG, L., 2018. Chitosan oligosaccharides improve glucolipid metabolism disorder in liver by suppression of obesity-related inflammation and restoration of peroxisome proliferator-activated receptor gamma (PPARγ). Marine Drugs, vol. 16, no. 11, pp. 455. http://dx.doi.org/10.3390/md16110455. PMid:30463189.
http://dx.doi.org/10.3390/md16110455...
; He et al., 2020HE, N., WANG, S., LV, Z., ZHAO, W. and LI, S., 2020. Low molecular weight chitosan oligosaccharides (LMW-COSs) prevent obesity-related metabolic abnormalities in association with the modification of gut microbiota in high-fat diet (HFD)-fed mice. Food & Function, vol. 11, no. 11, pp. 9947-9959. http://dx.doi.org/10.1039/D0FO01871F. PMid:33108433.
http://dx.doi.org/10.1039/D0FO01871F...
). These results mention that chitosan and its nano-formulation might be utilized to prevent or treat dyslipidemia and obesity.

The potential role of chitosan and its nano-formulation may inhibit adipogenesis, control liver lipid metabolism, enhance intestinal barrier malfunction, and dysbiosis of the gut microbiota (Figure 3). Moreover, Chen et al. (2020)CHEN, L., WANG, C. and WU, Y., 2020. Cholesterol (Blood lipid) lowering potential of Rosuvastatin chitosan nanoparticles for atherosclerosis: preclinical study in rabbit model. Acta Biochimica Polonica, vol. 67, no. 4, pp. 495-499. http://dx.doi.org/10.18388/abp.2020_5186. PMid:33090754.
http://dx.doi.org/10.18388/abp.2020_5186...
found that rosuvastatin-loaded chitosan nanoparticles are more effective in depressing blood fat than clean rosuvastatin. Its assistance in macerating the calcification of different valve tissues in rabbit models. Also, Luo et al. (2021)LUO, G., CHEN, W., LUO, J. and LIU, J., 2021. Control and monitoring of lipoprotein levels in atherosclerosis induced rabbits using novel nanoparticulate medication of Lovastatin and Rosuvastatin. Micro & Nano Letters, vol. 16, no. 11, pp. 558-565. http://dx.doi.org/10.1049/mna2.12081.
http://dx.doi.org/10.1049/mna2.12081...
presented that the management of chitosan NPs resulted in inferior blood LDL, total cholesterol, and uric acid. Furthermore, Abd-Elhakeem et al. (2016)ABD-ELHAKEEM, M.A., FARAG, N. and MAURICE, M., 2016. Effects of dietary chitosan nanoparticles on serum lipid concentration in hyperlipidemic rats induced by a high-fat diet. Egyptian Journal of Pure and Applied Science, vol. 54, no. 4, pp. 17-21. http://dx.doi.org/10.21608/ejaps.2016.184579.
http://dx.doi.org/10.21608/ejaps.2016.18...
signposted that giving rats chitosan and chitosan NPs reduced body weight gain and serum cholesterol levels.

Figure 3
Medical role of chitosan nanoparticles.

In the same context, Oksal et al. (2020)OKSAL, E., PANGESTIKA, I., MUHAMMAD, T.S.T., MOHAMAD, H., AMIR, H., KASSIM, M.N.I. and ANDRIANI, Y., 2020. In vitro and in vivo studies of nanoparticles of chitosan-Pandanus tectorius fruit extract as new alternative treatment for hypercholesterolemia via Scavenger Receptor Class B type 1 pathway. Saudi Pharmaceutical Journal, vol. 28, no. 10, pp. 1263-1275. http://dx.doi.org/10.1016/j.jsps.2020.08.017. PMid:33132720.
http://dx.doi.org/10.1016/j.jsps.2020.08...
mentioned that the chitosan-Pandanus tectorius fruit extract nanoparticles could decline the total cholesterol, LDL, and triglyceride amounts but also escalate the HDL amounts. Also, chitosan-Pandanus tectorius fruit extract nanoparticles will likely be applied as a novel unconventional management for hypercholesterolemia via the SR-B1 path. Additionally, Sriamornsak and Dass, (2022)SRIAMORNSAK, P. and DASS, C.R., 2022. Chitosan nanoparticles in atherosclerosis: development to preclinical testing. Pharmaceutics, vol. 14, no. 5, pp. 935. http://dx.doi.org/10.3390/pharmaceutics14050935. PMid:35631521.
http://dx.doi.org/10.3390/pharmaceutics1...
mentioned that chitosan and chitosan NPs could be utilized to create various medication formulations. They also naturally lower cholesterol. As illustrated in Table 1, we summarized some trials on the favorable impacts of chitosan and nano-formulation on obesity and dyslipidemia.

Table 1
Summary of some studies on the beneficial effects of chitosan and nano-formulation on obesity and dyslipidemia.

7. Impacts of Chitosan and Its Nanoformulation on Hyperglycemia and Diabetes Mellitus

Diabetes mellitus is distinguished by chronic hyperglycemia caused by unbefitting secretion or incompetent uptake of insulin (Kharroubi and Darwish, 2015KHARROUBI, A.T. and DARWISH, H.M., 2015. Diabetes mellitus: the epidemic of the century. World Journal of Diabetes, vol. 6, no. 6, pp. 850-867. http://dx.doi.org/10.4239/wjd.v6.i6.850. PMid:26131326.
http://dx.doi.org/10.4239/wjd.v6.i6.850...
). The anti-diabetic action of chitosan has been presented using different diabetic models. The management of chitosan enhanced the broad state and diabetic indications, diminished the amounts of glucose in the blood and urine, as well as regularized decreased glucose tolerance in newborn STZ (streptozotocin)- triggered type 2 diabetic in rats, a model of non-insulin- entrusted diabetes mellitus (Liu et al., 2009LIU, H.T., LI, W.M., XU, G., LI, X.Y., BAI, X.F., WEI, P., YU, C. and DU, Y.G., 2009. Chitosan oligosaccharides attenuate hydrogen peroxide-induced stress injury in human umbilical vein endothelial cells. Pharmacological Research, vol. 59, no. 3, pp. 167-175. http://dx.doi.org/10.1016/j.phrs.2008.12.001. PMid:19121394.
http://dx.doi.org/10.1016/j.phrs.2008.12...
). Ju et al. (2010)JU, C., YUE, W., YANG, Z., ZHANG, Q., YANG, X., LIU, Z. and ZHANG, F., 2010. Antidiabetic effect and mechanism of chitooligosaccharides. Biological & Pharmaceutical Bulletin, vol. 33, no. 9, pp. 1511-1516. http://dx.doi.org/10.1248/bpb.33.1511. PMid:20823566.
http://dx.doi.org/10.1248/bpb.33.1511...
demonstrated that in insulin-resistant rats produced by a high-energy diet combined with STZ, chitosan administration for eight weeks led to diminished fasting insulin amounts and fasting blood glucose, further elevated insulin sensitivity directory and enhanced oral glucose tolerance.

According to Katiyar et al. (2011)KATIYAR, D., SINGH, B., LALL, A.M. and HALDAR, C., 2011. Efficacy of chitooligosaccharides for the management of diabetes in alloxan induced mice: a correlative study with antihyperlipidemic and antioxidative activity. European Journal of Pharmaceutical Sciences, vol. 44, no. 4, pp. 534-543. http://dx.doi.org/10.1016/j.ejps.2011.09.015. PMid:21964204.
http://dx.doi.org/10.1016/j.ejps.2011.09...
, chitosan significantly improved renal dysfunction and blood glucose control in alloxan-induced diabetic rats. Chitosan has also been shown to be inferior to blood glucose in mice (Zheng et al., 2018ZHENG, J., YUAN, X., CHENG, G., JIAO, S., FENG, C., ZHAO, X., YIN, H., DU, Y. and LIU, H., 2018. Chitosan oligosaccharides improve the disturbance in glucose metabolism and reverse the dysbiosis of gut microbiota in diabetic mice. Carbohydrate Polymers, vol. 190, pp. 77-86. http://dx.doi.org/10.1016/j.carbpol.2018.02.058. PMid:29628262.
http://dx.doi.org/10.1016/j.carbpol.2018...
). In Korean patients between the ages of 20 and 75, a chitosan addition of 1.5g per day for three months effectively lowered the postprandial serum glucose amounts, according to Kim et al. (2014a).KIM, H.J., AHN, H.Y., KWAK, J.H., SHIN, D.Y., KWON, Y.I., OH, C.G. and LEE, J.H., 2014a. The effects of chitosan oligosaccharide (GO2KA1) supplementation on glucose control in subjects with prediabetes. Food & Function, vol. 5, no. 10, pp. 2662-2669. http://dx.doi.org/10.1039/C4FO00469H. PMid:25222285.
http://dx.doi.org/10.1039/C4FO00469H...
Additionally, Yuan et al. (2009)YUAN, W.P., LIU, B., LIU, C.H., WANG, X.J., ZHANG, M.S., MENG, X.M. and XIA, X.K., 2009. Antioxidant activity of chito-oligosaccharides on pancreatic islet cells in streptozotocin-induced diabetes in rats. World Journal of Gastroenterology, vol. 15, no. 11, pp. 1339-1345. http://dx.doi.org/10.3748/wjg.15.1339. PMid:19294763.
http://dx.doi.org/10.3748/wjg.15.1339...
described that chitosan reduced the damage of pancreatic islets, nuclear pyknosis of pancreatic cells, and atrophy of pancreatic cells in STZ-triggered diabetic rats. Additionally, in vitro research has demonstrated that chitosan can enhance pancreatic cell line apoptosis caused by STZ and stimulate cell growth (Ju et al., 2010JU, C., YUE, W., YANG, Z., ZHANG, Q., YANG, X., LIU, Z. and ZHANG, F., 2010. Antidiabetic effect and mechanism of chitooligosaccharides. Biological & Pharmaceutical Bulletin, vol. 33, no. 9, pp. 1511-1516. http://dx.doi.org/10.1248/bpb.33.1511. PMid:20823566.
http://dx.doi.org/10.1248/bpb.33.1511...
). Yuan et al. (2009)YUAN, W.P., LIU, B., LIU, C.H., WANG, X.J., ZHANG, M.S., MENG, X.M. and XIA, X.K., 2009. Antioxidant activity of chito-oligosaccharides on pancreatic islet cells in streptozotocin-induced diabetes in rats. World Journal of Gastroenterology, vol. 15, no. 11, pp. 1339-1345. http://dx.doi.org/10.3748/wjg.15.1339. PMid:19294763.
http://dx.doi.org/10.3748/wjg.15.1339...
indicated that chitosan increased the levels of superoxide dismutase and total antioxidant capability and depressed the amount of malondialdehyde (MDA) in the serum of STZ-triggered rats.

Karadeniz et al. (2010)KARADENIZ, F., ARTAN, M., KONG, C.S. and KIM, S.K., 2010. Chitooligosaccharides protect pancreatic β-cells from hydrogen peroxide-induced deterioration. Carbohydrate Polymers, vol. 82, no. 1, pp. 143-147. http://dx.doi.org/10.1016/j.carbpol.2010.04.046.
http://dx.doi.org/10.1016/j.carbpol.2010...
also showed that chitosan could have free radical scavenging properties to defend against the oxidative stress that hydrogen peroxide (H2O2) causes in cells. These results imply that chitosan may serve as a free radical scavenger or improve the antioxidant competence to protect pancreatic cells. It can be inferred that chitosan's ability to lower oxidative stress and prevent human islet amyloid polypeptide aggregation makes it an effective antidiabetic medication (Meng et al., 2020MENG, Q.Y., WANG, H., CUI, Z.B., YU, W.G. and LU, X.Z., 2020. Chitosan oligosaccharides attenuate amyloid formation of hIAPP and protect pancreatic β-cells from cytotoxicity. Molecules, vol. 25, no. 6, pp. 1314. http://dx.doi.org/10.3390/molecules25061314. PMid:32183067.
http://dx.doi.org/10.3390/molecules25061...
).

Insulin resistance is one of the first signs of diabetes. Chitosan has decreased insulin resistance, which is mentioned to lower sensitivity and responsiveness to insulin in board tissues such as the hepatic, adipocytes, and skeletal muscle tissues (Czech, 2017CZECH, M.P., 2017. Insulin action and resistance in obesity and type 2 diabetes. Nature Medicine, vol. 23, no. 7, pp. 804-814. http://dx.doi.org/10.1038/nm.4350. PMid:28697184.
http://dx.doi.org/10.1038/nm.4350...
). Ju et al. (2010)JU, C., YUE, W., YANG, Z., ZHANG, Q., YANG, X., LIU, Z. and ZHANG, F., 2010. Antidiabetic effect and mechanism of chitooligosaccharides. Biological & Pharmaceutical Bulletin, vol. 33, no. 9, pp. 1511-1516. http://dx.doi.org/10.1248/bpb.33.1511. PMid:20823566.
http://dx.doi.org/10.1248/bpb.33.1511...
revealed that chitosan supplementation increased the insulin sensitivity marker and glucose tolerance in high-energy diet-paired STZ-triggered diabetic mice. Additionally, mouse research found chitosan dramatically reduces insulin resistance (Zheng et al., 2018ZHENG, J., YUAN, X., CHENG, G., JIAO, S., FENG, C., ZHAO, X., YIN, H., DU, Y. and LIU, H., 2018. Chitosan oligosaccharides improve the disturbance in glucose metabolism and reverse the dysbiosis of gut microbiota in diabetic mice. Carbohydrate Polymers, vol. 190, pp. 77-86. http://dx.doi.org/10.1016/j.carbpol.2018.02.058. PMid:29628262.
http://dx.doi.org/10.1016/j.carbpol.2018...
). Numerous essential elements of the insulin signaling pathway, comprising the insulin receptor substrate, insulin receptor, phosphoinositide 3-kinase, and Akt, have been identified (James et al., 2021JAMES, D.E., STÖCKLI, J. and BIRNBAUM, M.J., 2021. The aetiology and molecular landscape of insulin resistance. Nature Reviews Molecular Cell Biology, vol. 22, no. 11, pp. 751-771. http://dx.doi.org/10.1038/s41580-021-00390-6. PMid:34285405.
http://dx.doi.org/10.1038/s41580-021-003...
). By blocking the actions of glucoamylase and intestinal sucrose and decreasing the mRNA transcript of the sucrase-isomaltose complex in mice, long-term supplementation with chitosan dramatically diminished the amounts of glycated hemoglobin A1c and blood glucose (Kim et al., 2014bKIM, J.G., JO, S.H., HA, K.S., KIM, S.C., KIM, Y.C., APOSTOLIDIS, E. and KWON, Y.I., 2014b. Effect of long-term supplementation of low molecular weight chitosan oligosaccharide (GO2KA1) on fasting blood glucose and HbA1c in db/db mice model and elucidation of mechanism of action. BMC Complementary and Alternative Medicine, vol. 14, no. 1, pp. 272. http://dx.doi.org/10.1186/1472-6882-14-272. PMid:25074485.
http://dx.doi.org/10.1186/1472-6882-14-2...
). A study by Jo et al. (2013)JO, S.H., HA, K.S., MOON, K.S., KIM, J.G., OH, C.G., KIM, Y.C., APOSTOLIDIS, E. and KWON, Y.I., 2013. Molecular weight dependent glucose lowering effect of low molecular weight chitosan oligosaccharide (GO2KA1) on postprandial blood glucose level in SD rats model. International Journal of Molecular Sciences, vol. 14, no. 7, pp. 14214-14224. http://dx.doi.org/10.3390/ijms140714214. PMid:23839092.
http://dx.doi.org/10.3390/ijms140714214...
described that chitosan diminished the SI multifaceted mRNA transcript and hindered the glucosidase activities in human intestinal cells.

Chitosan is suitable for use because of its precautionary, biodegradability, biocompatibility, strong adhesiveness, ease, and permeability in the intestinal region (Souto et al., 2019SOUTO, E.B., SOUTO, S.B., CAMPOS, J.R., SEVERINO, P., PASHIROVA, T.N., ZAKHAROVA, L.Y., SILVA, A.M., DURAZZO, A., LUCARINI, M., IZZO, A.A. and SANTINI, A., 2019. Nanoparticle delivery systems in the treatment of diabetes complications. Molecules, vol. 24, no. 23, pp. 4209. http://dx.doi.org/10.3390/molecules24234209. PMid:31756981.
http://dx.doi.org/10.3390/molecules24234...
). Additionally, it is known that chitosan-based nanoplatforms can deliver anti-cancer medications (Jaiswal et al., 2021JAISWAL, S., DUTTA, P.K., KUMAR, S. and CHAWLA, R., 2021. Chitosan modified by organo-functionalities as an efficient nanoplatform for anti-cancer drug delivery process. Journal of Drug Delivery Science and Technology, vol. 62, pp. 102407. http://dx.doi.org/10.1016/j.jddst.2021.102407.
http://dx.doi.org/10.1016/j.jddst.2021.1...
). There are signs that chitosan-based nanoparticles containing oleic acid can protect insulin from being degraded by enzymes (Elsayed et al., 2010ELSAYED, A., AL-REMAWI, M., FAROUK, A. and BADWAN, A., 2010. Insulin-chitosan polyelectrolyte _anocomplexes: preparation, characterization and stabilization of insulin. Sudan Journal of Medical Sciences, vol. 5, no. 2, pp. 99-109. http://dx.doi.org/10.4314/sjms.v5i2.57799.
http://dx.doi.org/10.4314/sjms.v5i2.5779...
). Also, applying a complexing manager in chitosan nanoparticles improved insulin absorption (Lin et al., 2007LIN, Y.H., MI, F.L., CHEN, C.T., CHANG, W.C., PENG, S.F., LIANG, H.F. and SUNG, H.W., 2007. Preparation and characterization of nanoparticles shelled with chitosan for oral insulin delivery. Biomacromolecules, vol. 8, no. 1, pp. 146-152. http://dx.doi.org/10.1021/bm0607776. PMid:17206800.
http://dx.doi.org/10.1021/bm0607776...
; Chuang et al., 2013CHUANG, E.Y., LIN, K.J., SU, F.Y., CHEN, H.L., MAITI, B., HO, Y.C., YEN, T.C., PANDA, N. and SUNG, H.W., 2013. Calcium depletion-mediated protease inhibition and apical-junctional-complex disassembly via an EGTA-conjugated carrier for oral insulin delivery. Journal of Controlled Release, vol. 169, no. 3, pp. 296-305. http://dx.doi.org/10.1016/j.jconrel.2012.11.011. PMid:23195534.
http://dx.doi.org/10.1016/j.jconrel.2012...
). To increase hydrophilicity, mucoadhesiveness, and permeability in an alkaline media, chitosan derivatives such as thiolated chitosan, trimethyl chitosan, carboxylated chitosan, etc., were utilized in the production of nanoparticles (Wong et al., 2017WONG, C.Y., AL-SALAMI, H. and DASS, C.R., 2017. Potential of insulin nanoparticle formulations for oral delivery and diabetes treatment. Journal of Controlled Release, vol. 264, pp. 247-275. http://dx.doi.org/10.1016/j.jconrel.2017.09.003. PMid:28887133.
http://dx.doi.org/10.1016/j.jconrel.2017...
). Additionally, at a pH of 7.4, it was found that trimethylated chitosan nanoparticles linked with insulin were steadier with constant insulin statements (Mi et al., 2008MI, F.L., WU, Y.Y., LIN, Y.H., SONAJE, K., HO, Y.C., CHEN, C.T., JUANG, J.H. and SUNG, H.W., 2008. Oral delivery of peptide drugs using nanoparticles self-assembled by poly (γ-glutamic acid) and a chitosan derivative functionalized by trimethylation. Bioconjugate Chemistry, vol. 19, no. 6, pp. 1248-1255. http://dx.doi.org/10.1021/bc800076n. PMid:18517235.
http://dx.doi.org/10.1021/bc800076n...
).

According to Abd El-Hameed (2020)ABD EL-HAMEED, A.M., 2020. Polydatin-loaded chitosan nanoparticles ameliorates early diabetic nephropathy by attenuating oxidative stress and inflammatory responses in streptozotocin-induced diabetic rat. Journal of Diabetes and Metabolic Disorders, vol. 19, no. 2, pp. 1599-1607. http://dx.doi.org/10.1007/s40200-020-00699-7. PMid:33520856.
http://dx.doi.org/10.1007/s40200-020-006...
, the new polydatin-coated (POL) chitosan-nan nanoparticle formulation is biocompatible. It may gradually improve diabetic rats' nephropathy compared to free POL. The research also found that POL-nanoparticles have nephroprotective activity on diabetic nephropathy may be owing to its antidiabetic capability via the elevation of insulin secretion, regulation of HbA1c and blood glucose, (1) blocking oxidative stress synthesis through its antioxidant upshot and dropping AGEs creation, (2) its function as an anti-inflammatory mediator, and (3) The greatest beneficial effect of POL chitosan nanoparticles may be accredited by improving absorption and prolonged-release possessions.

The study of Salem et al. (2021)SALEM, S.A., DIAB, M.S.M., SHEHATA, S.H. and SOLIMAN, T.N., 2021. Nanochitosan effect on biomolecular, hypolipidemic in rats and incorporation in functional yogurt. Pakistan Journal of Biological Sciences, vol. 24, no. 5, pp. 548-561. http://dx.doi.org/10.3923/pjbs.2021.548.561. PMid:34486330.
http://dx.doi.org/10.3923/pjbs.2021.548....
informed that chitosan nanoparticles could enhance the deficiency of fat metabolism as powerfully related to transcriptomics alterations correlated with lipogenesis and oxidative markers. Additionally, camel yogurt with 2% chitosan nanoparticles had good sensory and microbiological quality. Likewise, according to a study, 20 mg/kg of chitosan and nano-chitosan in guinea pigs dramatically decreased fasting blood glucose and enhanced renal function (Sami et al., 2022SAMI, M.M., AL-KAZZAZ, F.F. and ABDULSATTAR, S.A., 2022. The effect of using Chitosan and Nano-chitosan synthesized from blue carb in the treatment of hyperglycemia and glomerulus of diabetic guinea pigs. Egyptian Journal of Chemistry, vol. 65, no. 6, pp. 55-58. http://dx.doi.org/10.21608/EJCHEM.2021.85130.4151.
http://dx.doi.org/10.21608/EJCHEM.2021.8...
). According to Zhang et al. (2021)ZHANG, M., HOU, G., HU, P., FENG, D., WANG, J. and ZHU, W., 2021. Nano chitosan-zinc complex improves the growth performance and antioxidant capacity of the small intestine in weaned piglets. British Journal of Nutrition, vol. 126, no. 6, pp. 801-812. http://dx.doi.org/10.1017/S0007114520004766. PMid:33256856.
http://dx.doi.org/10.1017/S0007114520004...
, nano chitosan-zinc supplementation can enhance piglet small intestine antioxidant capacity and growth performance, reducing weaning stress. Chandrasekaran et al. (2020)CHANDRASEKARAN, M., KIM, K.D. and CHUN, S.C., 2020. Antibacterial activity of chitosan nanoparticles: a review. Processes, vol. 8, no. 9, pp. 1173. http://dx.doi.org/10.3390/pr8091173.
http://dx.doi.org/10.3390/pr8091173...
reported that both G- and G+ bacteria are inhibited by the biological use of chitosan nanoparticles, alone or in combination with other chemicals. Also, Zhang et al. (2012)ZHANG, X., SUN, M., ZHENG, A., CAO, D., BI, Y. and SUN, J., 2012. Preparation and characterization of insulin-loaded bioadhesive PLGA nanoparticles for oral administration. European Journal of Pharmaceutical Sciences, vol. 45, no. 5, pp. 632-638. http://dx.doi.org/10.1016/j.ejps.2012.01.002. PMid:22248882.
http://dx.doi.org/10.1016/j.ejps.2012.01...
reported that cationic chitosan might increase the strong adhesiveness of polylactic-co-glycolic acid nanoparticles in the GIT.

Additionally, Adetunji et al. (2022)ADETUNJI, C.O., MICHAEL, O.S., RATHEE, S., SINGH, K.R.B., AJAYI, O.O., ADETUNJI, J.B., OJHA, A., SINGH, J. and SINGH, R.P., 2022. Potentialities of nanomaterials for the management and treatment of metabolic syndrome: a new insight. Materials Today Advances, vol. 13, pp. 100198. http://dx.doi.org/10.1016/j.mtadv.2021.100198.
http://dx.doi.org/10.1016/j.mtadv.2021.1...
showed that nanoparticles significantly function in drug delivery to delicacy various metabolic syndromes. In the remedy of different sicknesses, including cancer and metabolic disorders, nanomaterials can potentially reduce the dosage of medications or their negative side effects. All these findings might suggest that chitosan and chitosan NPs can prevent diabetes by preventing the enzymes that break down carbohydrates in the intestine. According to in vivo and in vitro investigations, the beneficial effects of chitosan and nano-formulation on diabetes mellitus and hyperglycemia are illustrated in Table 2.

Table 2
Summary of some studies on the beneficial effects of chitosan and nano-formulation on diabetes mellitus and hyperglycemia.

8. Impacts of Chitosan and Its Nanoformulation on Hypertension

Cardiovascular illness, chronic renal disease, and cognitive impairment are all conditions that are thought to be significantly increased by hypertension (Iadecola and Gottesman, 2019IADECOLA, C. and GOTTESMAN, R.F., 2019. Neurovascular and cognitive dysfunction in hypertension: epidemiology, pathobiology, and treatment. Circulation Research, vol. 124, no. 7, pp. 1025-1044. http://dx.doi.org/10.1161/CIRCRESAHA.118.313260. PMid:30920929.
http://dx.doi.org/10.1161/CIRCRESAHA.118...
; Fuchs and Whelton, 2020FUCHS, F.D. and WHELTON, P.K., 2020. High blood pressure and cardiovascular disease. Hypertension, vol. 75, no. 2, pp. 285-292. http://dx.doi.org/10.1161/HYPERTENSIONAHA.119.14240. PMid:31865786.
http://dx.doi.org/10.1161/HYPERTENSIONAH...
). Heredities, the stimulation of the RAAS (renin-angiotensin-aldosterone system), the stimulation of the sympathetic nervous scheme, endothelial dysfunction, vascular remodeling, insulin resistance, and defective ion channels are specific the elements linked to the pathophysiology of hypertension (Oparil et al., 2003OPARIL, S., ZAMAN, M.A. and CALHOUN, D.A., 2003. Pathogenesis of hypertension. Annals of Internal Medicine, vol. 139, no. 9, pp. 761-776. http://dx.doi.org/10.7326/0003-4819-139-9-200311040-00011. PMid:14597461.
http://dx.doi.org/10.7326/0003-4819-139-...
). According to one of the investigations, a single oral dosage of chitosan trimer (2.14 mg/kg) effectively lowered blood tension in impulsively hypertensive rat models. Chitosan's ability to lower blood pressure may be connected to its ability to suppress RAAS and alleviate endothelial dysfunction.

Chitosan reduced ACE (angiotensin-I converting enzyme) activities at different polymerization levels (DP), ranging from 1 to 10 (Park et al., 2008PARK, P.J., AHN, C.B., JEON, Y.J. and JE, J.Y., 2008. Renin inhibition activity by chitooligosaccharides. Bioorganic & Medicinal Chemistry Letters, vol. 18, no. 7, pp. 2471-2474. http://dx.doi.org/10.1016/j.bmcl.2008.02.041. PMid:18313296.
http://dx.doi.org/10.1016/j.bmcl.2008.02...
). The chitosan trimer (DP = 3) displayed all the oligosaccharides' strongest inhibitory action. Additionally, the MW (molecular weight) degree of DD (deacetylation) of chitosan affects its ability to inhibit ACE. Chitosan with a regular MW of 1-5 kDa has greater ACE restrained action than chitosan with a regular MW of <1 kDa or 5-10 kDa (Park et al., 2003PARK, P.J., JE, J.Y. and KIM, S.K., 2003. Angiotensin I converting enzyme (ACE) inhibitory activity of hetero-chitooligosaccharides prepared from partially different deacetylated chitosans. Journal of Agricultural and Food Chemistry, vol. 51, no. 17, pp. 4930-4934. http://dx.doi.org/10.1021/jf0340557. PMid:12903948.
http://dx.doi.org/10.1021/jf0340557...
).

Liu et al. (2007b)LIU, H., CHEN, B., MAO, Z. and GAO, C., 2007b. Chitosan nanoparticles for loading of toothpaste actives and adhesion on tooth analogs. Journal of Applied Polymer Science, vol. 106, no. 6, pp. 4248-4256. http://dx.doi.org/10.1002/app.27078.
http://dx.doi.org/10.1002/app.27078...
stated that chitosan decreased intracellular oxidative markers, suppressed the construction of MDA, restored the actions of cellular antioxidants, increased levels of NO (nitric oxide) and NO synthase, and decreased cell apoptosis to decrease H2O2-triggered oxidative stress in endothelial cells. Also, Li et al. (2014)LI, Y., LIU, H., XU, Q.S., DU, Y.G. and XU, J., 2014. Chitosan oligosaccharides block LPS-induced O-GlcNAcylation of NF-κB and endothelial inflammatory response. Carbohydrate Polymers, vol. 99, pp. 568-578. http://dx.doi.org/10.1016/j.carbpol.2013.08.082. PMid:24274545.
http://dx.doi.org/10.1016/j.carbpol.2013...
demonstrated that chitosan reduced the O-GlcNAc transferase-dependent NF-B's O-GlcNAcylation, inhibiting LPS-induced vascular endothelial inflammatory response. These results demonstrated that chitosan might ameliorate endothelial dysfunction and exhibit an anti-hypertensive effect by reducing inflammation and oxidative stress.

Accordingly, Tao et al. (2021)TAO, W., WANG, G. and WEI, J., 2021. The role of chitosan oligosaccharide in metabolic syndrome: a review of possible mechanisms. Marine Drugs, vol. 19, no. 9, pp. 501. http://dx.doi.org/10.3390/md19090501. PMid:34564163.
http://dx.doi.org/10.3390/md19090501...
mentioned that chitosan has useful impacts on different metabolic syndromes constituents, including hyperglycemia, diabetes mellitus, obesity, dyslipidemia, and hypertension. According to Sharma et al. (2018)SHARMA, M., SHARMA, R. and JAIN, D.K., 2018. Preparation, characterization and evaluation of nebivolol loaded chitosan nanoparticles. Journal of Drug Delivery and Therapeutics, vol. 8, no. 2, pp. 118-122. http://dx.doi.org/10.22270/jddt.v8i2.1730.
http://dx.doi.org/10.22270/jddt.v8i2.173...
, the study's encouraging findings validated the possible use of chitosan in preparing nebivolol-loaded chitosan NPs. Additionally, chitosan-coated polymers are recommended to facilitate oral management by enhancing the drug's solubility (Niaz et al., 2016NIAZ, T., SHABBIR, S., MANZOOR, S., REHMAN, A., RAHMAN, A., NASIR, H. and IMRAN, M., 2016. Antihypertensive nano-ceuticales based on chitosan biopolymer: physico-chemical evaluation and release kinetics. Carbohydrate Polymers, vol. 142, pp. 268-274. http://dx.doi.org/10.1016/j.carbpol.2016.01.047. PMid:26917399.
http://dx.doi.org/10.1016/j.carbpol.2016...
). Like other polymeric nanoparticles (PNPs), Chitosan-based polymers have extended-release capabilities that can boost medicinal efficacy without increasing drug dosage, preventing negative side effects. Also, Chadha et al. (2012)CHADHA, R., BHANDARI, S., KATARIA, D., GUPTA, S. and SINGH JAIN, D., 2012. Exploring the potential of lecithin/chitosan nanoparticles in enhancement of antihypertensive efficacy of hydrochlorothiazide. Journal of Microencapsulation, vol. 29, no. 8, pp. 805-812. http://dx.doi.org/10.3109/02652048.2012.692399. PMid:22681125.
http://dx.doi.org/10.3109/02652048.2012....
presented that chitosan PNPs could treat hypertension caused by deoxycorticosterone acetate salt in rats in vivo. Additionally, Auwal et al. (2018)AUWAL, S.M., ZAREI, M., TAN, C.P., BASRI, M. and SAARI, N., 2018. Enhanced physicochemical stability and efficacy of angiotensin I-converting enzyme (ACE) - inhibitory biopeptides by chitosan nanoparticles optimized using Box-Behnken design. Scientific Reports, vol. 8, no. 1, pp. 10411. http://dx.doi.org/10.1038/s41598-018-28659-5. PMid:29991723.
http://dx.doi.org/10.1038/s41598-018-286...
reported that chitosan PNPs protecting food-based antihypertensive biopeptides against gastrointestinal degradation are a secure and possibly appealing source of nonpharmaceutical treatment of treatment-resistant hypertension. Furthermore, Chinh et al. (2018)CHINH, N.T., TRANG, N.T.T., MAI, T.T., THANH, D.T.M., TRUNG, T.H., TRUNG, T.H., QUAN, L.V., HOA, N.T., MAO, C.V., NGHIA, N.T., HAI, N.T., ANH, T.H., HUNG, T.V., CHOY, J.H., LOI, N.V., RAJESH, B. and HOANG, T., 2018. Polylactic acid/chitosan nanoparticles loading Nifedipine: characterization findings and in vivo investigation in animal. Journal of Nanoscience and Nanotechnology, vol. 18, no. 4, pp. 2294-2303. http://dx.doi.org/10.1166/jnn.2018.14537. PMid:29442895.
http://dx.doi.org/10.1166/jnn.2018.14537...
demonstrated that polylactic acid/chitosan nanoparticles carried the Ca2+ channel blocker nifedipine to inferior animal blood tensions.

In the other study, Auwal et al. (2017)AUWAL, S.M., ZAREI, M., TAN, C.P., BASRI, M. and SAARI, N., 2017. Improved in vivo efficacy of anti-hypertensive biopeptides encapsulated in chitosan nanoparticles fabricated by ionotropic gelation on spontaneously hypertensive rats. Nanomaterials, vol. 7, no. 12, pp. 421. http://dx.doi.org/10.3390/nano7120421. PMid:29207480.
http://dx.doi.org/10.3390/nano7120421...
found that ACE-inhibitory biopeptides stabilized by chitosan nanoparticles can successfully lower blood tension in hypertensive people for a protracted duration. In Table 3, we demonstrated the main findings of some investigations on the valuable effects of chitosan and nano-formulation on hypertension in Table 3.

Table 3
Summary of some studies on the beneficial effects of chitosan and nano-formulation on hypertension.

9. Effects on Molecular and Genetics Perspectives (Crispr-Cas9 Delivery)

Mutations instigate most hereditary metabolic syndromes in genes that code for enzymes; enzyme deficiency or inactivity leads to scarcities of the enzyme’s product or aggregation of material precursors or metabolites (Groop, 2000GROOP, L., 2000. Genetics of the metabolic syndrome. British Journal of Nutrition, vol. 83, suppl. 1, pp. S39-S48. http://dx.doi.org/10.1017/S0007114500000945. PMid:10889791.
http://dx.doi.org/10.1017/S0007114500000...
). Discovering and treating during the early stages of these syndromes focuses on emergency attention and certainty, improving organ function. Recently, gene editing or therapy has been a hopeful approach for treating challenging diseases, including metabolic syndromes. The fruitful delivery of genes is a perilous step for gene therapy. The utmost updated developed approach of gene editing is CRISPR-Cas9 (Clustered regularly interspaced short palindromic repeats- CRISPR associated 9) technology that, a competent gene-editing implemented according to the anti-viral mechanism controlled by some naturally occurring bacteria (Abdelnour et al., 2021ABDELNOUR, S.A., XIE, L., HASSANIN, A.A., ZUO, E. and LU, Y., 2021. The potential of CRISPR/Cas9 gene editing as a treatment strategy for inherited diseases. Frontiers in Cell and Developmental Biology, vol. 9, pp. 699597. http://dx.doi.org/10.3389/fcell.2021.699597. PMid:34977000.
http://dx.doi.org/10.3389/fcell.2021.699...
). This technique could allow for the deletion or insertion of a causative genetic component, paving a probability for the whole cure of the syndrome.

Viral delivery is the main gene editing choice and has been widely applied. However, it exhibits some drawbacks, such as replication competence, transduction efficiency, integration, small insert size, inactivation by the accompaniment path, and restricted host range owing to the necessity of cell division for transduction (Kay et al., 2001KAY, M.A., GLORIOSO, J.C. and NALDINI, L., 2001. Viral vectors for gene therapy: the art of turning infectious agents into vehicles of therapeutics. Nature Medicine, vol. 7, no. 1, pp. 33-40. http://dx.doi.org/10.1038/83324. PMid:11135613.
http://dx.doi.org/10.1038/83324...
). Based on the above drawbacks, searching for a biocompatible and safe delivery system is urgently necessary. In this era of biotechnology, numerous nanoparticles encompass lipid-based, glycolipid, inorganic polymers, gold nanoparticles, alginate, and chitosan nanoparticles are expansively used currently for preclinical and clinical investigations (Mout and Rotello, 2017MOUT, R. and ROTELLO, V.M., 2017. Cytosolic and nuclear delivery of CRISPR/Cas9-ribonucleoprotein for gene editing using arginine functionalized gold nanoparticles. Bio-Protocol, vol. 7, no. 20, e2586. http://dx.doi.org/10.21769/BioProtoc.2586. PMid:29226180.
http://dx.doi.org/10.21769/BioProtoc.258...
; Abdelnour et al., 2021ABDELNOUR, S.A., XIE, L., HASSANIN, A.A., ZUO, E. and LU, Y., 2021. The potential of CRISPR/Cas9 gene editing as a treatment strategy for inherited diseases. Frontiers in Cell and Developmental Biology, vol. 9, pp. 699597. http://dx.doi.org/10.3389/fcell.2021.699597. PMid:34977000.
http://dx.doi.org/10.3389/fcell.2021.699...
). Regarding the potential application of chitosan in the delivery system of gene editing, the study of (Saberi et al., 2009SABERI, M., BJELICA, D., SCHENK, S., IMAMURA, T., BANDYOPADHYAY, G., LI, P., JADHAR, V., VARGEESE, C., WANG, W., BOWMAN, K., ZHANG, Y., POLISKY, B. and OLEFSKY, J.M., 2009. Novel liver-specific TORC2 siRNA corrects hyperglycemia in rodent models of type 2 diabetes. American Journal of Physiology. Endocrinology and Metabolism, vol. 297, no. 5, pp. E1137-E1146. http://dx.doi.org/10.1152/ajpendo.00158.2009. PMid:19706791.
http://dx.doi.org/10.1152/ajpendo.00158....
) reported that siRNA delivery policy includes polyethylene glycol, chitosan lactate, conjugated with glycyrrhetinic acid and contains the CRTC2 gene as a target. This CRTC2-siRNA conjugate system effectively silences the CRTC2 gene that regulates hepatic gluconeogenesis in T2DM (Saberi et al., 2009SABERI, M., BJELICA, D., SCHENK, S., IMAMURA, T., BANDYOPADHYAY, G., LI, P., JADHAR, V., VARGEESE, C., WANG, W., BOWMAN, K., ZHANG, Y., POLISKY, B. and OLEFSKY, J.M., 2009. Novel liver-specific TORC2 siRNA corrects hyperglycemia in rodent models of type 2 diabetes. American Journal of Physiology. Endocrinology and Metabolism, vol. 297, no. 5, pp. E1137-E1146. http://dx.doi.org/10.1152/ajpendo.00158.2009. PMid:19706791.
http://dx.doi.org/10.1152/ajpendo.00158....
). Jean et al. (2012)JEAN, M., ALAMEH, M., JESUS, D., THIBAULT, M., LAVERTU, M., DARRAS, V., NELEA, M., BUSCHMANN, M.D. and MERZOUKI, A., 2012. Chitosan-based therapeutic nanoparticles for combination gene therapy and gene silencing of in vitro cell lines relevant to type 2 diabetes. European Journal of Pharmaceutical Sciences, vol. 45, no. 1-2, pp. 138-149. http://dx.doi.org/10.1016/j.ejps.2011.10.029. PMid:22085632.
http://dx.doi.org/10.1016/j.ejps.2011.10...
researched the silence of DPP-4, an antagonist of incretin, GLP-1 that encourages insulin relief and sustains glucose homeostasis.

The formulated chitosan-DPP-4-siRNA nano-complexes exhibited substantial silence of DPP-4 in cultured situations without seeming cytotoxicity Jean et al. (2012)JEAN, M., ALAMEH, M., JESUS, D., THIBAULT, M., LAVERTU, M., DARRAS, V., NELEA, M., BUSCHMANN, M.D. and MERZOUKI, A., 2012. Chitosan-based therapeutic nanoparticles for combination gene therapy and gene silencing of in vitro cell lines relevant to type 2 diabetes. European Journal of Pharmaceutical Sciences, vol. 45, no. 1-2, pp. 138-149. http://dx.doi.org/10.1016/j.ejps.2011.10.029. PMid:22085632.
http://dx.doi.org/10.1016/j.ejps.2011.10...
. Moreover, Sharma et al. (2021)SHARMA, D., ARORA, S., BANERJEE, A. and SINGH, J., 2021. Improved insulin sensitivity in obese-diabetic mice via chitosan Nanomicelles mediated silencing of pro-inflammatory Adipocytokines. Nanomedicine: Nanotechnology, Biology and Medicine, vol. 33, pp. 102357. http://dx.doi.org/10.1016/j.nano.2020.102357. PMid:33460779.
http://dx.doi.org/10.1016/j.nano.2020.10...
developed nano micelles incorporating chitosan that targets adiponectin, conjugated to the oleic acid and adipose homing peptide to ease the delivery of pADN (plasmid adiponectin) to adipocytes. This nano micelle of chitosan (112 nm) is cationic due to the presence of chitosan that shows a protective modulator for genes against enzymatic degradation with a highly encapsulated rate of around 93%. The outcome revealed betterment of insulin sensitivity for up to 6 weeks with single subcutaneous administration of pADN-chitosan-oleic-AHP in vivo and in vitro using a diabetic rat model (Banerjee et al., 2020BANERJEE, A., SHARMA, D., TRIVEDI, R. and SINGH, J., 2020. Treatment of insulin resistance in obesity-associated type 2 diabetes mellitus through adiponectin gene therapy. International Journal of Pharmaceutics, vol. 583, pp. 119357. http://dx.doi.org/10.1016/j.ijpharm.2020.119357. PMid:32334065.
http://dx.doi.org/10.1016/j.ijpharm.2020...
).

Commercial drugs used as anti-inflammatory mediators have numerous side effects and are inappropriate for long-term usage. A research group by Luo et al. (2018)LUO, Y.L., XU, C.F., LI, H.J., CAO, Z.T., LIU, J., WANG, J.L., DU, X.J., YANG, X.Z., GU, Z. and WANG, J., 2018. Macrophage-specific in vivo gene editing using cationic lipid-assisted polymeric nanoparticles. ACS Nano, vol. 12, no. 2, pp. 994-1005. http://dx.doi.org/10.1021/acsnano.7b07874. PMid:29314827.
http://dx.doi.org/10.1021/acsnano.7b0787...
targeted two such factors, e.g., monocyte chemoattractant protein-1 (MCP-1) and tumor necrosis factor-α (TNF-α) that are pro-inflammatory adipocytokines, existing in adipocytes macrophages (ATMs) and adipocytes and lead to knockdown by applied shRNAs against these features. The chitosan nanomicelle/pDNA polyplexes were subcutaneously treated in an obese-diabetic mice model. The outcome showed decreasing levels of numerous classes of pro-inflammatory cytokines such as IL-6 IL-1, TNF-α, and MCP-1, whereas the insulin-sensitizing adipokine, adiponectin levels were augmented. After the literature screening, limited works incorporate CRISPR-Cas9 (Luo et al., 2018LUO, Y.L., XU, C.F., LI, H.J., CAO, Z.T., LIU, J., WANG, J.L., DU, X.J., YANG, X.Z., GU, Z. and WANG, J., 2018. Macrophage-specific in vivo gene editing using cationic lipid-assisted polymeric nanoparticles. ACS Nano, vol. 12, no. 2, pp. 994-1005. http://dx.doi.org/10.1021/acsnano.7b07874. PMid:29314827.
http://dx.doi.org/10.1021/acsnano.7b0787...
; Banerjee et al., 2020BANERJEE, A., SHARMA, D., TRIVEDI, R. and SINGH, J., 2020. Treatment of insulin resistance in obesity-associated type 2 diabetes mellitus through adiponectin gene therapy. International Journal of Pharmaceutics, vol. 583, pp. 119357. http://dx.doi.org/10.1016/j.ijpharm.2020.119357. PMid:32334065.
http://dx.doi.org/10.1016/j.ijpharm.2020...
; Sharma et al., 2021SHARMA, D., ARORA, S., BANERJEE, A. and SINGH, J., 2021. Improved insulin sensitivity in obese-diabetic mice via chitosan Nanomicelles mediated silencing of pro-inflammatory Adipocytokines. Nanomedicine: Nanotechnology, Biology and Medicine, vol. 33, pp. 102357. http://dx.doi.org/10.1016/j.nano.2020.102357. PMid:33460779.
http://dx.doi.org/10.1016/j.nano.2020.10...
), nanoparticle-based delivery in treating metabolic syndromes. This strategy is indisputably a commanding policy, as per the recent study and it can proficiently silence the specific causative genes related to some metabolic syndromes. In the upcoming investigations, modulations of the transcriptomics of the causative genes were prepared and potentially applied to the miracle molecules, CRISPR-Cas9, delivered by chitosan nanoformulations.

10. The Potential Challenges of Utilizing Chitosan and its Nanoformulations

According to Yadav et al. (2023)YADAV, M., KAUSHIK, B., RAO, G.K., SRIVASTAVA, C.M. and VAYA, D., 2023. Advances and challenges in the use of chitosan and its derivatives in biomedical fields: a review. Carbohydrate Polymer Technologies and Applications, vol. 5, pp. 100323. http://dx.doi.org/10.1016/j.carpta.2023.100323.
http://dx.doi.org/10.1016/j.carpta.2023....
, advances in nanotechnology have created nanocomposites based on chitosan with better qualities. These nanocomposites have excellent thermal, mechanical, conductive, and antibacterial qualities. They are efficient in delivering drugs and aid in the healing of wounds. Biomedical sectors hold great promise for chitosan-based nanocomposites, notwithstanding obstacles related to toxicity and in vivo assessments. In vitro testing and fabrication have been the main topics of previous research, but more work is required to address these issues.

According to Kaur et al. (2023)KAUR, G., GOYAL, J., BEHERA, P.K., DEVI, S., SINGH, S.K., GARG, V. and MITTAL, N., 2023. Unraveling the role of chitosan for nasal drug delivery systems: a review. Carbohydrate Polymer Technologies and Applications, vol. 5, pp. 100316. http://dx.doi.org/10.1016/j.carpta.2023.100316.
http://dx.doi.org/10.1016/j.carpta.2023....
, nanoscience and technology were used more and more in targeted drug administration to increase therapeutic efficacy and safety over the previous 20 years. Biodegradable polymers, such as those found in CS drug delivery systems, provide an alternative to controlled drug release through the nose. These carriers represent a new paradigm in medication delivery because of their ability to treat neurodegenerative illnesses. Nasal drug delivery (NDD) is a non-invasive technique that shows great promise for efficiently administering a wide range of pharmaceuticals, including high molecular peptide and protein therapies and low molecular polar chemicals (Weyers et al., 2022WEYERS, M., PETERSON, B., HAMMAN, J.H. and STEENEKAMP, J.H., 2022. Formulation of chitosan microparticles for enhanced intranasal macromolecular compound delivery: factors that influence particle size during ionic gelation. Gels, vol. 8, no. 11, pp. 686. http://dx.doi.org/10.3390/gels8110686. PMid:36354594.
http://dx.doi.org/10.3390/gels8110686...
). Therefore, the nasal mucosa serves as the main portal through which various therapeutic drugs are directed to specific disease-causing locations that enter the body (Kurono, 2022KURONO, Y., 2022. The mucosal immune system of the upper respiratory tract and recent progress in mucosal vaccines. Auris, Nasus, Larynx, vol. 49, no. 1, pp. 1-10. http://dx.doi.org/10.1016/j.anl.2021.07.003. PMid:34304944.
http://dx.doi.org/10.1016/j.anl.2021.07....
). As a result, in contrast with parenteral and oral administrations, the nasal cavity's high vascularity expedites regional and systemic pharmaceutical absorption via the nasal mucosa, enabling fast therapeutic action (Chavda et al., 2022CHAVDA, V.P., JOGI, G., SHAH, N., ATHALYE, M.N., BAMANIYA, N., VORA, L.K. and PAIVA-SANTOS, A.C., 2022. Advanced particulate carrier-mediated technologies for nasal drug delivery. Journal of Drug Delivery Science and Technology, vol. 74, pp. 103569. http://dx.doi.org/10.1016/j.jddst.2022.103569.
http://dx.doi.org/10.1016/j.jddst.2022.1...
). Additionally, its ability to get past the hepatic first-pass metabolism and blood-brain barrier (BBB) is another important benefit (Nojoki et al., 2022NOJOKI, F., EBRAHIMI-HOSSEINZADEH, B., HATAMIAN-ZARMI, A., KHODAGHOLI, F. and KHEZRI, K., 2022. Design and development of chitosan-insulin-transfersomes (Transfersulin) as effective intranasal nanovesicles for the treatment of Alzheimer’s disease: in vitro, in vivo, and ex vivo evaluations. Biomedicine and Pharmacotherapy, vol. 153, pp. 113450. http://dx.doi.org/10.1016/j.biopha.2022.113450. PMid:36076565.
http://dx.doi.org/10.1016/j.biopha.2022....
; Khatri et al., 2023KHATRI, D.K., PREETI, K., TONAPE, S., BHATTACHARJEE, S., PATEL, M., SHAH, S., SINGH, P.K., SRIVASTAVA, S., GUGULOTHU, D., VORA, L. and SINGH, S.B., 2023. Nanotechnological advances for nose to brain delivery of therapeutics to improve the Parkinson therapy. Current Neuropharmacology, vol. 21, no. 3, pp. 493-516. http://dx.doi.org/10.2174/1570159X20666220507022701. PMid:35524671.
http://dx.doi.org/10.2174/1570159X206662...
). Because of this, lower pharmaceutical doses may be needed to produce more beneficial effects with fewer negative effects.

By necrotic biofilm construction, decreasing its significant components, and preventing microbial proliferation, El-Naggar et al. (2023)EL-NAGGAR, N.E.A., ELTARAHONY, M., HAFEZ, E.E. and BASHIR, S.I., 2023. Green fabrication of chitosan nanoparticles using Lavendula angustifolia, optimization, characterization and in-vitro antibiofilm activity. Scientific Reports, vol. 13, no. 1, pp. 11127. http://dx.doi.org/10.1038/s41598-023-37660-6. PMid:37429892.
http://dx.doi.org/10.1038/s41598-023-376...
suggested that the encouraging findings of the research study in biofilm inhibition motivate usage as a natural, safe, and biocompatible anti-adherent covering in antibiofouling membranes, medical bandages/tissues, and packaging for food. According to Virmani et al. (2023)VIRMANI, T., KUMAR, G., SHARMA, A., PATHAK, K., AKHTAR, M.S., AFZAL, O. and ALTAMIMI, A.S., 2023. Amelioration of cancer employing chitosan, its derivatives, and chitosan-based nanoparticles: recent updates. Polymers, vol. 15, no. 13, pp. 2928. http://dx.doi.org/10.3390/polym15132928. PMid:37447573.
http://dx.doi.org/10.3390/polym15132928...
, chitosan and its modified derivatives are employed in pharmaceuticals to produce nanoparticles, medication delivery, and cancer site targeting. These nanoparticles are desirable for many anticancer medications due to their increased potency, efficacy, cytotoxicity, and biocompatibility.

Chitosan is widely used in biotechnology, medicine, and agriculture due to its unique properties. However, its insoluble nature and poor mechanical properties limit its use in biomedical fields. Modifications can increase solubility, creating new derivatives with enhanced properties. Chitosan is also used in wound dressings, speeding up healing and protecting against infection. On the other hand, chitosan, a versatile drug delivery system, has been studied extensively over the past 20 years. However, cytotoxic impacts are still present, and further research is needed to reduce toxicity. Despite numerous studies, there are still few uses for chitosan in the medical field. Further investigation into drug delivery methods, toxicology, and security concerns is crucial.

11. Conclusions

Chitosan is one of the utmost discovered bio-based polymers. According to WHO reports, chitosan is commonly approved and recognized as safe eminence as a food element. Due to its biodegradability and biocompatibility, chitosan has a variety of multifaceted uses, with a distinctive prominence on therapeutic uses and drug delivery schemes. Moreover, the new form of chitosan nanoparticles or chitosan alone has numerous proposes in non-parenteral drug management for metabolic syndromes such as insulin resistance, obesity, diabetes mellitus, dyslipidemia, hyperglycemia, and hypertension due to their physical structures and absence of toxicity. In addition, chitosan nanoparticles have been considered in the pitch of nanomedicine for the formation of novel therapeutic drug schemes due to their enhancement of the bioavailability of drugs and their inferior toxicity, sensitivity, and specificity. Recently, the chitosan nanoparticle-based delivery of CRISPR-Cas9 has been applied in treating metabolic syndromes. Furthermore, the wide range of chitosan NPs has revealed therapeutic likely in various metabolic syndromes.

References

  • ABD EL-HACK, M.E., EL-SAADONY, M.T., SHAFI, M.E., ZABERMAWI, N.M., ARIF, M., BATIHA, G.E., KHAFAGA, A.F., ABD EL-HAKIM, Y.M. and AL-SAGHEER, A.A., 2020. Antimicrobial and antioxidant properties of chitosan and its derivatives and their applications: a review. International Journal of Biological Macromolecules, vol. 164, pp. 2726-2744. http://dx.doi.org/10.1016/j.ijbiomac.2020.08.153 PMid:32841671.
    » http://dx.doi.org/10.1016/j.ijbiomac.2020.08.153
  • ABD EL-HAMEED, A.M., 2020. Polydatin-loaded chitosan nanoparticles ameliorates early diabetic nephropathy by attenuating oxidative stress and inflammatory responses in streptozotocin-induced diabetic rat. Journal of Diabetes and Metabolic Disorders, vol. 19, no. 2, pp. 1599-1607. http://dx.doi.org/10.1007/s40200-020-00699-7 PMid:33520856.
    » http://dx.doi.org/10.1007/s40200-020-00699-7
  • ABD-ELHAKEEM, M.A., FARAG, N. and MAURICE, M., 2016. Effects of dietary chitosan nanoparticles on serum lipid concentration in hyperlipidemic rats induced by a high-fat diet. Egyptian Journal of Pure and Applied Science, vol. 54, no. 4, pp. 17-21. http://dx.doi.org/10.21608/ejaps.2016.184579
    » http://dx.doi.org/10.21608/ejaps.2016.184579
  • ABDELNOUR, S.A., XIE, L., HASSANIN, A.A., ZUO, E. and LU, Y., 2021. The potential of CRISPR/Cas9 gene editing as a treatment strategy for inherited diseases. Frontiers in Cell and Developmental Biology, vol. 9, pp. 699597. http://dx.doi.org/10.3389/fcell.2021.699597 PMid:34977000.
    » http://dx.doi.org/10.3389/fcell.2021.699597
  • ABDULLAH, S.S., MASOOD, S., ZANEB, H., RABBANI, I., AKBAR, J., KUTHU, Z.H., MASOOD, A. and VARGAS-BELLO-PÉREZ, E., 2024. Effects of copper nanoparticles on performance, muscle and bone characteristics and serum metabolites in broilers. Brazilian Journal of Biology = Revista Brasileira de Biologia, vol. 84, e261578. http://dx.doi.org/10.1590/1519-6984.261578 PMid:35730812.
    » http://dx.doi.org/10.1590/1519-6984.261578
  • ADETUNJI, C.O., MICHAEL, O.S., RATHEE, S., SINGH, K.R.B., AJAYI, O.O., ADETUNJI, J.B., OJHA, A., SINGH, J. and SINGH, R.P., 2022. Potentialities of nanomaterials for the management and treatment of metabolic syndrome: a new insight. Materials Today Advances, vol. 13, pp. 100198. http://dx.doi.org/10.1016/j.mtadv.2021.100198
    » http://dx.doi.org/10.1016/j.mtadv.2021.100198
  • AGNIHOTRI, S.A., MALLIKARJUNA, N.N. and AMINABHAVI, T.M., 2004. Recent advances on chitosan-based micro-and nanoparticles in drug delivery. Journal of Controlled Release, vol. 100, no. 1, pp. 5-28. http://dx.doi.org/10.1016/j.jconrel.2004.08.010 PMid:15491807.
    » http://dx.doi.org/10.1016/j.jconrel.2004.08.010
  • AHSAN, S.M., THOMAS, M., REDDY, K.K., SOORAPARAJU, S.G., ASTHANA, A. and BHATNAGAR, I., 2018. Chitosan as biomaterial in drug delivery and tissue engineering. International Journal of Biological Macromolecules, vol. 110, pp. 97-109. http://dx.doi.org/10.1016/j.ijbiomac.2017.08.140 PMid:28866015.
    » http://dx.doi.org/10.1016/j.ijbiomac.2017.08.140
  • ALAQEEL, N.K., 2024. Antioxidants from different citrus peels provide protection against cancer. Brazilian Journal of Biology = Revista Brasileira de Biologia, vol. 84, e271619. http://dx.doi.org/10.1590/1519-6984.271619 PMid:37436265.
    » http://dx.doi.org/10.1590/1519-6984.271619
  • ALARCÓN-PAYÁN, D.A., KOYANI, R.D. and VAZQUEZ-DUHALT, R., 2017. Chitosan-based biocatalytic nanoparticles for pollutant removal from wastewater. Enzyme and Microbial Technology, vol. 100, pp. 71-78. http://dx.doi.org/10.1016/j.enzmictec.2017.02.008 PMid:28284314.
    » http://dx.doi.org/10.1016/j.enzmictec.2017.02.008
  • AMATO, A., MIGNECO, L.M., MARTINELLI, A., PIETRELLI, L., PIOZZI, A. and FRANCOLINI, I., 2018. Antimicrobial activity of catechol functionalized-chitosan versus Staphylococcus epidermidis Carbohydrate Polymers, vol. 179, pp. 273-281. http://dx.doi.org/10.1016/j.carbpol.2017.09.073 PMid:29111051.
    » http://dx.doi.org/10.1016/j.carbpol.2017.09.073
  • AUWAL, S.M., ZAREI, M., TAN, C.P., BASRI, M. and SAARI, N., 2017. Improved in vivo efficacy of anti-hypertensive biopeptides encapsulated in chitosan nanoparticles fabricated by ionotropic gelation on spontaneously hypertensive rats. Nanomaterials, vol. 7, no. 12, pp. 421. http://dx.doi.org/10.3390/nano7120421 PMid:29207480.
    » http://dx.doi.org/10.3390/nano7120421
  • AUWAL, S.M., ZAREI, M., TAN, C.P., BASRI, M. and SAARI, N., 2018. Enhanced physicochemical stability and efficacy of angiotensin I-converting enzyme (ACE) - inhibitory biopeptides by chitosan nanoparticles optimized using Box-Behnken design. Scientific Reports, vol. 8, no. 1, pp. 10411. http://dx.doi.org/10.1038/s41598-018-28659-5 PMid:29991723.
    » http://dx.doi.org/10.1038/s41598-018-28659-5
  • AZAD, A.K., SERMSINTHAM, N., CHANDRKRACHANG, S. and STEVENS, W.F., 2004. Chitosan membrane as a wound‐healing dressing: characterization and clinical application. Journal of Biomedical Materials Research Part B: Applied Biomaterials, vol. 69B, no. 2, pp. 216-222. http://dx.doi.org/10.1002/jbm.b.30000 PMid:15116411.
    » http://dx.doi.org/10.1002/jbm.b.30000
  • BAHAR, B., O’DOHERTY, J.V., O’DOHERTY, A.M. and SWEENEY, T., 2013. Chito-oligosaccharide inhibits the de-methylation of a ‘CpG’ Island within the leptin (LEP) promoter during adipogenesis of 3T3-L1 cells. PLoS One, vol. 8, no. 3, e60011. http://dx.doi.org/10.1371/journal.pone.0060011 PMid:23544120.
    » http://dx.doi.org/10.1371/journal.pone.0060011
  • BAI, Y., ZHENG, J., YUAN, X., JIAO, S., FENG, C., DU, Y., LIU, H. and ZHENG, L., 2018. Chitosan oligosaccharides improve glucolipid metabolism disorder in liver by suppression of obesity-related inflammation and restoration of peroxisome proliferator-activated receptor gamma (PPARγ). Marine Drugs, vol. 16, no. 11, pp. 455. http://dx.doi.org/10.3390/md16110455 PMid:30463189.
    » http://dx.doi.org/10.3390/md16110455
  • BANERJEE, A., SHARMA, D., TRIVEDI, R. and SINGH, J., 2020. Treatment of insulin resistance in obesity-associated type 2 diabetes mellitus through adiponectin gene therapy. International Journal of Pharmaceutics, vol. 583, pp. 119357. http://dx.doi.org/10.1016/j.ijpharm.2020.119357 PMid:32334065.
    » http://dx.doi.org/10.1016/j.ijpharm.2020.119357
  • BASHIR, S.M., AHMED RATHER, G., PATRÍCIO, A., HAQ, Z., SHEIKH, A.A., SHAH, M.Z.U.H., SINGH, H., KHAN, A.A., IMTIYAZ, S., AHMAD, S.B., NABI, S., RAKHSHAN, R., HASSAN, S. and FONTE, P., 2022. Chitosan nanoparticles: a versatile platform for biomedical applications. Materials, vol. 15, no. 19, pp. 6521. http://dx.doi.org/10.3390/ma15196521 PMid:36233864.
    » http://dx.doi.org/10.3390/ma15196521
  • BENEDIKTSDÓTTIR, B.E., BALDURSSON, Ó. and MÁSSON, M., 2014. Challenges in evaluation of chitosan and trimethylated chitosan (TMC) as mucosal permeation enhancers: from synthesis to in vitro application. Journal of Controlled Release, vol. 173, pp. 18-31. http://dx.doi.org/10.1016/j.jconrel.2013.10.022 PMid:24511609.
    » http://dx.doi.org/10.1016/j.jconrel.2013.10.022
  • BENEDIKTSDÓTTIR, B.E., GAWARE, V.S., RÚNARSSON, Ö.V., JÓNSDÓTTIR, S., JENSEN, K.J. and MÁSSON, M., 2011. Synthesis of N,N,N-trimethyl chitosan homopolymer and highly substituted N-alkyl-N,N-dimethyl chitosan derivatives with the aid of di-tert-butyldimethylsilyl chitosan. Carbohydrate Polymers, vol. 86, no. 4, pp. 1451-1460. http://dx.doi.org/10.1016/j.carbpol.2011.06.007
    » http://dx.doi.org/10.1016/j.carbpol.2011.06.007
  • BERGER, N.A., 2014. Obesity and cancer pathogenesis. Annals of the New York Academy of Sciences, vol. 1311, no. 1, pp. 57-76. http://dx.doi.org/10.1111/nyas.12416 PMid:24725147.
    » http://dx.doi.org/10.1111/nyas.12416
  • CALVO, P., REMUÑÁN‐LÓPEZ, C., VILA‐JATO, J.L. and ALONSO, M.J., 1997. Novel hydrophilic chitosan‐polyethylene oxide nanoparticles as protein carriers. Journal of Applied Polymer Science, vol. 63, no. 1, pp. 125-132. http://dx.doi.org/10.1002/(SICI)1097-4628(19970103)63:1<125::AID-APP13>3.0.CO;2-4
    » http://dx.doi.org/10.1002/(SICI)1097-4628(19970103)63:1<125::AID-APP13>3.0.CO;2-4
  • CANI, P.D. and JORDAN, B.F., 2018. Gut microbiota-mediated inflammation in obesity: a link with gastrointestinal cancer. Nature Reviews. Gastroenterology & Hepatology, vol. 15, no. 11, pp. 671-682. http://dx.doi.org/10.1038/s41575-018-0025-6 PMid:29844585.
    » http://dx.doi.org/10.1038/s41575-018-0025-6
  • CAREY, F.A. and SUNDBERG, R.J., 2007. Advanced organic chemistry: part B: reactions and synthesis Boston: Springer, vol. 3.
  • CHADHA, R., BHANDARI, S., KATARIA, D., GUPTA, S. and SINGH JAIN, D., 2012. Exploring the potential of lecithin/chitosan nanoparticles in enhancement of antihypertensive efficacy of hydrochlorothiazide. Journal of Microencapsulation, vol. 29, no. 8, pp. 805-812. http://dx.doi.org/10.3109/02652048.2012.692399 PMid:22681125.
    » http://dx.doi.org/10.3109/02652048.2012.692399
  • CHAN, H.K. and KWOK, P.C.L., 2011. Production methods for nanodrug particles using the bottom-up approach. Advanced Drug Delivery Reviews, vol. 63, no. 6, pp. 406-416. http://dx.doi.org/10.1016/j.addr.2011.03.011 PMid:21457742.
    » http://dx.doi.org/10.1016/j.addr.2011.03.011
  • CHANDRASEKARAN, M., KIM, K.D. and CHUN, S.C., 2020. Antibacterial activity of chitosan nanoparticles: a review. Processes, vol. 8, no. 9, pp. 1173. http://dx.doi.org/10.3390/pr8091173
    » http://dx.doi.org/10.3390/pr8091173
  • CHAVDA, V.P., JOGI, G., SHAH, N., ATHALYE, M.N., BAMANIYA, N., VORA, L.K. and PAIVA-SANTOS, A.C., 2022. Advanced particulate carrier-mediated technologies for nasal drug delivery. Journal of Drug Delivery Science and Technology, vol. 74, pp. 103569. http://dx.doi.org/10.1016/j.jddst.2022.103569
    » http://dx.doi.org/10.1016/j.jddst.2022.103569
  • CHEN, K., ZHANG, J., BEERAKA, N.M., TANG, C., BABAYEVA, Y.V., SINELNIKOV, M.Y., ZHANG, X., ZHANG, J., LIU, J., RESHETOV, I.V., SUKOCHEVA, O.A., LU, P. and FAN, R., 2022. Advances in the prevention and treatment of obesity-driven effects in breast cancers. Frontiers in Oncology, vol. 12, pp. 820968. http://dx.doi.org/10.3389/fonc.2022.820968 PMid:35814391.
    » http://dx.doi.org/10.3389/fonc.2022.820968
  • CHEN, L., WANG, C. and WU, Y., 2020. Cholesterol (Blood lipid) lowering potential of Rosuvastatin chitosan nanoparticles for atherosclerosis: preclinical study in rabbit model. Acta Biochimica Polonica, vol. 67, no. 4, pp. 495-499. http://dx.doi.org/10.18388/abp.2020_5186 PMid:33090754.
    » http://dx.doi.org/10.18388/abp.2020_5186
  • CHINH, N.T., TRANG, N.T.T., MAI, T.T., THANH, D.T.M., TRUNG, T.H., TRUNG, T.H., QUAN, L.V., HOA, N.T., MAO, C.V., NGHIA, N.T., HAI, N.T., ANH, T.H., HUNG, T.V., CHOY, J.H., LOI, N.V., RAJESH, B. and HOANG, T., 2018. Polylactic acid/chitosan nanoparticles loading Nifedipine: characterization findings and in vivo investigation in animal. Journal of Nanoscience and Nanotechnology, vol. 18, no. 4, pp. 2294-2303. http://dx.doi.org/10.1166/jnn.2018.14537 PMid:29442895.
    » http://dx.doi.org/10.1166/jnn.2018.14537
  • CHIU, C.Y., CHANG, T.C., LIU, S.H. and CHIANG, M.T., 2017. The regulatory effects of fish oil and chitosan on hepatic lipogenic signals in high-fat diet-induced obese rats. Yao Wu Shi Pin Fen Xi, vol. 25, no. 4, pp. 919-930. http://dx.doi.org/10.1016/j.jfda.2016.11.015 PMid:28987369.
    » http://dx.doi.org/10.1016/j.jfda.2016.11.015
  • CHO, E.J., RAHMAN, A., KIM, S.W., BAEK, Y.M., HWANG, H.J., OH, J.Y., HWANG, H.S., LEE, S.H. and YUN, J.W., 2008. Chitosan oligosaccharides inhibit adipogenesis in 3T3-L1 adipocytes. Journal of Microbiology and Biotechnology, vol. 18, no. 1, pp. 80-87. PMid:18239421.
  • CHOU, C.-K., CHEN, S.-M., LI, Y.-C., HUANG, T.-C. and LEE, J.-A., 2015. Low-molecular-weight chitosan scavenges methylglyoxal and N ε-(carboxyethyl)lysine, the major factors contributing to the pathogenesis of nephropathy. SpringerPlus, vol. 4, no. 1, pp. 312. http://dx.doi.org/10.1186/s40064-015-1106-4 PMid:25674489.
    » http://dx.doi.org/10.1186/s40064-015-1106-4
  • CHUANG, E.Y., LIN, K.J., SU, F.Y., CHEN, H.L., MAITI, B., HO, Y.C., YEN, T.C., PANDA, N. and SUNG, H.W., 2013. Calcium depletion-mediated protease inhibition and apical-junctional-complex disassembly via an EGTA-conjugated carrier for oral insulin delivery. Journal of Controlled Release, vol. 169, no. 3, pp. 296-305. http://dx.doi.org/10.1016/j.jconrel.2012.11.011 PMid:23195534.
    » http://dx.doi.org/10.1016/j.jconrel.2012.11.011
  • COLONNA, C., CONTI, B., PERUGINI, P., PAVANETTO, F., MODENA, T., DORATI, R. and GENTA, I., 2007. Chitosan glutamate nanoparticles for protein delivery: development and effect on prolidase stability. Journal of Microencapsulation, vol. 24, no. 6, pp. 553-564. http://dx.doi.org/10.1080/02652040701449608 PMid:17654175.
    » http://dx.doi.org/10.1080/02652040701449608
  • CORRADINI, E., MOURA, M. and MATTOSO, L., 2010. A preliminary study of the incorparation of NPK fertilizer into chitosan nanoparticles. Express Polymer Letters, vol. 4, no. 8, pp. 509-515. http://dx.doi.org/10.3144/expresspolymlett.2010.64
    » http://dx.doi.org/10.3144/expresspolymlett.2010.64
  • CROISIER, F. and JÉRÔME, C., 2013. Chitosan-based biomaterials for tissue engineering. European Polymer Journal, vol. 49, no. 4, pp. 780-792. http://dx.doi.org/10.1016/j.eurpolymj.2012.12.009
    » http://dx.doi.org/10.1016/j.eurpolymj.2012.12.009
  • CZECH, M.P., 2017. Insulin action and resistance in obesity and type 2 diabetes. Nature Medicine, vol. 23, no. 7, pp. 804-814. http://dx.doi.org/10.1038/nm.4350 PMid:28697184.
    » http://dx.doi.org/10.1038/nm.4350
  • DAS, L., DAS, P., BHOWAL, A. and BHATTACHARIEE, C., 2020. Synthesis of hybrid hydrogel nano-polymer composite using Graphene oxide, Chitosan and PVA and its application in waste water treatment. Environmental Technology & Innovation, vol. 18, pp. 100664. http://dx.doi.org/10.1016/j.eti.2020.100664
    » http://dx.doi.org/10.1016/j.eti.2020.100664
  • DEVARAJ, S., SINGH, U. and JIALAL, I., 2009. Human C-reactive protein and the metabolic syndrome. Current Opinion in Lipidology, vol. 20, no. 3, pp. 182-189. http://dx.doi.org/10.1097/MOL.0b013e32832ac03e PMid:19369869.
    » http://dx.doi.org/10.1097/MOL.0b013e32832ac03e
  • DUTTA, P.K., DUTTA, J. and TRIPATHI, V.S., 2004 [viewed 15 November 2023]. Chitin and chitosan: chemistry, properties and applications. Journal of Scientific & Industrial Research [online], vol. 63, pp. 20-31. Available from: http://nopr.niscpr.res.in/handle/123456789/5397
    » http://nopr.niscpr.res.in/handle/123456789/5397
  • ECKEL, R.H., GRUNDY, S.M. and ZIMMET, P.Z., 2005. The metabolic syndrome. Lancet, vol. 365, no. 9468, pp. 1415-1428. http://dx.doi.org/10.1016/S0140-6736(05)66378-7 PMid:15836891.
    » http://dx.doi.org/10.1016/S0140-6736(05)66378-7
  • EGAN, Á.M., O’DOHERTY, J.V., VIGORS, S. and SWEENEY, T., 2016. Prawn shell chitosan exhibits anti-obesogenic potential through alterations to appetite, affecting feeding behaviour and satiety signals in vivo. PLoS One, vol. 11, no. 2, e0149820. http://dx.doi.org/10.1371/journal.pone.0149820 PMid:26901760.
    » http://dx.doi.org/10.1371/journal.pone.0149820
  • EGAN, Á.M., SWEENEY, T., HAYES, M. and O’DOHERTY, J.V., 2015. Prawn shell chitosan has anti-obesogenic properties, influencing both nutrient digestibility and microbial populations in a pig model. PLoS One, vol. 10, no. 12, e0144127. http://dx.doi.org/10.1371/journal.pone.0144127 PMid:26636332.
    » http://dx.doi.org/10.1371/journal.pone.0144127
  • EL-NAGGAR, N.E.A., ELTARAHONY, M., HAFEZ, E.E. and BASHIR, S.I., 2023. Green fabrication of chitosan nanoparticles using Lavendula angustifolia, optimization, characterization and in-vitro antibiofilm activity. Scientific Reports, vol. 13, no. 1, pp. 11127. http://dx.doi.org/10.1038/s41598-023-37660-6 PMid:37429892.
    » http://dx.doi.org/10.1038/s41598-023-37660-6
  • ELSAYED, A., AL-REMAWI, M., FAROUK, A. and BADWAN, A., 2010. Insulin-chitosan polyelectrolyte _anocomplexes: preparation, characterization and stabilization of insulin. Sudan Journal of Medical Sciences, vol. 5, no. 2, pp. 99-109. http://dx.doi.org/10.4314/sjms.v5i2.57799
    » http://dx.doi.org/10.4314/sjms.v5i2.57799
  • EL-SHABOURI, M.H., 2002. Positively charged nanoparticles for improving the oral bioavailability of cyclosporin-A. International Journal of Pharmaceutics, vol. 249, no. 1-2, pp. 101-108. http://dx.doi.org/10.1016/S0378-5173(02)00461-1 PMid:12433438.
    » http://dx.doi.org/10.1016/S0378-5173(02)00461-1
  • ERBACHER, P., ZOU, S., BETTINGER, T., STEFFAN, A.M. and REMY, J.S., 1998. Chitosan-based vector/DNA complexes for gene delivery: biophysical characteristics and transfection ability. Pharmaceutical Research, vol. 15, no. 9, pp. 1332-1339. http://dx.doi.org/10.1023/A:1011981000671 PMid:9755882.
    » http://dx.doi.org/10.1023/A:1011981000671
  • FAHED, G., AOUN, L., BOU ZERDAN, M., ALLAM, S., BOU ZERDAN, M., BOUFERRAA, Y. and ASSI, H.I., 2022. Metabolic syndrome: updates on pathophysiology and management in 2021. International Journal of Molecular Sciences, vol. 23, no. 2, pp. 786. http://dx.doi.org/10.3390/ijms23020786 PMid:35054972.
    » http://dx.doi.org/10.3390/ijms23020786
  • FANG, X., ZHANG, J., ROMAN, R.J. and FAN, F., 2022. From 1901 to 2022, how far are we from truly understanding the pathogenesis of age-related dementia? GeroScience, vol. 44, no. 3, pp. 1879-1883. http://dx.doi.org/10.1007/s11357-022-00591-7 PMid:35585301.
    » http://dx.doi.org/10.1007/s11357-022-00591-7
  • FINI, A. and ORIENTI, I., 2003. The role of chitosan in drug delivery: current and potential applications. American Journal of Drug Delivery, vol. 1, no. 1, pp. 43-59. http://dx.doi.org/10.2165/00137696-200301010-00004
    » http://dx.doi.org/10.2165/00137696-200301010-00004
  • FUCHS, F.D. and WHELTON, P.K., 2020. High blood pressure and cardiovascular disease. Hypertension, vol. 75, no. 2, pp. 285-292. http://dx.doi.org/10.1161/HYPERTENSIONAHA.119.14240 PMid:31865786.
    » http://dx.doi.org/10.1161/HYPERTENSIONAHA.119.14240
  • FURUIKE, T., KOMOTO, D., HASHIMOTO, H. and TAMURA, H., 2017. Preparation of chitosan hydrogel and its solubility in organic acids. International Journal of Biological Macromolecules, vol. 104, no. Pt B, pp. 1620-1625. http://dx.doi.org/10.1016/j.ijbiomac.2017.02.099 PMid:28258006.
    » http://dx.doi.org/10.1016/j.ijbiomac.2017.02.099
  • FURUKAWA, S., FUJITA, T., SHIMABUKURO, M., IWAKI, M., YAMADA, Y., NAKAJIMA, Y., NAKAYAMA, O., MAKISHIMA, M., MATSUDA, M. and SHIMOMURA, I., 2004. Increased oxidative stress in obesity and its impact on metabolic syndrome. The Journal of Clinical Investigation, vol. 114, no. 12, pp. 1752-1761. http://dx.doi.org/10.1172/JCI21625 PMid:15599400.
    » http://dx.doi.org/10.1172/JCI21625
  • GALLEGO, I., VILLATE-BEITIA, I., MARTÍNEZ-NAVARRETE, G., MENÉNDEZ, M., LÓPEZ-MÉNDEZ, T., SOTO-SÁNCHEZ, C., ZÁRATE, J., PURAS, G., FERNÁNDEZ, E. and PEDRAZ, J.L., 2019. Non-viral vectors based on cationic niosomes and minicircle DNA technology enhance gene delivery efficiency for biomedical applications in retinal disorders. Nanomedicine: Nanotechnology, Biology, and Medicine, vol. 17, pp. 308-318. http://dx.doi.org/10.1016/j.nano.2018.12.018 PMid:30790710.
    » http://dx.doi.org/10.1016/j.nano.2018.12.018
  • GLOBAL BURDEN OF DISEASE COLLABORATIVE NETWORK - GBD, 2017. Global Burden of Disease Study 2015 (GBD 2015) obesity and overweight prevalence 1980-2015 Seattle: Institute for Health Metrics and Evaluation.
  • GRENHA, A., GOMES, M.E., RODRIGUES, M., SANTO, V.E., MANO, J.F., NEVES, N.M. and REIS, R.L., 2010. Development of new chitosan/carrageenan nanoparticles for drug delivery applications. Journal of Biomedical Materials Research Part A, vol. 92A, no. 4, pp. 1265-1272. http://dx.doi.org/10.1002/jbm.a.32466 PMid:19322874.
    » http://dx.doi.org/10.1002/jbm.a.32466
  • GROOP, L., 2000. Genetics of the metabolic syndrome. British Journal of Nutrition, vol. 83, suppl. 1, pp. S39-S48. http://dx.doi.org/10.1017/S0007114500000945 PMid:10889791.
    » http://dx.doi.org/10.1017/S0007114500000945
  • GUAN, G., AZAD, M.A.K., LIN, Y., KIM, S.W., TIAN, Y., LIU, G. and WANG, H., 2019. Biological effects and applications of chitosan and chito-oligosaccharides. Frontiers in Physiology, vol. 10, pp. 516. http://dx.doi.org/10.3389/fphys.2019.00516 PMid:31133871.
    » http://dx.doi.org/10.3389/fphys.2019.00516
  • HE, N., WANG, S., LV, Z., ZHAO, W. and LI, S., 2020. Low molecular weight chitosan oligosaccharides (LMW-COSs) prevent obesity-related metabolic abnormalities in association with the modification of gut microbiota in high-fat diet (HFD)-fed mice. Food & Function, vol. 11, no. 11, pp. 9947-9959. http://dx.doi.org/10.1039/D0FO01871F PMid:33108433.
    » http://dx.doi.org/10.1039/D0FO01871F
  • HERDIANA, Y., HUSNI, P., NURHASANAH, S., SHAMSUDDIN, S. and WATHONI, N., 2023. Chitosan-based nano systems for natural antioxidants in breast cancer therapy. Polymers, vol. 15, no. 13, pp. 2953. http://dx.doi.org/10.3390/polym15132953 PMid:37447598.
    » http://dx.doi.org/10.3390/polym15132953
  • HONG, S.P., KIM, M.H., OH, S.W., HAN, C.K. and KIM, Y.H., 1998. ACE inhibitory and antihypertensive effect of chitosan oligosaccharides in SHR. Korean Journal of Food Science Technology, vol. 30, no. 6, pp. 1476-1479.
  • HUANG, L., CHEN, J., CAO, P., PAN, H., DING, C., XIAO, T., ZHANG, P., GUO, J. and SU, Z., 2015. Anti-obese effect of glucosamine and chitosan oligosaccharide in high-fat diet-induced obese rats. Marine Drugs, vol. 13, no. 5, pp. 2732-2756. http://dx.doi.org/10.3390/md13052732 PMid:25942093.
    » http://dx.doi.org/10.3390/md13052732
  • HUANG, P.L., 2009. A comprehensive definition for metabolic syndrome. Disease Models & Mechanisms, vol. 2, no. 5-6, pp. 231-237. http://dx.doi.org/10.1242/dmm.001180 PMid:19407331.
    » http://dx.doi.org/10.1242/dmm.001180
  • HUDISH, L.I., REUSCH, J.E. and SUSSEL, L., 2019. β Cell dysfunction during progression of metabolic syndrome to type 2 diabetes. The Journal of Clinical Investigation, vol. 129, no. 10, pp. 4001-4008. http://dx.doi.org/10.1172/JCI129188 PMid:31424428.
    » http://dx.doi.org/10.1172/JCI129188
  • HUO, M., ZHANG, Y., ZHOU, J., ZOU, A., YU, D., WU, Y., LI, J. and LI, H., 2010. Synthesis and characterization of low-toxic amphiphilic chitosan derivatives and their application as micelle carrier for antitumor drug. International Journal of Pharmaceutics, vol. 394, no. 1-2, pp. 162-173. http://dx.doi.org/10.1016/j.ijpharm.2010.05.001 PMid:20457237.
    » http://dx.doi.org/10.1016/j.ijpharm.2010.05.001
  • IADECOLA, C. and GOTTESMAN, R.F., 2019. Neurovascular and cognitive dysfunction in hypertension: epidemiology, pathobiology, and treatment. Circulation Research, vol. 124, no. 7, pp. 1025-1044. http://dx.doi.org/10.1161/CIRCRESAHA.118.313260 PMid:30920929.
    » http://dx.doi.org/10.1161/CIRCRESAHA.118.313260
  • ISLAM, M.M., SHAHRUZZAMAN, M., BISWAS, S., SAKIB, M.N. and RASHID, T.U., 2020. Chitosan based bioactive materials in tissue engineering applications-a review. Bioactive Materials, vol. 5, no. 1, pp. 164-183. http://dx.doi.org/10.1016/j.bioactmat.2020.01.012 PMid:32083230.
    » http://dx.doi.org/10.1016/j.bioactmat.2020.01.012
  • JAISWAL, S., DUTTA, P.K., KUMAR, S. and CHAWLA, R., 2021. Chitosan modified by organo-functionalities as an efficient nanoplatform for anti-cancer drug delivery process. Journal of Drug Delivery Science and Technology, vol. 62, pp. 102407. http://dx.doi.org/10.1016/j.jddst.2021.102407
    » http://dx.doi.org/10.1016/j.jddst.2021.102407
  • JAMES, D.E., STÖCKLI, J. and BIRNBAUM, M.J., 2021. The aetiology and molecular landscape of insulin resistance. Nature Reviews Molecular Cell Biology, vol. 22, no. 11, pp. 751-771. http://dx.doi.org/10.1038/s41580-021-00390-6 PMid:34285405.
    » http://dx.doi.org/10.1038/s41580-021-00390-6
  • JEAN, M., ALAMEH, M., JESUS, D., THIBAULT, M., LAVERTU, M., DARRAS, V., NELEA, M., BUSCHMANN, M.D. and MERZOUKI, A., 2012. Chitosan-based therapeutic nanoparticles for combination gene therapy and gene silencing of in vitro cell lines relevant to type 2 diabetes. European Journal of Pharmaceutical Sciences, vol. 45, no. 1-2, pp. 138-149. http://dx.doi.org/10.1016/j.ejps.2011.10.029 PMid:22085632.
    » http://dx.doi.org/10.1016/j.ejps.2011.10.029
  • JO, S.H., HA, K.S., MOON, K.S., KIM, J.G., OH, C.G., KIM, Y.C., APOSTOLIDIS, E. and KWON, Y.I., 2013. Molecular weight dependent glucose lowering effect of low molecular weight chitosan oligosaccharide (GO2KA1) on postprandial blood glucose level in SD rats model. International Journal of Molecular Sciences, vol. 14, no. 7, pp. 14214-14224. http://dx.doi.org/10.3390/ijms140714214 PMid:23839092.
    » http://dx.doi.org/10.3390/ijms140714214
  • JU, C., YUE, W., YANG, Z., ZHANG, Q., YANG, X., LIU, Z. and ZHANG, F., 2010. Antidiabetic effect and mechanism of chitooligosaccharides. Biological & Pharmaceutical Bulletin, vol. 33, no. 9, pp. 1511-1516. http://dx.doi.org/10.1248/bpb.33.1511 PMid:20823566.
    » http://dx.doi.org/10.1248/bpb.33.1511
  • KAELIN JUNIOR, W.G. and MCKNIGHT, S.L., 2013. Influence of metabolism on epigenetics and disease. Cell, vol. 153, no. 1, pp. 56-69. http://dx.doi.org/10.1016/j.cell.2013.03.004 PMid:23540690.
    » http://dx.doi.org/10.1016/j.cell.2013.03.004
  • KAMAL, M., KISHK, W.H., KHALIL, H.A., ABDEL-KHALEK, A.M., AYOUB, M.A., SWELUM, A.A., ALQHTANI, A.H., BA-AWADH, H.A. and ABD EL-HACK, M.E., 2023a. Effect of dietary chitosan supplementation on productive and physiological performance parameters of growing New Zealand white rabbits. International Journal of Biological Macromolecules, vol. 230, pp. 123166. http://dx.doi.org/10.1016/j.ijbiomac.2023.123166 PMid:36623627.
    » http://dx.doi.org/10.1016/j.ijbiomac.2023.123166
  • KAMAL, M., YOUSSEF, I.M., KHALIL, H.A., AYOUB, M.A. and HASHEM, N.M., 2023b. Multifunctional role of chitosan in farm animals: a comprehensive review. Annals of Animal Science, vol. 23, no. 1, pp. 69-86. http://dx.doi.org/10.2478/aoas-2022-0054
    » http://dx.doi.org/10.2478/aoas-2022-0054
  • KAMEL, M. and EL-SAYED, A., 2019. Utilization of herpesviridae as recombinant viral vectors in vaccine development against animal pathogens. Virus Research, vol. 270, pp. 197648. http://dx.doi.org/10.1016/j.virusres.2019.197648 PMid:31279828.
    » http://dx.doi.org/10.1016/j.virusres.2019.197648
  • KARADENIZ, F., ARTAN, M., KONG, C.S. and KIM, S.K., 2010. Chitooligosaccharides protect pancreatic β-cells from hydrogen peroxide-induced deterioration. Carbohydrate Polymers, vol. 82, no. 1, pp. 143-147. http://dx.doi.org/10.1016/j.carbpol.2010.04.046
    » http://dx.doi.org/10.1016/j.carbpol.2010.04.046
  • KARAGOZLU, M.Z. and KIM, S.K., 2014. Anticancer effects of chitin and chitosan derivatives. Advances in Food and Nutrition Research, vol. 72, pp. 215-225. http://dx.doi.org/10.1016/B978-0-12-800269-8.00012-9 PMid:25081085.
    » http://dx.doi.org/10.1016/B978-0-12-800269-8.00012-9
  • KATIYAR, D., SINGH, B., LALL, A.M. and HALDAR, C., 2011. Efficacy of chitooligosaccharides for the management of diabetes in alloxan induced mice: a correlative study with antihyperlipidemic and antioxidative activity. European Journal of Pharmaceutical Sciences, vol. 44, no. 4, pp. 534-543. http://dx.doi.org/10.1016/j.ejps.2011.09.015 PMid:21964204.
    » http://dx.doi.org/10.1016/j.ejps.2011.09.015
  • KAUR, G., GOYAL, J., BEHERA, P.K., DEVI, S., SINGH, S.K., GARG, V. and MITTAL, N., 2023. Unraveling the role of chitosan for nasal drug delivery systems: a review. Carbohydrate Polymer Technologies and Applications, vol. 5, pp. 100316. http://dx.doi.org/10.1016/j.carpta.2023.100316
    » http://dx.doi.org/10.1016/j.carpta.2023.100316
  • KAY, M.A., GLORIOSO, J.C. and NALDINI, L., 2001. Viral vectors for gene therapy: the art of turning infectious agents into vehicles of therapeutics. Nature Medicine, vol. 7, no. 1, pp. 33-40. http://dx.doi.org/10.1038/83324 PMid:11135613.
    » http://dx.doi.org/10.1038/83324
  • KHALAF, E.M., ABOOD, N.A., ATTA, R.Z., RAMÍREZ-CORONEL, A.A., ALAZRAGI, R., PARRA, R.M.R., ABED, O.H., ABOSAOODA, M., JALIL, A.T., MUSTAFA, Y.F., NARMANI, A. and FARHOOD, B., 2023. Recent progressions in biomedical and pharmaceutical applications of chitosan nanoparticles: a comprehensive review. International Journal of Biological Macromolecules, vol. 231, pp. 123354. http://dx.doi.org/10.1016/j.ijbiomac.2023.123354 PMid:36681228.
    » http://dx.doi.org/10.1016/j.ijbiomac.2023.123354
  • KHARROUBI, A.T. and DARWISH, H.M., 2015. Diabetes mellitus: the epidemic of the century. World Journal of Diabetes, vol. 6, no. 6, pp. 850-867. http://dx.doi.org/10.4239/wjd.v6.i6.850 PMid:26131326.
    » http://dx.doi.org/10.4239/wjd.v6.i6.850
  • KHATRI, D.K., PREETI, K., TONAPE, S., BHATTACHARJEE, S., PATEL, M., SHAH, S., SINGH, P.K., SRIVASTAVA, S., GUGULOTHU, D., VORA, L. and SINGH, S.B., 2023. Nanotechnological advances for nose to brain delivery of therapeutics to improve the Parkinson therapy. Current Neuropharmacology, vol. 21, no. 3, pp. 493-516. http://dx.doi.org/10.2174/1570159X20666220507022701 PMid:35524671.
    » http://dx.doi.org/10.2174/1570159X20666220507022701
  • KIM, H.J., AHN, H.Y., KWAK, J.H., SHIN, D.Y., KWON, Y.I., OH, C.G. and LEE, J.H., 2014a. The effects of chitosan oligosaccharide (GO2KA1) supplementation on glucose control in subjects with prediabetes. Food & Function, vol. 5, no. 10, pp. 2662-2669. http://dx.doi.org/10.1039/C4FO00469H PMid:25222285.
    » http://dx.doi.org/10.1039/C4FO00469H
  • KIM, J.G., JO, S.H., HA, K.S., KIM, S.C., KIM, Y.C., APOSTOLIDIS, E. and KWON, Y.I., 2014b. Effect of long-term supplementation of low molecular weight chitosan oligosaccharide (GO2KA1) on fasting blood glucose and HbA1c in db/db mice model and elucidation of mechanism of action. BMC Complementary and Alternative Medicine, vol. 14, no. 1, pp. 272. http://dx.doi.org/10.1186/1472-6882-14-272 PMid:25074485.
    » http://dx.doi.org/10.1186/1472-6882-14-272
  • KOCKISCH, S., REES, G.D., YOUNG, S.A., TSIBOUKLIS, J. and SMART, J.D., 2003. Polymeric microspheres for drug delivery to the oral cavity: an in vitro evaluation of mucoadhesive potential. Journal of Pharmaceutical Sciences, vol. 92, no. 8, pp. 1614-1623. http://dx.doi.org/10.1002/jps.10423 PMid:12884248.
    » http://dx.doi.org/10.1002/jps.10423
  • KONG, S., DING, C., HUANG, L., BAI, Y., XIAO, T., GUO, J. and SU, Z., 2017. The effects of COST on the differentiation of 3T3-L1 preadipocytes and the mechanism of action. Saudi Journal of Biological Sciences, vol. 24, no. 2, pp. 251-255. http://dx.doi.org/10.1016/j.sjbs.2016.09.008 PMid:28149159.
    » http://dx.doi.org/10.1016/j.sjbs.2016.09.008
  • KOYANI, R.D., ANDRADE, M., QUESTER, K., GAYTÁN, P., HUERTA-SAQUERO, A. and VAZQUEZ-DUHALT, R., 2018. Surface modification of protein enhances encapsulation in chitosan nanoparticles. Applied Nanoscience, vol. 8, no. 5, pp. 1197-1203. http://dx.doi.org/10.1007/s13204-018-0779-8
    » http://dx.doi.org/10.1007/s13204-018-0779-8
  • KUMAR, M.N.V.R., 2000. A review of chitin and chitosan applications. Reactive & Functional Polymers, vol. 46, no. 1, pp. 1-27. http://dx.doi.org/10.1016/S1381-5148(00)00038-9
    » http://dx.doi.org/10.1016/S1381-5148(00)00038-9
  • KUMAR, S.G., RAHMAN, M.A., LEE, S.H., HWANG, H.S., KIM, H.A. and YUN, J.W., 2009. Plasma proteome analysis for anti‐obesity and anti‐diabetic potentials of chitosan oligosaccharides in ob/ob mice. Proteomics, vol. 9, no. 8, pp. 2149-2162. http://dx.doi.org/10.1002/pmic.200800571 PMid:19296549.
    » http://dx.doi.org/10.1002/pmic.200800571
  • KUNJACHAN, S., JOSE, S. and LAMMERS, T., 2010. Understanding the mechanism of ionic gelation for synthesis of chitosan nanoparticles using qualitative techniques. Asian Journal of Pharmaceutics, vol. 4, no. 2, pp. 148. http://dx.doi.org/10.4103/0973-8398.68467
    » http://dx.doi.org/10.4103/0973-8398.68467
  • KURITA, K., IKEDA, H., YOSHIDA, Y., SHIMOJOH, M. and HARATA, M., 2002. Chemoselective protection of the amino groups of chitosan by controlled phthaloylation: facile preparation of a precursor useful for chemical modifications. Biomacromolecules, vol. 3, no. 1, pp. 1-4. http://dx.doi.org/10.1021/bm0101163 PMid:11866549.
    » http://dx.doi.org/10.1021/bm0101163
  • KURONO, Y., 2022. The mucosal immune system of the upper respiratory tract and recent progress in mucosal vaccines. Auris, Nasus, Larynx, vol. 49, no. 1, pp. 1-10. http://dx.doi.org/10.1016/j.anl.2021.07.003 PMid:34304944.
    » http://dx.doi.org/10.1016/j.anl.2021.07.003
  • LARSEN, J.R., DIMA, L., CORRELL, C.U. and MANU, P., 2018. The pharmacological management of metabolic syndrome. Expert Review of Clinical Pharmacology, vol. 11, no. 4, pp. 397-410. http://dx.doi.org/10.1080/17512433.2018.1429910 PMid:29345505.
    » http://dx.doi.org/10.1080/17512433.2018.1429910
  • LEE, J.Y., KIM, T.Y., KANG, H., OH, J., PARK, J.W., KIM, S.C., KIM, M., APOSTOLIDIS, E., KIM, Y.C. and KWON, Y.I., 2021. Anti-obesity and anti-adipogenic effects of chitosan oligosaccharide (GO2KA1) in SD rats and in 3T3-L1 preadipocytes models. Molecules, vol. 26, no. 2, pp. 331. http://dx.doi.org/10.3390/molecules26020331 PMid:33440605.
    » http://dx.doi.org/10.3390/molecules26020331
  • LENNON, H., SPERRIN, M., BADRICK, E. and RENEHAN, A.G., 2016. The obesity paradox in cancer: a review. Current Oncology Reports, vol. 18, no. 9, pp. 56. http://dx.doi.org/10.1007/s11912-016-0539-4 PMid:27475805.
    » http://dx.doi.org/10.1007/s11912-016-0539-4
  • LI, Y., LIU, H., XU, Q.S., DU, Y.G. and XU, J., 2014. Chitosan oligosaccharides block LPS-induced O-GlcNAcylation of NF-κB and endothelial inflammatory response. Carbohydrate Polymers, vol. 99, pp. 568-578. http://dx.doi.org/10.1016/j.carbpol.2013.08.082 PMid:24274545.
    » http://dx.doi.org/10.1016/j.carbpol.2013.08.082
  • LIN, Y.H., MI, F.L., CHEN, C.T., CHANG, W.C., PENG, S.F., LIANG, H.F. and SUNG, H.W., 2007. Preparation and characterization of nanoparticles shelled with chitosan for oral insulin delivery. Biomacromolecules, vol. 8, no. 1, pp. 146-152. http://dx.doi.org/10.1021/bm0607776 PMid:17206800.
    » http://dx.doi.org/10.1021/bm0607776
  • LIU, B., LIU, W.S., HAN, B.Q. and SUN, Y.Y., 2007a. Antidiabetic effects of chitooligosaccharides on pancreatic islet cells in streptozotocin-induced diabetic rats. World Journal of Gastroenterology, vol. 13, no. 5, pp. 725-731. http://dx.doi.org/10.3748/wjg.v13.i5.725 PMid:17278195.
    » http://dx.doi.org/10.3748/wjg.v13.i5.725
  • LIU, H., CHEN, B., MAO, Z. and GAO, C., 2007b. Chitosan nanoparticles for loading of toothpaste actives and adhesion on tooth analogs. Journal of Applied Polymer Science, vol. 106, no. 6, pp. 4248-4256. http://dx.doi.org/10.1002/app.27078
    » http://dx.doi.org/10.1002/app.27078
  • LIU, H.T., LI, W.M., XU, G., LI, X.Y., BAI, X.F., WEI, P., YU, C. and DU, Y.G., 2009. Chitosan oligosaccharides attenuate hydrogen peroxide-induced stress injury in human umbilical vein endothelial cells. Pharmacological Research, vol. 59, no. 3, pp. 167-175. http://dx.doi.org/10.1016/j.phrs.2008.12.001 PMid:19121394.
    » http://dx.doi.org/10.1016/j.phrs.2008.12.001
  • LIU, S.H., CHIU, C.Y., SHI, C.M. and CHIANG, M.T., 2018. Functional comparison of high and low molecular weight chitosan on lipid metabolism and signals in high-fat diet-fed rats. Marine Drugs, vol. 16, no. 8, pp. 251. http://dx.doi.org/10.3390/md16080251 PMid:30060615.
    » http://dx.doi.org/10.3390/md16080251
  • LIU, Y., CHEN, C., WANG, X., SUN, Y., ZHANG, J., CHEN, J. and SHI, Y., 2022. An epigenetic role of mitochondria in cancer. Cells, vol. 11, no. 16, pp. 2518. http://dx.doi.org/10.3390/cells11162518 PMid:36010594.
    » http://dx.doi.org/10.3390/cells11162518
  • LIU, Y., SUN, Y., GUO, Y., SHI, X., CHEN, X., FENG, W., WU, L.L., ZHANG, J., YU, S., WANG, Y. and SHI, Y., 2023. An overview: the diversified role of mitochondria in cancer metabolism. International Journal of Biological Sciences, vol. 19, no. 3, pp. 897-915. http://dx.doi.org/10.7150/ijbs.81609 PMid:36778129.
    » http://dx.doi.org/10.7150/ijbs.81609
  • LUO, G., CHEN, W., LUO, J. and LIU, J., 2021. Control and monitoring of lipoprotein levels in atherosclerosis induced rabbits using novel nanoparticulate medication of Lovastatin and Rosuvastatin. Micro & Nano Letters, vol. 16, no. 11, pp. 558-565. http://dx.doi.org/10.1049/mna2.12081
    » http://dx.doi.org/10.1049/mna2.12081
  • LUO, Y.L., XU, C.F., LI, H.J., CAO, Z.T., LIU, J., WANG, J.L., DU, X.J., YANG, X.Z., GU, Z. and WANG, J., 2018. Macrophage-specific in vivo gene editing using cationic lipid-assisted polymeric nanoparticles. ACS Nano, vol. 12, no. 2, pp. 994-1005. http://dx.doi.org/10.1021/acsnano.7b07874 PMid:29314827.
    » http://dx.doi.org/10.1021/acsnano.7b07874
  • MENG, Q.Y., WANG, H., CUI, Z.B., YU, W.G. and LU, X.Z., 2020. Chitosan oligosaccharides attenuate amyloid formation of hIAPP and protect pancreatic β-cells from cytotoxicity. Molecules, vol. 25, no. 6, pp. 1314. http://dx.doi.org/10.3390/molecules25061314 PMid:32183067.
    » http://dx.doi.org/10.3390/molecules25061314
  • MI, F.L., WU, Y.Y., LIN, Y.H., SONAJE, K., HO, Y.C., CHEN, C.T., JUANG, J.H. and SUNG, H.W., 2008. Oral delivery of peptide drugs using nanoparticles self-assembled by poly (γ-glutamic acid) and a chitosan derivative functionalized by trimethylation. Bioconjugate Chemistry, vol. 19, no. 6, pp. 1248-1255. http://dx.doi.org/10.1021/bc800076n PMid:18517235.
    » http://dx.doi.org/10.1021/bc800076n
  • MOUT, R. and ROTELLO, V.M., 2017. Cytosolic and nuclear delivery of CRISPR/Cas9-ribonucleoprotein for gene editing using arginine functionalized gold nanoparticles. Bio-Protocol, vol. 7, no. 20, e2586. http://dx.doi.org/10.21769/BioProtoc.2586 PMid:29226180.
    » http://dx.doi.org/10.21769/BioProtoc.2586
  • MUXIKA, A., ETXABIDE, A., URANGA, J., GUERRERO, P. and DE LA CABA, K., 2017. Chitosan as a bioactive polymer: processing, properties and applications. International Journal of Biological Macromolecules, vol. 105, no. Pt 2, pp. 1358-1368. http://dx.doi.org/10.1016/j.ijbiomac.2017.07.087 PMid:28735006.
    » http://dx.doi.org/10.1016/j.ijbiomac.2017.07.087
  • NAGY, V., 2018 [viewed 15 November 2023]. Chitosan-natural antioxidant conjugates: synthesis, antimicrobial and antioxidant properties [online]. Reykjavík: University of Iceland. Doctoral dissertation. Available from: http://hdl.handle.net/1946/32021
    » http://hdl.handle.net/1946/32021
  • NAKHAEI, H., MOGHARNASI, M. and FANAEI, H., 2019. Effect of swimming training on levels of asprosin, lipid profile, glucose and insulin resistance in rats with metabolic syndrome. Obesity Medicine, vol. 15, pp. 100111. http://dx.doi.org/10.1016/j.obmed.2019.100111
    » http://dx.doi.org/10.1016/j.obmed.2019.100111
  • NAVEED, M., PHIL, L., SOHAIL, M., HASNAT, M., BAIG, M.M.F.A., IHSAN, A.U., SHUMZAID, M., KAKAR, M.U., KHAN, T.M., AKABAR, M. HUSSAIN, M.I. and ZHOU Q.-G., 2019. Chitosan oligosaccharide (COS): an overview. International Journal of Biological Macromolecules, vol. 129, pp. 827-843. http://dx.doi.org/10.1016/j.ijbiomac.2019.01.192 PMid:30708011.
    » http://dx.doi.org/10.1016/j.ijbiomac.2019.01.192
  • NAYAK, D., MINZ, A.P., ASHE, S., RAUTA, P.R., KUMARI, M., CHOPRA, P. and NAYAK, B., 2016. Synergistic combination of antioxidants, silver nanoparticles and chitosan in a nanoparticle based formulation: characterization and cytotoxic effect on MCF-7 breast cancer cell lines. Journal of Colloid and Interface Science, vol. 470, pp. 142-152. http://dx.doi.org/10.1016/j.jcis.2016.02.043 PMid:26939078.
    » http://dx.doi.org/10.1016/j.jcis.2016.02.043
  • NEGM, N.A., HEFNI, H.H., ABD-ELAAL, A.A., BADR, E.A. and ABOU KANA, M.T., 2020. Advancement on modification of chitosan biopolymer and its potential applications. International Journal of Biological Macromolecules, vol. 152, pp. 681-702. http://dx.doi.org/10.1016/j.ijbiomac.2020.02.196 PMid:32084486.
    » http://dx.doi.org/10.1016/j.ijbiomac.2020.02.196
  • NGO, D.H. and KIM, S.K., 2014. Antioxidant effects of chitin, chitosan, and their derivatives. Advances in Food and Nutrition Research, vol. 73, pp. 15-31. http://dx.doi.org/10.1016/B978-0-12-800268-1.00002-0 PMid:25300540.
    » http://dx.doi.org/10.1016/B978-0-12-800268-1.00002-0
  • NIAZ, T., SHABBIR, S., MANZOOR, S., REHMAN, A., RAHMAN, A., NASIR, H. and IMRAN, M., 2016. Antihypertensive nano-ceuticales based on chitosan biopolymer: physico-chemical evaluation and release kinetics. Carbohydrate Polymers, vol. 142, pp. 268-274. http://dx.doi.org/10.1016/j.carbpol.2016.01.047 PMid:26917399.
    » http://dx.doi.org/10.1016/j.carbpol.2016.01.047
  • NOJOKI, F., EBRAHIMI-HOSSEINZADEH, B., HATAMIAN-ZARMI, A., KHODAGHOLI, F. and KHEZRI, K., 2022. Design and development of chitosan-insulin-transfersomes (Transfersulin) as effective intranasal nanovesicles for the treatment of Alzheimer’s disease: in vitro, in vivo, and ex vivo evaluations. Biomedicine and Pharmacotherapy, vol. 153, pp. 113450. http://dx.doi.org/10.1016/j.biopha.2022.113450 PMid:36076565.
    » http://dx.doi.org/10.1016/j.biopha.2022.113450
  • NWE, N., FURUIKE, T. and TAMURA, H., 2009. The mechanical and biological properties of chitosan scaffolds for tissue regeneration templates are significantly enhanced by chitosan from Gongronella butleri. Materials, vol. 2, no. 2, pp. 374-398. http://dx.doi.org/10.3390/ma2020374
    » http://dx.doi.org/10.3390/ma2020374
  • OHYA, Y., SHIRATANI, M., KOBAYASHI, H. and OUCHI, T., 1994. Release behavior of 5-fluorouracil from chitosan-gel nanospheres immobilizing 5-fluorouracil coated with polysaccharides and their cell specific cytotoxicity. Journal of Macromolecular Science, Part A: Pure and Applied Chemistry, vol. 31, no. 5, pp. 629-642. http://dx.doi.org/10.1080/10601329409349743
    » http://dx.doi.org/10.1080/10601329409349743
  • OKSAL, E., PANGESTIKA, I., MUHAMMAD, T.S.T., MOHAMAD, H., AMIR, H., KASSIM, M.N.I. and ANDRIANI, Y., 2020. In vitro and in vivo studies of nanoparticles of chitosan-Pandanus tectorius fruit extract as new alternative treatment for hypercholesterolemia via Scavenger Receptor Class B type 1 pathway. Saudi Pharmaceutical Journal, vol. 28, no. 10, pp. 1263-1275. http://dx.doi.org/10.1016/j.jsps.2020.08.017 PMid:33132720.
    » http://dx.doi.org/10.1016/j.jsps.2020.08.017
  • OPARIL, S., ZAMAN, M.A. and CALHOUN, D.A., 2003. Pathogenesis of hypertension. Annals of Internal Medicine, vol. 139, no. 9, pp. 761-776. http://dx.doi.org/10.7326/0003-4819-139-9-200311040-00011 PMid:14597461.
    » http://dx.doi.org/10.7326/0003-4819-139-9-200311040-00011
  • ORELLANO, M.S., PORPORATTO, C., SILBER, J.J., FALCONE, R.D. and CORREA, N.M., 2017. AOT reverse micelles as versatile reaction media for chitosan nanoparticles synthesis. Carbohydrate Polymers, vol. 171, pp. 85-93. http://dx.doi.org/10.1016/j.carbpol.2017.04.074 PMid:28578974.
    » http://dx.doi.org/10.1016/j.carbpol.2017.04.074
  • OTHMAN, N., MASARUDIN, M.J., KUEN, C.Y., DASUAN, N.A., ABDULLAH, L.C. and JAMIL, S.N.A.M., 2018. Synthesis and optimization of chitosan nanoparticles loaded with L-ascorbic acid and thymoquinone. Nanomaterials, vol. 8, no. 11, pp. 920. http://dx.doi.org/10.3390/nano8110920 PMid:30405074.
    » http://dx.doi.org/10.3390/nano8110920
  • PAN, H., FU, C., HUANG, L., JIANG, Y., DENG, X., GUO, J. and SU, Z., 2018. Anti-obesity effect of chitosan oligosaccharide capsules (COSCs) in obese rats by ameliorating leptin resistance and adipogenesis. Marine Drugs, vol. 16, no. 6, pp. 198. http://dx.doi.org/10.3390/md16060198 PMid:29874843.
    » http://dx.doi.org/10.3390/md16060198
  • PARK, P.J., AHN, C.B., JEON, Y.J. and JE, J.Y., 2008. Renin inhibition activity by chitooligosaccharides. Bioorganic & Medicinal Chemistry Letters, vol. 18, no. 7, pp. 2471-2474. http://dx.doi.org/10.1016/j.bmcl.2008.02.041 PMid:18313296.
    » http://dx.doi.org/10.1016/j.bmcl.2008.02.041
  • PARK, P.J., JE, J.Y. and KIM, S.K., 2003. Angiotensin I converting enzyme (ACE) inhibitory activity of hetero-chitooligosaccharides prepared from partially different deacetylated chitosans. Journal of Agricultural and Food Chemistry, vol. 51, no. 17, pp. 4930-4934. http://dx.doi.org/10.1021/jf0340557 PMid:12903948.
    » http://dx.doi.org/10.1021/jf0340557
  • PEERS, S., MONTEMBAULT, A. and LADAVIÈRE, C., 2020. Chitosan hydrogels for sustained drug delivery. Journal of Controlled Release, vol. 326, pp. 150-163. http://dx.doi.org/10.1016/j.jconrel.2020.06.012 PMid:32562854.
    » http://dx.doi.org/10.1016/j.jconrel.2020.06.012
  • QI, L., XU, Z., JIANG, X., HU, C. and ZOU, X., 2004. Preparation and antibacterial activity of chitosan nanoparticles. Carbohydrate Research, vol. 339, no. 16, pp. 2693-2700. http://dx.doi.org/10.1016/j.carres.2004.09.007 PMid:15519328.
    » http://dx.doi.org/10.1016/j.carres.2004.09.007
  • QUESTER, K., RODRÍGUEZ-GONZÁLEZ, S., GONZÁLEZ-DÁVALOS, L., LOZANO-FLORES, C., GONZÁLEZ-GALLARDO, A., ZAPIAIN-MERINO, S.J., SHIMADA, A., MORA, O. and VAZQUEZ-DUHALT, R., 2022. Chitosan nanoparticles containing lipoic acid with antioxidant properties as a potential nutritional supplement. Animals, vol. 12, no. 4, pp. 417. http://dx.doi.org/10.3390/ani12040417 PMid:35203125.
    » http://dx.doi.org/10.3390/ani12040417
  • RAHMAN, M.A., KUMAR, S.G., KIM, S.W., HWANG, H.J., BAEK, Y.M., LEE, S.H., HWANG, H.S., SHON, Y.H., NAM, K.S. and YUN, J.W., 2008. Proteomic analysis for inhibitory effect of chitosan oligosaccharides on 3T3‐L1 adipocyte differentiation. Proteomics, vol. 8, no. 3, pp. 569-581. http://dx.doi.org/10.1002/pmic.200700888 PMid:18175373.
    » http://dx.doi.org/10.1002/pmic.200700888
  • RAMKUMAR, S., RAGHUNATH, A. and RAGHUNATH, S., 2016. Statin therapy: review of safety and potential side effects. Acta Cardiologica Sinica, vol. 32, no. 6, pp. 631-639. http://dx.doi.org/10.6515/ACS20160611A PMid:27899849.
    » http://dx.doi.org/10.6515/ACS20160611A
  • ROCHA, A.R.F., MORAIS, N.S., PRIORE, S.E. and FRANCESCHINI, S.C.C., 2022. Inflammatory biomarkers and components of metabolic syndrome in adolescents: a systematic review. Inflammation, vol. 45, no. 1, pp. 14-30. http://dx.doi.org/10.1007/s10753-021-01549-1 PMid:34546513.
    » http://dx.doi.org/10.1007/s10753-021-01549-1
  • ROSSI, J.L.S., BARBALHO, S.M., ARAUJO, R.R., BECHARA, M.D., SLOAN, K.P. and SLOAN, L.A., 2022. Metabolic syndrome and cardiovascular diseases: going beyond traditional risk factors. Diabetes Metabolism Research and Reviews, vol. 38, no. 3, e3502. http://dx.doi.org/10.1002/dmrr.3502 PMid:34614543.
    » http://dx.doi.org/10.1002/dmrr.3502
  • SABERI, M., BJELICA, D., SCHENK, S., IMAMURA, T., BANDYOPADHYAY, G., LI, P., JADHAR, V., VARGEESE, C., WANG, W., BOWMAN, K., ZHANG, Y., POLISKY, B. and OLEFSKY, J.M., 2009. Novel liver-specific TORC2 siRNA corrects hyperglycemia in rodent models of type 2 diabetes. American Journal of Physiology. Endocrinology and Metabolism, vol. 297, no. 5, pp. E1137-E1146. http://dx.doi.org/10.1152/ajpendo.00158.2009 PMid:19706791.
    » http://dx.doi.org/10.1152/ajpendo.00158.2009
  • SABOYA, P.P., BODANESE, L.C., ZIMMERMANN, P.R., GUSTAVO, A.D.S., MACAGNAN, F.E., FEOLI, A.P. and OLIVEIRA, M.D.S., 2017. Lifestyle intervention on metabolic syndrome and its impact on quality of life: a randomized controlled trial. Arquivos Brasileiros de Cardiologia, vol. 108, no. 1, pp. 60-69. http://dx.doi.org/10.5935/abc.20160186 PMid:27982160.
    » http://dx.doi.org/10.5935/abc.20160186
  • SAFDAR, R., OMAR, A.A., ARUNAGIRI, A., REGUPATHI, I. and THANABALAN, M., 2019. Potential of chitosan and its derivatives for controlled drug release applications: a review. Journal of Drug Delivery Science and Technology, vol. 49, pp. 642-659. http://dx.doi.org/10.1016/j.jddst.2018.10.020
    » http://dx.doi.org/10.1016/j.jddst.2018.10.020
  • SAKLAYEN, M.G., 2018. The global epidemic of the metabolic syndrome. Current Hypertension Reports, vol. 20, no. 2, pp. 12. http://dx.doi.org/10.1007/s11906-018-0812-z PMid:29480368.
    » http://dx.doi.org/10.1007/s11906-018-0812-z
  • SALEM, S.A., DIAB, M.S.M., SHEHATA, S.H. and SOLIMAN, T.N., 2021. Nanochitosan effect on biomolecular, hypolipidemic in rats and incorporation in functional yogurt. Pakistan Journal of Biological Sciences, vol. 24, no. 5, pp. 548-561. http://dx.doi.org/10.3923/pjbs.2021.548.561 PMid:34486330.
    » http://dx.doi.org/10.3923/pjbs.2021.548.561
  • SAMI, M.M., AL-KAZZAZ, F.F. and ABDULSATTAR, S.A., 2022. The effect of using Chitosan and Nano-chitosan synthesized from blue carb in the treatment of hyperglycemia and glomerulus of diabetic guinea pigs. Egyptian Journal of Chemistry, vol. 65, no. 6, pp. 55-58. http://dx.doi.org/10.21608/EJCHEM.2021.85130.4151
    » http://dx.doi.org/10.21608/EJCHEM.2021.85130.4151
  • SÁNCHEZ-JIMÉNEZ, F., PÉREZ-PÉREZ, A., DE LA CRUZ-MERINO, L. and SÁNCHEZ-MARGALET, V., 2019. Obesity and breast cancer: role of leptin. Frontiers in Oncology, vol. 9, pp. 596. http://dx.doi.org/10.3389/fonc.2019.00596 PMid:31380268.
    » http://dx.doi.org/10.3389/fonc.2019.00596
  • SARMENTO, B., MARTINS, S., RIBEIRO, A., VEIGA, F., NEUFELD, R. and FERREIRA, D., 2006. Development and comparison of different nanoparticulate polyelectrolyte complexes as insulin carriers. International Journal of Peptide Research and Therapeutics, vol. 12, no. 2, pp. 131-138. http://dx.doi.org/10.1007/s10989-005-9010-3
    » http://dx.doi.org/10.1007/s10989-005-9010-3
  • SHARIATINIA, Z., 2019. Pharmaceutical applications of chitosan. Advances in Colloid and Interface Science, vol. 263, pp. 131-194. http://dx.doi.org/10.1016/j.cis.2018.11.008 PMid:30530176.
    » http://dx.doi.org/10.1016/j.cis.2018.11.008
  • SHARMA, D., ARORA, S., BANERJEE, A. and SINGH, J., 2021. Improved insulin sensitivity in obese-diabetic mice via chitosan Nanomicelles mediated silencing of pro-inflammatory Adipocytokines. Nanomedicine: Nanotechnology, Biology and Medicine, vol. 33, pp. 102357. http://dx.doi.org/10.1016/j.nano.2020.102357 PMid:33460779.
    » http://dx.doi.org/10.1016/j.nano.2020.102357
  • SHARMA, M., SHARMA, R. and JAIN, D.K., 2018. Preparation, characterization and evaluation of nebivolol loaded chitosan nanoparticles. Journal of Drug Delivery and Therapeutics, vol. 8, no. 2, pp. 118-122. http://dx.doi.org/10.22270/jddt.v8i2.1730
    » http://dx.doi.org/10.22270/jddt.v8i2.1730
  • SHEKHAR, S., LIU, Y., WANG, S., ZHANG, H., FANG, X., ZHANG, J., FAN, L., ZHENG, B., ROMAN, R.J., WANG, Z., FAN, F. and BOOZ, G.W., 2021. Novel mechanistic insights and potential therapeutic impact of TRPC6 in neurovascular coupling and ischemic stroke. International Journal of Molecular Sciences, vol. 22, no. 4, pp. 2074. http://dx.doi.org/10.3390/ijms22042074 PMid:33669830.
    » http://dx.doi.org/10.3390/ijms22042074
  • SLATE-ROMANO, J.J., YANO, N. and ZHAO, T.C., 2022. Irisin reduces inflammatory signaling pathways in inflammation-mediated metabolic syndrome. Molecular and Cellular Endocrinology, vol. 552, pp. 111676. http://dx.doi.org/10.1016/j.mce.2022.111676 PMid:35569582.
    » http://dx.doi.org/10.1016/j.mce.2022.111676
  • SOUTO, E.B., SOUTO, S.B., CAMPOS, J.R., SEVERINO, P., PASHIROVA, T.N., ZAKHAROVA, L.Y., SILVA, A.M., DURAZZO, A., LUCARINI, M., IZZO, A.A. and SANTINI, A., 2019. Nanoparticle delivery systems in the treatment of diabetes complications. Molecules, vol. 24, no. 23, pp. 4209. http://dx.doi.org/10.3390/molecules24234209 PMid:31756981.
    » http://dx.doi.org/10.3390/molecules24234209
  • SRIAMORNSAK, P. and DASS, C.R., 2022. Chitosan nanoparticles in atherosclerosis: development to preclinical testing. Pharmaceutics, vol. 14, no. 5, pp. 935. http://dx.doi.org/10.3390/pharmaceutics14050935 PMid:35631521.
    » http://dx.doi.org/10.3390/pharmaceutics14050935
  • SRIKANTH, S. and DEEDWANIA, P., 2016. Management of dyslipidemia in patients with hypertension, diabetes, and metabolic syndrome. Current Hypertension Reports, vol. 18, no. 10, pp. 76. http://dx.doi.org/10.1007/s11906-016-0683-0 PMid:27730495.
    » http://dx.doi.org/10.1007/s11906-016-0683-0
  • SUGANO, M., WATANABE, S., KISHI, A., IZUME, M. and OHTAKARA, A., 1988. Hypocholesterolemic action of chitosans with different viscosity in rats. Lipids, vol. 23, no. 3, pp. 187-191. http://dx.doi.org/10.1007/BF02535456 PMid:2836688.
    » http://dx.doi.org/10.1007/BF02535456
  • SUN, T., ZHOU, D., XIE, J. and MAO, F., 2007. Preparation of chitosan oligomers and their antioxidant activity. European Food Research and Technology, vol. 225, no. 3-4, pp. 451-456. http://dx.doi.org/10.1007/s00217-006-0439-1
    » http://dx.doi.org/10.1007/s00217-006-0439-1
  • SWIERCZEWSKA, M., HAN, H.S., KIM, K., PARK, J.H. and LEE, S., 2016. Polysaccharide-based nanoparticles for theranostic nanomedicine. Advanced Drug Delivery Reviews, vol. 99, no. Pt A, pp. 70-84. http://dx.doi.org/10.1016/j.addr.2015.11.015 PMid:26639578.
    » http://dx.doi.org/10.1016/j.addr.2015.11.015
  • TAN, H., CHU, C.R., PAYNE, K.A. and MARRA, K.G., 2009. Injectable in situ forming biodegradable chitosan-hyaluronic acid-based hydrogels for cartilage tissue engineering. Biomaterials, vol. 30, no. 13, pp. 2499-2506. http://dx.doi.org/10.1016/j.biomaterials.2008.12.080 PMid:19167750.
    » http://dx.doi.org/10.1016/j.biomaterials.2008.12.080
  • TAO, W., SUN, W., LIU, L., WANG, G., XIAO, Z., PEI, X. and WANG, M., 2019. Chitosan oligosaccharide attenuates nonalcoholic fatty liver disease induced by high fat diet through reducing lipid accumulation, inflammation and oxidative stress in C57BL/6 mice. Marine Drugs, vol. 17, no. 11, pp. 645. http://dx.doi.org/10.3390/md17110645 PMid:31744059.
    » http://dx.doi.org/10.3390/md17110645
  • TAO, W., WANG, G. and WEI, J., 2021. The role of chitosan oligosaccharide in metabolic syndrome: a review of possible mechanisms. Marine Drugs, vol. 19, no. 9, pp. 501. http://dx.doi.org/10.3390/md19090501 PMid:34564163.
    » http://dx.doi.org/10.3390/md19090501
  • TIAN, X.-X. and GROVES, M.J., 1999. Formulation and biological activity of antineoplastic proteoglycans derived from Mycobacterium vaccae in chitosan nanoparticles. The Journal of Pharmacy and Pharmacology, vol. 51, no. 2, pp. 151-157. http://dx.doi.org/10.1211/0022357991772268 PMid:10217313.
    » http://dx.doi.org/10.1211/0022357991772268
  • TING, D.R. and SHEN, Y., 2005. Antibacterial finishing with chitosan derivatives and their nano-particles. Dyeing Finishing, vol. 14, no. 4, pp. 12-14.
  • TOKUMITSU, H., ICHIKAWA, H., FUKUMORI, Y. and BLOCK, L.H., 1999. Preparation of gadopentetic acid-loaded chitosan microparticles for gadolinium neutron-capture therapy of cancer by a novel emulsion-droplet coalescence technique. Chemical & Pharmaceutical Bulletin, vol. 47, no. 6, pp. 838-842. http://dx.doi.org/10.1248/cpb.47.838 PMid:10399838.
    » http://dx.doi.org/10.1248/cpb.47.838
  • TRIVEDI, V.R., SATIA, M.C., DESCHAMPS, A., MAQUET, V., SHAH, R.B., ZINZUWADIA, P.H. and TRIVEDI, J.V., 2015. Single-blind, placebo controlled randomised clinical study of chitosan for body weight reduction. Nutrition Journal, vol. 15, no. 1, pp. 3. http://dx.doi.org/10.1186/s12937-016-0122-8 PMid:26747458.
    » http://dx.doi.org/10.1186/s12937-016-0122-8
  • VENKATESAN, J. and KIM, S.K., 2010. Chitosan composites for bone tissue engineering: an overview. Marine Drugs, vol. 8, no. 8, pp. 2252-2266. http://dx.doi.org/10.3390/md8082252 PMid:20948907.
    » http://dx.doi.org/10.3390/md8082252
  • VIRMANI, T., KUMAR, G., SHARMA, A., PATHAK, K., AKHTAR, M.S., AFZAL, O. and ALTAMIMI, A.S., 2023. Amelioration of cancer employing chitosan, its derivatives, and chitosan-based nanoparticles: recent updates. Polymers, vol. 15, no. 13, pp. 2928. http://dx.doi.org/10.3390/polym15132928 PMid:37447573.
    » http://dx.doi.org/10.3390/polym15132928
  • WANG, G.S. and HOYTE, C., 2019. Review of biguanide (metformin) toxicity. Journal of Intensive Care Medicine, vol. 34, no. 11-12, pp. 863-876. http://dx.doi.org/10.1177/0885066618793385 PMid:30126348.
    » http://dx.doi.org/10.1177/0885066618793385
  • WANG, J., BYRNE, J.D., NAPIER, M.E. and DESIMONE, J.M., 2011. More effective nanomedicines through particle design. Small, vol. 7, no. 14, pp. 1919-1931. http://dx.doi.org/10.1002/smll.201100442 PMid:21695781.
    » http://dx.doi.org/10.1002/smll.201100442
  • WEI, L., TAN, W., WANG, G., LI, Q., DONG, F. and GUO, Z., 2019. The antioxidant and antifungal activity of chitosan derivatives bearing Schiff bases and quaternary ammonium salts. Carbohydrate Polymers, vol. 226, pp. 115256. http://dx.doi.org/10.1016/j.carbpol.2019.115256 PMid:31582056.
    » http://dx.doi.org/10.1016/j.carbpol.2019.115256
  • WEYERS, M., PETERSON, B., HAMMAN, J.H. and STEENEKAMP, J.H., 2022. Formulation of chitosan microparticles for enhanced intranasal macromolecular compound delivery: factors that influence particle size during ionic gelation. Gels, vol. 8, no. 11, pp. 686. http://dx.doi.org/10.3390/gels8110686 PMid:36354594.
    » http://dx.doi.org/10.3390/gels8110686
  • WISSE, B.E., 2004. The inflammatory syndrome: the role of adipose tissue cytokines in metabolic disorders linked to obesity. Journal of the American Society of Nephrology, vol. 15, no. 11, pp. 2792-2800. http://dx.doi.org/10.1097/01.ASN.0000141966.69934.21 PMid:15504932.
    » http://dx.doi.org/10.1097/01.ASN.0000141966.69934.21
  • WONG, C.Y., AL-SALAMI, H. and DASS, C.R., 2017. Potential of insulin nanoparticle formulations for oral delivery and diabetes treatment. Journal of Controlled Release, vol. 264, pp. 247-275. http://dx.doi.org/10.1016/j.jconrel.2017.09.003 PMid:28887133.
    » http://dx.doi.org/10.1016/j.jconrel.2017.09.003
  • WUTS, P.G. and GREENE, T.W., 2006. Greene’s protective groups in organic synthesis New York: John Wiley & Sons. http://dx.doi.org/10.1002/0470053488
    » http://dx.doi.org/10.1002/0470053488
  • XIAO, S., MAO, L., XIAO, J., WU, Y. and LIU, H., 2021. Selenium nanoparticles inhibit the formation of atherosclerosis in apolipoprotein E deficient mice by alleviating hyperlipidemia and oxidative stress. European Journal of Pharmacology, vol. 902, pp. 174120. http://dx.doi.org/10.1016/j.ejphar.2021.174120 PMid:33905703.
    » http://dx.doi.org/10.1016/j.ejphar.2021.174120
  • YADAV, M., KAUSHIK, B., RAO, G.K., SRIVASTAVA, C.M. and VAYA, D., 2023. Advances and challenges in the use of chitosan and its derivatives in biomedical fields: a review. Carbohydrate Polymer Technologies and Applications, vol. 5, pp. 100323. http://dx.doi.org/10.1016/j.carpta.2023.100323
    » http://dx.doi.org/10.1016/j.carpta.2023.100323
  • YUAN, W.P., LIU, B., LIU, C.H., WANG, X.J., ZHANG, M.S., MENG, X.M. and XIA, X.K., 2009. Antioxidant activity of chito-oligosaccharides on pancreatic islet cells in streptozotocin-induced diabetes in rats. World Journal of Gastroenterology, vol. 15, no. 11, pp. 1339-1345. http://dx.doi.org/10.3748/wjg.15.1339 PMid:19294763.
    » http://dx.doi.org/10.3748/wjg.15.1339
  • ZHANG, J., LIU, J., LI, L. and XIA, W., 2008. Dietary chitosan improves hypercholesterolemia in rats fed high-fat diets. Nutrition Research, vol. 28, no. 6, pp. 383-390. http://dx.doi.org/10.1016/j.nutres.2007.12.013 PMid:19083436.
    » http://dx.doi.org/10.1016/j.nutres.2007.12.013
  • ZHANG, M., HOU, G., HU, P., FENG, D., WANG, J. and ZHU, W., 2021. Nano chitosan-zinc complex improves the growth performance and antioxidant capacity of the small intestine in weaned piglets. British Journal of Nutrition, vol. 126, no. 6, pp. 801-812. http://dx.doi.org/10.1017/S0007114520004766 PMid:33256856.
    » http://dx.doi.org/10.1017/S0007114520004766
  • ZHANG, X., SUN, M., ZHENG, A., CAO, D., BI, Y. and SUN, J., 2012. Preparation and characterization of insulin-loaded bioadhesive PLGA nanoparticles for oral administration. European Journal of Pharmaceutical Sciences, vol. 45, no. 5, pp. 632-638. http://dx.doi.org/10.1016/j.ejps.2012.01.002 PMid:22248882.
    » http://dx.doi.org/10.1016/j.ejps.2012.01.002
  • ZHENG, J., YUAN, X., CHENG, G., JIAO, S., FENG, C., ZHAO, X., YIN, H., DU, Y. and LIU, H., 2018. Chitosan oligosaccharides improve the disturbance in glucose metabolism and reverse the dysbiosis of gut microbiota in diabetic mice. Carbohydrate Polymers, vol. 190, pp. 77-86. http://dx.doi.org/10.1016/j.carbpol.2018.02.058 PMid:29628262.
    » http://dx.doi.org/10.1016/j.carbpol.2018.02.058
  • ZHOU, T.X., CHEN, Y.J., YOO, J.S., HUANG, Y., LEE, J.H., JANG, H.D., SHIN, S.O., KIM, H.J., CHO, J.H. and KIM, I.H., 2009. Effects of chitooligosaccharide supplementation on performance, blood characteristics, relative organ weight, and meat quality in broiler chickens. Poultry Science, vol. 88, no. 3, pp. 593-600. http://dx.doi.org/10.3382/ps.2008-00285 PMid:19211530.
    » http://dx.doi.org/10.3382/ps.2008-00285

Publication Dates

  • Publication in this collection
    26 Feb 2024
  • Date of issue
    2023

History

  • Received
    12 July 2023
  • Accepted
    15 Nov 2023
Instituto Internacional de Ecologia R. Bento Carlos, 750, 13560-660 São Carlos SP - Brasil, Tel. e Fax: (55 16) 3362-5400 - São Carlos - SP - Brazil
E-mail: bjb@bjb.com.br