Hydroxycinnamic Acids and Derivatives Formulations for Skin Damages and Disorders: A Review
Abstract
:1. Introduction
2. Hydroxycinnamic Acids and Derivatives: General Uses and Current Applications
2.1. Cinnamic Acid
2.2. Ferulic Acid
2.3. p-Coumaric Acid
2.4. Caffeic Acid
2.5. Syringic Acid
2.6. Vanillic Acid
2.7. Rosmarinic Acid
2.8. Chlorogenic Acid
3. Hydroxycinnamic Acids and Derivatives: Activities on Skin Disorders
3.1. Wound Healing
3.2. Burns and UV-Induced Damages
3.3. Dermatitis and Inflammation
3.4. Psoriasis
4. Hydroxycinnamic Acids and Derivatives: Advanced Formulations
4.1. Micro and Nano-Particles
4.2. Fibers
4.3. Hydrogels and Scaffolds
4.4. Films and Creams
5. Conclusions and Future Perspectives
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Conflicts of Interest
Abbreviations
AA | Arachidonic acid |
AD | Atopic dermatitis |
AP-1 | Activator Protein-1 |
CA | Caffeic Acid |
CAPE | Caffeic Acid Phenethyl Ester |
CH | Chitosan |
CinAc | Cinnamic Acid |
CGA | Chlorogenic Acid |
COX-2 | cyclooxygenase-2 |
DMSO | Dimethyl Sulfoxide |
DNCB | 2,4-Dinitrochlorobenzene |
DNFB | 1-Fluoro-2,4-dinitrobenzene |
DPPH | 2,2-Diphenyl-1-picrylhydrazyl |
ECM | Extracellular Matrix |
EGFR | Epidermal Growth Factor Receptor |
FA | Ferulic Acid |
GPX | Glutathione Peroxidase |
GRAS | generally recognize As Safety |
GSH | glutathione |
HaCaT | Human Dermal Keratinocyte |
HDFa | Human Dermal Fibroblast adult |
ICAM-1 | Intracellular Adhesion Molecule-1 |
IFN-γ | Interferon-gamma |
IgE | Immunoglobulin E |
IL-10 | Interleukin-10 |
IL-12 | Interleukin-12 |
IL-13 | Interleukin-13 |
IL-17 | Interleukin-17 |
IL-17A | Interleukin-17 A |
IL-17RA | Interleukin-17 receptor A |
IL-1β | Interleukin-1β |
IL-2 | Interleukin-2 |
IL-23 | Interleukin-23 |
IL-31 | Interleukin-31 |
IL-4 | Interleukin-4 |
IL-5 | Interleukin-5 |
IL-6 | Interleukin-6 |
IL-8 | Interleukin-8 |
IMQ | Imiquimod |
IRAK1 | Interleukin-1 Receptor Associated Kinase 1 |
IRAK4 | Interleukin-1 Receptor Associated Kinase 4 |
JAK | Janus Kinase |
LPS | Lipopolysaccharide |
MAPK | Mitogen-activated protein kinase |
MDCK | Madin-Darby Canine Kidney |
MESL | oil-in-water-type Microemulsion |
MMP-1 | Matrix Metalloproteinase-1 |
MMP-3 | Matrix Metalloproteinase-3 |
MMP-9 | Matrix Metalloproteinase-9 |
MyD88 | Myeloid Differentiation Factor 88 |
NADH | Nicotinamide adenine dinucleotide hydride |
NFE2 | Nuclear Factor Erythroid 2 |
NF-κB | Nuclear Factor kappa-light-chain-enhancer of activated B cells |
NHDF-neo | normal human dermal fibroblast-neonatal |
NLC | Nanostructured Lipid Carrier |
NLRP3 | Nucleotide-binding oligomerization domain |
NO | Nitric Oxide |
NRF2 | nuclear factor erythroid 2–related factor 2 |
PCA | p-coumaric acid |
PCL | Poly ε-caprolactone |
PGE2 | Prostaglandin E2 |
pHEMA | poly(2-hydroxyethyl methacrylate) |
p-IκB-α | nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor alpha |
PIK3/AKT | Phosphoinositide 3-kinases; alfa serine/threonine-protein kinases |
PLA | Poly Lactid Acid |
PLGA | Poly Lactic Co-Glycolic Acid |
PPARγ | Peroxisome proliferator-activated receptor γ |
PTP- κ | Protein tyrosine phosphatases-kappa |
PVP | Polyvinylpyrrolidone |
RA | rosmarinic acid |
ROS | reactive oxygen species |
SA | syringic acid |
SOD | Superoxide dismutase |
STAT | signal transducers and activators of transcription |
Th2 | T helper type 2 cells |
Th17 | T helper type 17 cells |
TLR-3 | Tool-Like Receptor-3 |
TLR-4 | Tool-Like Receptor-4 |
TNFα | Tumor Necrosis Factor α |
TPA | 12-O-tetradecanoylphorbol-13-acetate |
VA | vanillic acid |
γ-PGA | Poly-γ-Glutamic Acid |
References
- Suarato, G.; Bertorelli, R.; Athanassiou, A. Borrowing from Nature: Biopolymers and biocomposites as smart wound care materials. Front. Bioeng. Biotechnol. 2018, 6, 137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Raghav, A.; Khan, Z.A.; Labala, R.K.; Ahmad, J.; Noor, S.; Mishra, B.K. Financial burden of diabetic foot ulcers to world: A progressive topic to discuss always. Ther. Adv. Endocrinol. Metab. 2018, 9, 29–31. [Google Scholar] [CrossRef] [PubMed]
- Thomsen, S.F.; Skov, L.; Dodge, R.; Hedegaard, M.S.; Kjellberg, J. Socioeconomic costs and health inequalities from psoriasis: A cohort study. Dermatology 2019, 235, 372–379. [Google Scholar] [CrossRef]
- Drucker, A.M.; Wang, A.R.; Li, W.-Q.; Sevetson, E.; Block, J.K.; Qureshi, A.A. The burden of atopic dermatitis: Summary of a report for the National Eczema Association. J. Investig. Dermatol. 2017, 137, 26–30. [Google Scholar] [CrossRef] [Green Version]
- Chandra, A.; Ray, A.; Senapati, S.; Chatterjee, R. Genetic and epigenetic basis of psoriasis pathogenesis. Mol. Immunol. 2015, 64, 313–323. [Google Scholar] [CrossRef] [PubMed]
- Frykberg, R.G.; Banks, J. Challenges in the treatment of chronic wounds. Adv. Wound Care 2015, 4, 560–582. [Google Scholar] [CrossRef] [Green Version]
- Kaddoura, I.; Abu-Sittah, G.; Ibrahim, A.; Karamanoukian, R.; Papazian, N. Burn injury: Review of pathophysiology and therapeutic modalities in major burns. Ann. Burn. Fire Disasters 2017, 30, 95. [Google Scholar]
- Hall, T.J.; Villapún, V.M.; Addison, O.; Webber, M.A.; Lowther, M.; Louth, S.E.; Mountcastle, S.E.; Brunet, M.Y.; Cox, S.C. A call for action to the biomaterial community to tackle antimicrobial resistance. Biomater. Sci. 2020, 8, 4951–4974. [Google Scholar] [CrossRef]
- Ejtahed, H.-S.; Hasani-Ranjbar, S.; Siadat, S.D.; Larijani, B. The most important challenges ahead of microbiome pattern in the post era of the COVID-19 pandemic. J. Diabetes Metab. Disord. 2020, 19, 2031–2033. [Google Scholar] [CrossRef]
- Proksch, E. pH in nature, humans and skin. J. Dermatol. 2018, 45, 1044–1052. [Google Scholar] [CrossRef]
- Da Silva, N.T.; Quintana, H.T.; Bortolin, J.A.; Ribeiro, D.A.; de Oliveira, F. Burn injury induces skeletal muscle degeneration, inflammatory host response, and oxidative stress in wistar rats. J. Burn. Care Res. 2015, 36, 428–433. [Google Scholar] [CrossRef] [PubMed]
- Dunnill, C.; Patton, T.; Brennan, J.; Barrett, J.; Dryden, M.; Cooke, J.; Leaper, D.; Georgopoulos, N.T. Reactive oxygen species (ROS) and wound healing: The functional role of ROS and emerging ROS-modulating technologies for augmentation of the healing process. Int. Wound J. 2017, 14, 89–96. [Google Scholar] [CrossRef]
- Todke, P.; Shah, V.H. Psoriasis: Implication to disease and therapeutic strategies, with an emphasis on drug delivery approaches. Int. J. Dermatol. 2018, 57, 1387–1402. [Google Scholar] [CrossRef]
- Hon, K.L.; Leung, A.K.; Barankin, B. Barrier repair therapy in atopic dermatitis: An overview. Am. J. Clin. Dermatol. 2013, 14, 389–399. [Google Scholar] [CrossRef] [PubMed]
- Nobili, S.; Lippi, D.; Witort, E.; Donnini, M.; Bausi, L.; Mini, E.; Capaccioli, S. Natural compounds for cancer treatment and prevention. Pharmacol. Res. 2009, 59, 365–378. [Google Scholar] [CrossRef] [PubMed]
- Tundis, R.; Loizzo, M.; Bonesi, M.; Menichini, F. Potential role of natural compounds against skin aging. Curr. Med. Chem. 2015, 22, 1515–1538. [Google Scholar] [CrossRef]
- Marrelli, M.; Menichini, G.; Provenzano, E.; Conforti, F. Applications of natural compounds in the photodynamic therapy of skin cancer. Curr. Med. Chem. 2014, 21, 1371–1390. [Google Scholar] [CrossRef]
- Sychrová, A.; Koláriková, I.; Žemlička, M.; Šmejkal, K. Natural compounds with dual antimicrobial and anti-inflammatory effects. Phytochem. Rev. 2020, 19, 1471–1502. [Google Scholar] [CrossRef]
- Yong, H.; Liu, Y.; Yun, D.; Zong, S.; Jin, C.; Liu, J. Chitosan films functionalized with different hydroxycinnamic acids: Preparation, characterization and application for pork preservation. Foods 2021, 10, 536. [Google Scholar] [CrossRef] [PubMed]
- Taofiq, O.; González-Paramás, A.M.; Barreiro, M.F.; Ferreira, I.C. Hydroxycinnamic acids and their derivatives: Cosmeceutical significance, challenges and future perspectives, a review. Molecules 2017, 22, 281. [Google Scholar] [CrossRef]
- Ou, S.; Kwok, K.C. Ferulic acid: Pharmaceutical functions, preparation and applications in foods. J. Sci. Food Agric. 2004, 84, 1261–1269. [Google Scholar] [CrossRef]
- Adefegha, S.A. Functional foods and nutraceuticals as dietary intervention in chronic diseases; novel perspectives for health promotion and disease prevention. J. Diet. Suppl. 2018, 15, 977–1009. [Google Scholar] [CrossRef]
- Wu, D.; Zhou, J.; Creyer, M.N.; Yim, W.; Chen, Z.; Messersmith, P.B.; Jokerst, J.V. Phenolic-enabled nanotechnology: Versatile particle engineering for biomedicine. Chem. Soc. Rev. 2021, 50, 4432–4483. [Google Scholar] [CrossRef] [PubMed]
- Li, Z.; Jiang, H.; Xu, C.; Gu, L. A review: Using nanoparticles to enhance absorption and bioavailability of phenolic phytochemicals. Food Hydrocoll. 2015, 43, 153–164. [Google Scholar] [CrossRef]
- Peña-Torres, E.F.; González-Ríos, H.; Avendaño-Reyes, L.; Valenzuela-Grijalva, N.V.; Pinelli-Saavedra, A.; Muhlia-Almazán, A.; Peña-Ramos, E.A. Hydroxycinnamic acids in animal production: Pharmacokinetics, pharmacodynamics and growth promoting effects. Review. Rev. Mex. Cienc. Pecu. 2019, 10, 391–415. [Google Scholar] [CrossRef]
- El-Seedi, H.R.; El-Said, A.M.; Khalifa, S.A.; Goransson, U.; Bohlin, L.; Borg-Karlson, A.-K.; Verpoorte, R. Biosynthesis, natural sources, dietary intake, pharmacokinetic properties, and biological activities of hydroxycinnamic acids. J. Agric. Food Chem. 2012, 60, 10877–10895. [Google Scholar] [CrossRef] [PubMed]
- Coman, V.; Vodnar, D.C. Hydroxycinnamic acids and human health: Recent advances. J. Sci. Food Agric. 2020, 100, 483–499. [Google Scholar] [CrossRef]
- Adisakwattana, S. Cinnamic acid and its derivatives: Mechanisms for prevention and management of diabetes and its complications. Nutrients 2017, 9, 163. [Google Scholar] [CrossRef] [Green Version]
- Neelam; Khatkar, A.; Sharma, K.K. Phenylpropanoids and its derivatives: Biological activities and its role in food, pharmaceutical and cosmetic industries. Crit. Rev. Food Sci. Nutr. 2020, 60, 2655–2675. [Google Scholar] [CrossRef]
- Gunia-Krzyżak, A.; Słoczyńska, K.; Popiół, J.; Koczurkiewicz, P.; Marona, H.; Pękala, E. Cinnamic acid derivatives in cosmetics: Current use and future prospects. Int. J. Cosmet. Sci. 2018, 40, 356–366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Singh, N.; Rao, A.S.; Nandal, A.; Kumar, S.; Yadav, S.S.; Ganaie, S.A.; Narasimhan, B. Phytochemical and pharmacological review of Cinnamomum verum J. Presl-a versatile spice used in food and nutrition. Food Chem. 2021, 338, 127773. [Google Scholar] [CrossRef] [PubMed]
- Forero-Doria, O.; Araya-Maturana, R.; Barrientos-Retamal, A.; Morales-Quintana, L.; Guzmán, L. N-alkylimidazolium salts functionalized with p-coumaric and cinnamic acid: A study of their antimicrobial and antibiofilm effects. Molecules 2019, 24, 3484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ruwizhi, N.; Aderibigbe, B.A. Cinnamic Acid Derivatives and Their Biological Efficacy. Int. J. Mol. Sci. 2020, 21, 5712. [Google Scholar] [CrossRef]
- Pontiki, E.; Hadjipavlou-Litina, D. Multi-target cinnamic acids for oxidative stress and inflammation: Design, synthesis, biological evaluation and modeling studies. Molecules 2019, 24, 12. [Google Scholar] [CrossRef] [Green Version]
- Adisakwattana, S.; Moonsan, P.; Yibchok-Anun, S. Insulin-releasing properties of a series of cinnamic acid derivatives in vitro and in vivo. J. Agric. Food Chem. 2008, 56, 7838–7844. [Google Scholar] [CrossRef]
- Hafizur, R.M.; Hameed, A.; Shukrana, M.; Raza, S.A.; Chishti, S.; Kabir, N.; Siddiqui, R.A. Cinnamic acid exerts anti-diabetic activity by improving glucose tolerance in vivo and by stimulating insulin secretion in vitro. Phytomedicine 2015, 22, 297–300. [Google Scholar] [CrossRef]
- Zhu, R.; Liu, H.; Liu, C.; Wang, L.; Ma, R.; Chen, B.; Li, L.; Niu, J.; Fu, M.; Zhang, D. Cinnamaldehyde in diabetes: A review of pharmacology, pharmacokinetics and safety. Pharmacol. Res. 2017, 122, 78–89. [Google Scholar] [CrossRef] [PubMed]
- Kong, Y.H.; Jo, Y.O.; Cho, C.-W.; Son, D.; Park, S.; Rho, J.; Choi, S.Y. Inhibitory effects of cinnamic acid on melanin biosynthesis in skin. Biol. Pharm. Bull. 2008, 31, 946–948. [Google Scholar] [CrossRef] [Green Version]
- Mathew, S.; Abraham, T.E. Ferulic acid: An antioxidant found naturally in plant cell walls and feruloyl esterases involved in its release and their applications. Crit. Rev. Biotechnol. 2004, 24, 59–83. [Google Scholar] [CrossRef]
- Faulds, C.B.; Williamson, G. The role of hydroxycinnamates in the plant cell wall. J. Sci. Food Agric. 1999, 79, 393–395. [Google Scholar] [CrossRef]
- Contardi, M.; Kossyvaki, D.; Picone, P.; Summa, M.; Guo, X.; Heredia-Guerrero, J.A.; Giacomazza, D.; Carzino, R.; Goldoni, L.; Scoponi, G. Electrospun Polyvinylpyrrolidone (PVP) hydrogels containing hydroxycinnamic acid derivatives as potential wound dressings. Chem. Eng. J. 2021, 409, 128144. [Google Scholar] [CrossRef]
- Sharma, S.; Jaiswal, A.K.; Duffy, B.; Jaiswal, S. Ferulic acid incorporated active films based on poly (lactide)/poly (butylene adipate-co-terephthalate) blend for food packaging. Food Packag. Shelf Life 2020, 24, 100491. [Google Scholar] [CrossRef]
- Ou, S.; Wang, Y.; Tang, S.; Huang, C.; Jackson, M.G. Role of ferulic acid in preparing edible films from soy protein isolate. J. Food Eng. 2005, 70, 205–210. [Google Scholar] [CrossRef]
- Patzke, H.; Schieber, A. Growth-inhibitory activity of phenolic compounds applied in an emulsifiable concentrate-ferulic acid as a natural pesticide against Botrytis cinerea. Food Res. Int. 2018, 113, 18–23. [Google Scholar] [CrossRef]
- Han, H.S.; Seol, H.; Kang, D.H.; Ahmed, M.S.; You, J.-M.; Jeon, S. Electrochemical oxidation and determination of dopamine in the presence of AA using ferulic acid functionalized electrochemically reduced graphene. Sens. Actuators B Chem. 2014, 204, 289–296. [Google Scholar] [CrossRef]
- Picone, P.; Bondi, M.L.; Picone, P.; Bondi, M.L.; Montana, G.; Bruno, A.; Pitarresi, G.; Giammona, G.; Di Carlo, M. Ferulic acid inhibits oxidative stress and cell death induced by Ab oligomers: Improved delivery by solid lipid nanoparticles. Free Radic. Res. 2009, 43, 1133–1145. [Google Scholar] [CrossRef]
- Hassanzadeh, P.; Arbabi, E.; Atyabi, F.; Dinarvand, R. Ferulic acid exhibits antiepileptogenic effect and prevents oxidative stress and cognitive impairment in the kindling model of epilepsy. Life Sci. 2017, 179, 9–14. [Google Scholar] [CrossRef] [PubMed]
- Nankar, R.; Prabhakar, P.; Doble, M. Hybrid drug combination: Combination of ferulic acid and metformin as anti-diabetic therapy. Phytomedicine 2017, 37, 10–13. [Google Scholar] [CrossRef]
- Wang, S.; Gao, Z.; Chen, X.; Lian, X.; Zhu, H.; Zheng, J.; Sun, L. The anticoagulant ability of ferulic acid and its applications for improving the blood compatibility of silk fibroin. Biomed. Mater. 2008, 3, 044106. [Google Scholar] [CrossRef]
- Zhang, E.; Shen, F. Blood compatibility of a ferulic acid (FA)-eluting PHBHHx system for biodegradable magnesium stent application. Mater. Sci. Eng. C 2015, 52, 37–45. [Google Scholar] [CrossRef]
- Mancuso, C.; Santangelo, R. Ferulic acid: Pharmacological and toxicological aspects. Food Chem. Toxicol. 2014, 65, 185–195. [Google Scholar] [CrossRef] [PubMed]
- Zheng, Y.; You, X.; Guan, S.; Huang, J.; Wang, L.; Zhang, J.; Wu, J. Poly (ferulic acid) with an anticancer effect as a drug nanocarrier for enhanced colon cancer therapy. Adv. Funct. Mater. 2019, 29, 1808646. [Google Scholar] [CrossRef]
- Fetoni, A.R.; Mancuso, C.; Eramo, S.L.M.; Ralli, M.; Piacentini, R.; Barone, E.; Paludetti, G.; Troiani, D. In vivo protective effect of ferulic acid against noise-induced hearing loss in the guinea-pig. Neuroscience 2010, 169, 1575–1588. [Google Scholar] [CrossRef] [PubMed]
- Tsai, C.-Y.; Woung, L.-C.; Yen, J.-C.; Tseng, P.-C.; Chiou, S.-H.; Sung, Y.-J.; Liu, K.-T.; Cheng, Y.-H. Thermosensitive chitosan-based hydrogels for sustained release of ferulic acid on corneal wound healing. Carbohydr. Polym. 2016, 135, 308–315. [Google Scholar] [CrossRef]
- Grimaudo, M.A.; Concheiro, A.; Alvarez-Lorenzo, C. Crosslinked Hyaluronan Electrospun Nanofibers for Ferulic Acid Ocular Delivery. Pharmaceutics 2020, 12, 274. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Contardi, M.; Montano, S.; Liguori, G.; Heredia-Guerrero, J.A.; Galli, P.; Athanassiou, A.; Bayer, I.S. treatment of coral Wounds by combining an Antiseptic Bilayer film and an injectable Antioxidant Biopolymer. Sci. Rep. 2020, 10, 1–10. [Google Scholar] [CrossRef]
- Ziyatdinova, G.; Kozlova, E.; Budnikov, H. Selective electrochemical sensor based on the electropolymerized p-coumaric acid for the direct determination of l-cysteine. Electrochim. Acta 2018, 270, 369–377. [Google Scholar] [CrossRef]
- Esakkimuthu, S.; Krishnamurthy, V.; Wang, S.; Hu, X.; Swaminathan, K.; Abomohra, A.E.-F. Application of p-coumaric acid for extraordinary lipid production in Tetradesmus obliquus: A sustainable approach towards enhanced biodiesel production. Renew. Energy 2020, 157, 368–376. [Google Scholar] [CrossRef]
- Rodriguez, A.; Kildegaard, K.R.; Li, M.; Borodina, I.; Nielsen, J. Establishment of a yeast platform strain for production of p-coumaric acid through metabolic engineering of aromatic amino acid biosynthesis. Metab. Eng. 2015, 31, 181–188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Boo, Y.C. p-Coumaric acid as an active ingredient in cosmetics: A review focusing on its antimelanogenic effects. Antioxidants 2019, 8, 275. [Google Scholar] [CrossRef] [Green Version]
- Song, K.; An, S.M.; Kim, M.; Koh, J.-S.; Boo, Y.C. Comparison of the antimelanogenic effects of p-coumaric acid and its methyl ester and their skin permeabilities. J. Dermatol. Sci. 2011, 63, 17–22. [Google Scholar] [CrossRef]
- Guglielmi, F.; Luceri, C.; Giovannelli, L.; Dolara, P.; Lodovici, M. Effect of 4-coumaric and 3, 4-dihydroxybenzoic acid on oxidative DNA damage in rat colonic mucosa. Br. J. Nutr. 2003, 89, 581–587. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Boeing, T.; Costa, P.; Venzon, L.; Meurer, M.; Mariano, L.N.B.; França, T.C.S.; Gouveia, L.; de Bassi, A.C.; Steimbach, V.; de Souza, P. Gastric healing effect of p-coumaric acid isolated from Baccharis dracunculifolia DC on animal model. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2021, 394, 49–57. [Google Scholar] [CrossRef] [PubMed]
- Yeh, C.-T.; Ching, L.-C.; Yen, G.-C. Inducing gene expression of cardiac antioxidant enzymes by dietary phenolic acids in rats. J. Nutr. Biochem. 2009, 20, 163–171. [Google Scholar] [CrossRef] [PubMed]
- Abdel-Moneim, A.; Abd El-Twab, S.M.; Yousef, A.I.; Reheim, E.S.A.; Ashour, M.B. Modulation of hyperglycemia and dyslipidemia in experimental type 2 diabetes by gallic acid and p-coumaric acid: The role of adipocytokines and PPARγ. Biomed. Pharmacother. 2018, 105, 1091–1097. [Google Scholar] [CrossRef]
- Lopes, S.P.; Yepes, L.M.; Pérez-Castillo, Y.; Robledo, S.M.; de Sousa, D.P. Alkyl and Aryl Derivatives Based on p-Coumaric Acid Modification and Inhibitory Action against Leishmania braziliensis and Plasmodium falciparum. Molecules 2020, 25, 3178. [Google Scholar] [CrossRef] [PubMed]
- Zhang, M.; Zhou, J.; Wang, L.; Li, B.; Guo, J.; Guan, X.; Han, Q.; Zhang, H. Caffeic acid reduces cutaneous tumor necrosis factor alpha (TNF-α), IL-6 and IL-1β levels and ameliorates skin edema in acute and chronic model of cutaneous inflammation in mice. Biol. Pharm. Bull. 2014, 37, 347–354. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Magnani, C.; Isaac, V.L.B.; Correa, M.A.; Salgado, H.R.N. Caffeic acid: A review of its potential use in medications and cosmetics. Analytical Methods 2014, 6, 3203–3210. [Google Scholar] [CrossRef]
- Działo, M.; Mierziak, J.; Korzun, U.; Preisner, M.; Szopa, J.; Kulma, A. The potential of plant phenolics in prevention and therapy of skin disorders. Int. J.Mol. Sci. 2016, 17, 160. [Google Scholar] [CrossRef] [Green Version]
- Choi, H.G.; Tran, P.T.; Lee, J.-H.; Min, B.S.; Kim, J.A. Anti-inflammatory activity of caffeic acid derivatives isolated from the roots of Salvia miltiorrhiza Bunge. Arch. Pharmacal Res. 2018, 41, 64–70. [Google Scholar] [CrossRef]
- Dimitris, D.; Ekaterina-Michaela, T.; Christina, K.; Ioannis, S.; Ioanna, S.K.; Aggeliki, L.; Sophia, H.; Michael, R.; Helen, S. Melissa officinalis ssp. altissima extracts: A therapeutic approach targeting psoriasis in mice. J. Ethnopharmacol. 2020, 246, 112208. [Google Scholar] [CrossRef] [PubMed]
- Sundarrajan, S.; Arumugam, M. A systems pharmacology perspective to decipher the mechanism of action of Parangichakkai chooranam, a Siddha formulation for the treatment of psoriasis. Biomed. Pharmacother. 2017, 88, 74–86. [Google Scholar] [CrossRef]
- Dudonné, S.; Poupard, P.; Coutiere, P.; Woillez, M.; Richard, T.; Mérillon, J.-M.; Vitrac, X. Phenolic composition and antioxidant properties of poplar bud (Populus nigra) extract: Individual antioxidant contribution of phenolics and transcriptional effect on skin aging. J. Agric. Food Chem. 2011, 59, 4527–4536. [Google Scholar] [CrossRef] [PubMed]
- Im, K.; Lee, J.Y.; Byeon, H.; Hwang, K.W.; Kang, W.; Whang, W.K.; Min, H. In Vitro antioxidative and anti-inflammatory activities of the ethanol extract of eggplant (Solanum melongena) stalks in macrophage RAW 264.7 cells. Food Agric. Immunol. 2016, 27, 758–771. [Google Scholar] [CrossRef] [Green Version]
- Heidari, M.; Bahramsoltani, R.; Abdolghaffari, A.H.; Rahimi, R.; Esfandyari, M.; Baeeri, M.; Hassanzadeh, G.; Abdollahi, M.; Farzaei, M.H. Efficacy of topical application of standardized extract of Tragopogon graminifolius in the healing process of experimental burn wounds. J. Tradit. Complement. Med. 2019, 9, 54–59. [Google Scholar] [CrossRef]
- Juneja, K.; Mishra, R.; Chauhan, S.; Gupta, S.; Roy, P.; Sircar, D. Metabolite profiling and wound-healing activity of Boerhavia diffusa leaf extracts using in vitro and in vivo models. J. Tradit. Complement. Med. 2020, 10, 52–59. [Google Scholar] [CrossRef] [PubMed]
- Seo, S.H.; Lee, S.H.; Cha, P.H.; Kim, M.Y.; Min, D.S.; Choi, K.Y. Polygonum aviculare L. and its active compounds, quercitrin hydrate, caffeic acid, and rutin, activate the Wnt/β-catenin pathway and induce cutaneous wound healing. Phytother. Res. 2016, 30, 848–854. [Google Scholar] [CrossRef]
- Song, Y.; Zeng, R.; Hu, L.; Maffucci, K.G.; Ren, X.; Qu, Y. In vivo wound healing and in vitro antioxidant activities of Bletilla striata phenolic extracts. Biomed. Pharmacother. 2017, 93, 451–461. [Google Scholar] [CrossRef] [PubMed]
- Liu, J.; Wang, X.; Bai, R.; Zhang, N.; Kan, J.; Jin, C. Synthesis, characterization, and antioxidant activity of caffeic-acid-grafted corn starch. Starch Stärke 2018, 70, 1700141. [Google Scholar] [CrossRef]
- Alevizou, E.I.; Voutsas, E.C. Solubilities of p-coumaric and caffeic acid in ionic liquids and organic solvents. J. Chem. Thermodyn. 2013, 62, 69–78. [Google Scholar] [CrossRef]
- Rodrigues, J.; Araújo, R.; Prather, K.L.; Kluskens, L.; Rodrigues, L. Heterologous production of caffeic acid from tyrosine in Escherichia coli. Enzym. Microb. Technol. 2015, 71, 36–44. [Google Scholar] [CrossRef] [Green Version]
- Yu, S.-H.; Hsieh, H.-Y.; Pang, J.-C.; Tang, D.-W.; Shih, C.-M.; Tsai, M.-L.; Tsai, Y.-C.; Mi, F.-L. Active films from water-soluble chitosan/cellulose composites incorporating releasable caffeic acid for inhibition of lipid oxidation in fish oil emulsions. Food Hydrocoll. 2013, 32, 9–19. [Google Scholar] [CrossRef]
- Lee, P.; Ward, K.; Tschulik, K.; Chapman, G.; Compton, R. Electrochemical detection of glutathione using a poly (caffeic acid) nanocarbon composite modified electrode. Electroanalysis 2014, 26, 366–373. [Google Scholar] [CrossRef]
- Damasceno, S.S.; Santos, N.A.; Santos, I.M.; Souza, A.L.; Souza, A.G.; Queiroz, N. Caffeic and ferulic acids: An investigation of the effect of antioxidants on the stability of soybean biodiesel during storage. Fuel 2013, 107, 641–646. [Google Scholar] [CrossRef]
- Utsunomiya, H.; Ichinose, M.; Ikeda, K.; Uozaki, M.; Morishita, J.; Kuwahara, T.; Koyama, A.H.; Yamasaki, H. Inhibition by caffeic acid of the influenza A virus multiplication in vitro. Int. J. Mol. Med. 2014, 34, 1020–1024. [Google Scholar] [CrossRef] [Green Version]
- Srinivasulu, C.; Ramgopal, M.; Ramanjaneyulu, G.; Anuradha, C.; Kumar, C.S. Syringic acid (SA)‒a review of its occurrence, biosynthesis, pharmacological and industrial importance. Biomed. Pharmacother. 2018, 108, 547–557. [Google Scholar] [CrossRef] [PubMed]
- Wang, Z.; Zhang, J.; Wu, F.; Zhou, X. Changes in rhizosphere microbial communities in potted cucumber seedlings treated with syringic acid. PLoS ONE 2018, 13, e0200007. [Google Scholar] [CrossRef] [PubMed]
- Attanasio, A.; Diano, N.; Grano, V.; Sicuranza, S.; Rossi, S.; Bencivenga, U.; Fraconte, L.; Martino, S.D.; Canciglia, P.; Mita, D.G. Nonisothermal bioreactors in the treatment of vegetation waters from olive oil: Laccase versus syringic acid as bioremediation model. Biotechnol. Prog. 2005, 21, 806–815. [Google Scholar] [CrossRef]
- Gimeno, O.; Fernandez, L.A.; Carbajo, M.; Beltran, F.; Rivas, J. Photocatalytic ozonation of phenolic wastewaters: Syringic acid, tyrosol and gallic acid. J. Environ. Sci. Health Part A 2007, 43, 61–69. [Google Scholar] [CrossRef]
- Muthukumaran, J.; Srinivasan, S.; Venkatesan, R.S.; Ramachandran, V.; Muruganathan, U. Syringic acid, a novel natural phenolic acid, normalizes hyperglycemia with special reference to glycoprotein components in experimental diabetic rats. J. Acute Dis. 2013, 2, 304–309. [Google Scholar] [CrossRef] [Green Version]
- Li, Y.; Zhang, L.; Wang, X.; Wu, W.; Qin, R. Effect of Syringic acid on antioxidant biomarkers and associated inflammatory markers in mice model of asthma. Drug Dev. Res. 2019, 80, 253–261. [Google Scholar] [CrossRef]
- Srinivasan, S.; Muthukumaran, J.; Muruganathan, U.; Venkatesan, R.S.; Jalaludeen, A.M. Antihyperglycemic effect of syringic acid on attenuating the key enzymes of carbohydrate metabolism in experimental diabetic rats. Biomed. Prev. Nutr. 2014, 4, 595–602. [Google Scholar] [CrossRef]
- Karamac, M.; Kosiñska, A.; Pegg, R.B. Comparison of radical-scavenging activities for selected phenolic acids. Pol. J. Food Nutr. Sci. 2005, 14, 165–170. [Google Scholar]
- Abaza, M.-S.; Al-Attiyah, R.A.; Bhardwaj, R.; Abbadi, G.; Koyippally, M.; Afzal, M. Syringic acid from Tamarix aucheriana possesses antimitogenic and chemo-sensitizing activities in human colorectal cancer cells. Pharm. Biol. 2013, 51, 1110–1124. [Google Scholar] [CrossRef] [Green Version]
- Zhang, L.; Liu, R.; Niu, W. Phytochemical and antiproliferative activity of proso millet. PLoS ONE 2014, 9, e104058. [Google Scholar] [CrossRef] [Green Version]
- Cheemanapalli, S.; Anuradha, M.C.; Madhusudhana, P.; Mahesh, M.; Raghavendra, B.P.; Kumar, S.C. Exploring the binding affinity of novel syringic acid analogues and critical determinants of selectivity as potent proteasome inhibitors. Anti Cancer Agents Med. Chem. 2016, 16, 1496–1510. [Google Scholar] [CrossRef]
- Ghitescu, R.-E.; Popa, A.-M.; Popa, V.I.; Rossi, R.M.; Fortunato, G. Encapsulation of polyphenols into pHEMA e-spun fibers and determination of their antioxidant activities. Int. J. Pharm. 2015, 494, 278–287. [Google Scholar] [CrossRef]
- Vishnu, K.; Chatterjee, N.S.; Ajeeshkumar, K.; Lekshmi, R.; Tejpal, C.; Mathew, S.; Ravishankar, C. Microencapsulation of sardine oil: Application of vanillic acid grafted chitosan as a bio-functional wall material. Carbohydr. Polym. 2017, 174, 540–548. [Google Scholar] [CrossRef]
- Asha, K.; Ajeeshkumar, K.; Chatterjee, N.S.; Anandan, R.; Mathew, S. Microencapsulation of β-carotene with vanillic acid grafted chitosan improves stability and glutathione content in rats. Asclepius 2018, 1, 1–7. [Google Scholar]
- Da Silva, L.; Silva, F.; Kubota, L.; Lopes, C.; Lima, P.; Costa, E.; Júnior, W.P.; Goulart, M. Amperometric sensor based on carbon nanotubes and electropolymerized vanillic acid for simultaneous determination of ascorbic acid, dopamine, and uric acid. J. Solid State Electrochem. 2016, 20, 2389–2393. [Google Scholar] [CrossRef]
- Chou, T.H.; Ding, H.Y.; Hung, W.J.; Liang, C.H. Antioxidative characteristics and inhibition of α-melanocyte-stimulating hormone-stimulated melanogenesis of vanillin and vanillic acid from Origanum vulgare. Exp. Dermatol. 2010, 19, 742–750. [Google Scholar] [CrossRef]
- Kumar, S.; Prahalathan, P.; Raja, B. Antihypertensive and antioxidant potential of vanillic acid, a phenolic compound in L-NAME-induced hypertensive rats: A dose-dependence study. Redox Rep. 2011, 16, 208–215. [Google Scholar] [CrossRef]
- Kumar, S.; Prahalathan, P.; Raja, B. Vanillic acid: A potential inhibitor of cardiac and aortic wall remodeling in l-NAME induced hypertension through upregulation of endothelial nitric oxide synthase. Environ. Toxicol. Pharmacol. 2014, 38, 643–652. [Google Scholar] [CrossRef]
- Delaquis, P.; Stanich, K.; Toivonen, P. Effect of pH on the inhibition of Listeria spp. by vanillin and vanillic acid. J. Food Prot. 2005, 68, 1472–1476. [Google Scholar] [CrossRef] [PubMed]
- Prince, P.S.M.; Rajakumar, S.; Dhanasekar, K. Protective effects of vanillic acid on electrocardiogram, lipid peroxidation, antioxidants, proinflammatory markers and histopathology in isoproterenol induced cardiotoxic rats. Eur. J. Pharmacol. 2011, 668, 233–240. [Google Scholar] [CrossRef]
- Velli, S.K.; Sundaram, J.; Murugan, M.; Balaraman, G.; Thiruvengadam, D. Protective effect of vanillic acid against benzo (a) pyrene induced lung cancer in Swiss albino mice. J. Biochem. Mol. Toxicol. 2019, 33, e22382. [Google Scholar] [CrossRef]
- Ji, G.; Sun, R.; Hu, H.; Xu, F.; Yu, X.; Veeraraghavan, V.P.; Mohan, S.K.; Chi, X. Vannilic acid ameliorates hyperglycemia-induced oxidative stress and inflammation in streptozotocin-induced diabetic rats. J. King Saud Univ. Sci. 2020, 32, 2905–2911. [Google Scholar] [CrossRef]
- Ghasemian, M.; Owlia, S.; Owlia, M.B. Review of Anti-Inflammatory Herbal Medicines. Adv. Pharmacol. Sci. 2016, 2016, 9130979. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ge, L.; Zhu, M.; Li, X.; Xu, Y.; Ma, X.; Shi, R.; Li, D.; Mu, C. Development of active rosmarinic acid-gelatin biodegradable films with antioxidant and long-term antibacterial activities. Food Hydrocoll. 2018, 83, 308–316. [Google Scholar] [CrossRef]
- Marchev, A.S.; Vasileva, L.V.; Amirova, K.M.; Savova, M.S.; Koycheva, I.K.; Balcheva-Sivenova, Z.P.; Vasileva, S.M.; Georgiev, M.I. Rosmarinic acid-From bench to valuable applications in food industry. Trends Food Sci. Technol. 2021. [Google Scholar] [CrossRef]
- Vatankhah, E.; Hamedi, S.; Ramezani, O. Surfactant-assisted incorporation of rosmarinic acid into electrosprayed poly (lactic-co-glycolic acid) microparticles with potential for cosmetic and pharmaceutical applications. Polym. Test. 2020, 81, 106180. [Google Scholar] [CrossRef]
- Kim, H.J.; Kim, T.H.; Kang, K.C.; Pyo, H.B.; Jeong, H.H. Microencapsulation of rosmarinic acid using polycaprolactone and various surfactants. Int. J. Cosmet. Sci. 2010, 32, 185–191. [Google Scholar] [CrossRef]
- Amoah, S.K.; Sandjo, L.P.; Kratz, J.M.; Biavatti, M.W. Rosmarinic Acid--Pharmaceutical and Clinical Aspects. Planta. Med. 2016, 82, 388–406. [Google Scholar] [CrossRef] [Green Version]
- Nadeem, M.; Imran, M.; Aslam Gondal, T.; Imran, A.; Shahbaz, M.; Muhammad Amir, R.; Wasim Sajid, M.; Batool Qaisrani, T.; Atif, M.; Hussain, G.; et al. Therapeutic Potential of Rosmarinic Acid: A Comprehensive Review. Appl. Sci. 2019, 9, 3139. [Google Scholar] [CrossRef] [Green Version]
- Jiang, W.-L.; Chen, X.-G.; Qu, G.-W.; Yue, X.-D.; Zhu, H.-B.; Tian, J.-W.; Fu, F.-H. Rosmarinic acid protects against experimental sepsis by inhibiting proinflammatory factor release and ameliorating hemodynamics. Shock 2009, 32, 608–613. [Google Scholar] [CrossRef]
- Scheckel, K.A.; Degner, S.C.; Romagnolo, D.F. Rosmarinic acid antagonizes activator protein-1-dependent activation of cyclooxygenase-2 expression in human cancer and nonmalignant cell lines. J. Nutr. 2008, 138, 2098–2105. [Google Scholar] [CrossRef]
- Clifford, M.N. Chlorogenic acids and other cinnamates–nature, occurrence and dietary burden. J. Sci. Food Agric. 1999, 79, 362–372. [Google Scholar] [CrossRef]
- Fazel Nabavi, S.; Tejada, S.; Setzer, N.W.; Gortzi, O.; Sureda, A.; Braidy, N.; Daglia, M.; Manayi, A.; Mohammad Nabavi, S. Chlorogenic acid and mental diseases: From chemistry to medicine. Curr. Neuropharmacol. 2017, 15, 471–479. [Google Scholar] [CrossRef] [Green Version]
- Fu, S.; Wu, C.; Wu, T.; Yu, H.; Yang, S.; Hu, Y. Preparation and characterisation of chlorogenic acid-gelatin: A type of biologically active film for coating preservation. Food Chem. 2017, 221, 657–663. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; Wang, H.; Niu, N.; Chen, Z.; Li, S.; Liu, S.-X.; Li, J. Fluorescent Poly (vinyl alcohol) Films Containing Chlorogenic Acid Carbon Nanodots for Food Monitoring. ACS Appl. Nano Mater. 2020, 3, 7611–7620. [Google Scholar] [CrossRef]
- Salimi, A.; Hallaj, R.; Ghadermazi, M. Modification of carbon ceramic electrode prepared with sol–gel technique by a thin film of chlorogenic acid: Application to amperometric detection of NADH. Talanta 2005, 65, 888–894. [Google Scholar] [CrossRef]
- Naveed, M.; Hejazi, V.; Abbas, M.; Kamboh, A.A.; Khan, G.J.; Shumzaid, M.; Ahmad, F.; Babazadeh, D.; FangFang, X.; Modarresi-Ghazani, F. Chlorogenic acid (CGA): A pharmacological review and call for further research. Biomed. Pharmacother. 2018, 97, 67–74. [Google Scholar] [CrossRef]
- Yun, N.; Kang, J.-W.; Lee, S.-M. Protective effects of chlorogenic acid against ischemia/reperfusion injury in rat liver: Molecular evidence of its antioxidant and anti-inflammatory properties. J. Nutr. Biochem. 2012, 23, 1249–1255. [Google Scholar] [CrossRef] [PubMed]
- Suzuki, A.; Yamamoto, N.; Jokura, H.; Yamamoto, M.; Fujii, A.; Tokimitsu, I.; Saito, I. Chlorogenic acid attenuates hypertension and improves endothelial function in spontaneously hypertensive rats. J. Hypertens. 2006, 24, 1065–1073. [Google Scholar] [CrossRef]
- Lu, H.; Tian, Z.; Cui, Y.; Liu, Z.; Ma, X. Chlorogenic acid: A comprehensive review of the dietary sources, processing effects, bioavailability, beneficial properties, mechanisms of action, and future directions. Compr. Rev. Food Sci. Food Saf. 2020, 19, 3130–3158. [Google Scholar] [CrossRef]
- Shin, H.S.; Satsu, H.; Bae, M.-J.; Zhao, Z.; Ogiwara, H.; Totsuka, M.; Shimizu, M. Anti-inflammatory effect of chlorogenic acid on the IL-8 production in Caco-2 cells and the dextran sulphate sodium-induced colitis symptoms in C57BL/6 mice. Food Chem. 2015, 168, 167–175. [Google Scholar] [CrossRef]
- Feng, R.; Lu, Y.; Bowman, L.L.; Qian, Y.; Castranova, V.; Ding, M. Inhibition of activator protein-1, NF-κB, and MAPKs and induction of phase 2 detoxifying enzyme activity by chlorogenic acid. J. Biol. Chem. 2005, 280, 27888–27895. [Google Scholar] [CrossRef] [Green Version]
- Han, G.; Ceilley, R. Chronic wound healing: A review of current management and treatments. Adv. Ther. 2017, 34, 599–610. [Google Scholar] [CrossRef] [Green Version]
- Gurtner, G.C.; Werner, S.; Barrandon, Y.; Longaker, M.T. Wound repair and regeneration. Nature 2008, 453, 314–321. [Google Scholar] [CrossRef]
- Gethin, G. Understanding the inflammatory process in wound healing. Br. J. Community Nurs. 2012, 17, S17–S22. [Google Scholar] [CrossRef]
- Blakytny, R.; Jude, E. The molecular biology of chronic wounds and delayed healing in diabetes. Diabet. Med. 2006, 23, 594–608. [Google Scholar] [CrossRef]
- Rosique, R.G.; Rosique, M.J.; Farina Junior, J.A. Curbing inflammation in skin wound healing: A review. Int. J. Inflamm. 2015, 2015, 316235. [Google Scholar] [CrossRef] [Green Version]
- Tottoli, E.M.; Dorati, R.; Genta, I.; Chiesa, E.; Pisani, S.; Conti, B. Skin wound healing process and new emerging technologies for skin wound care and regeneration. Pharmaceutics 2020, 12, 735. [Google Scholar] [CrossRef]
- Ghaisas, M.M.; Kshirsagar, S.B.; Sahane, R.S. Evaluation of wound healing activity of ferulic acid in diabetic rats. Int. Wound J. 2014, 11, 523–532. [Google Scholar] [CrossRef]
- Ren, J.; Yang, M.; Xu, F.; Chen, J.; Ma, S. Acceleration of wound healing activity with syringic acid in streptozotocin induced diabetic rats. Life Sci. 2019, 233, 116728. [Google Scholar] [CrossRef]
- Koganov, M.M.; Dueva, O.V.; Tsorin, B.L. Activities of plant-derived phenols in a fibroblast cell culture model. J. Nat. Prod. 1999, 62, 481–483. [Google Scholar] [CrossRef]
- Song, H.S.; Park, T.W.; Sohn, U.D.; Shin, Y.K.; Choi, B.C.; Kim, C.J.; Sim, S.S. The effect of caffeic acid on wound healing in skin-incised mice. Korean J. Physiol. Pharmacol. 2008, 12, 343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alexandru, V.; Gaspar, A.; Savin, S.; Toma, A.; Tatia, R.; Gille, E. Phenolic content, antioxidant activity and effect on collagen synthesis of a traditional wound healing polyherbal formula. Studia Univ. Vasile Goldis Arad. Ser. Stiintele Vietii 2015, 25, 41. [Google Scholar]
- Bagdas, D.; Etoz, B.C.; Gul, Z.; Ziyanok, S.; Inan, S.; Turacozen, O.; Gul, N.Y.; Topal, A.; Cinkilic, N.; Tas, S. In vivo systemic chlorogenic acid therapy under diabetic conditions: Wound healing effects and cytotoxicity/genotoxicity profile. Food Chem. Toxicol. 2015, 81, 54–61. [Google Scholar] [CrossRef]
- Bagdas, D.; Gul, N.Y.; Topal, A.; Tas, S.; Ozyigit, M.O.; Cinkilic, N.; Gul, Z.; Etoz, B.C.; Ziyanok, S.; Inan, S. Pharmacologic overview of systemic chlorogenic acid therapy on experimental wound healing. Naunyn Schmiedeberg’s Arch. Pharmacol. 2014, 387, 1101–1116. [Google Scholar] [CrossRef] [PubMed]
- Davis, F.M.; denDekker, A.; Joshi, A.D.; Wolf, S.J.; Audu, C.; Melvin, W.J.; Mangum, K.; Riordan, M.O.; Kunkel, S.L.; Gallagher, K.A. Palmitate-TLR4 signaling regulates the histone demethylase, JMJD3, in macrophages and impairs diabetic wound healing. Eur. J. Immunol. 2020, 50, 1929–1940. [Google Scholar] [CrossRef]
- Jere, S.W.; Abrahamse, H.; Houreld, N.N. The JAK/STAT signaling pathway and photobiomodulation in chronic wound healing. Cytokine Growth Factor Rev. 2017, 38, 73–79. [Google Scholar] [CrossRef]
- Vinaik, R.; Abdullahi, A.; Barayan, D.; Jeschke, M.G. NLRP3 inflammasome activity is required for wound healing after burns. Transl. Res. 2020, 217, 47–60. [Google Scholar] [CrossRef]
- Hiebert, P.; Werner, S. Regulation of Wound Healing by the NRF2 Transcription Factor—More Than Cytoprotection. Int. J. Mol. Sci. 2019, 20, 3856. [Google Scholar] [CrossRef] [Green Version]
- Thuraisingam, T.; Xu, Y.Z.; Eadie, K.; Heravi, M.; Guiot, M.-C.; Greemberg, R.; Gaestel, M.; Radzioch, D. MAPKAPK-2 signaling is critical for cutaneous wound healing. J. Investig. Dermatol. 2010, 130, 278–286. [Google Scholar] [CrossRef] [Green Version]
- Kępa, M.; Miklasińska-Majdanik, M.; Wojtyczka, R.D.; Idzik, D.; Korzeniowski, K.; Smoleń-Dzirba, J.; Wąsik, T.J. Antimicrobial potential of caffeic acid against Staphylococcus aureus clinical strains. BioMed. Res. Int. 2018, 2018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gheibi, N.; Divan Khosroshahi, N.; Habibi, M. Effect of Caffeic Acid and Low-Power Laser Light Co-Exposure on Viability of Pseudomonas aeruginosa. J. Biotechnol. Health Sci. 2015. [Google Scholar] [CrossRef] [Green Version]
- Borges, A.; Ferreira, C.; Saavedra, M.J.; Simões, M. Antibacterial activity and mode of action of ferulic and gallic acids against pathogenic bacteria. Microb. Drug Resist. 2013, 19, 256–265. [Google Scholar] [CrossRef]
- Ignatova, M.; Manolova, N.; Rashkov, I.; Markova, N. Antibacterial and antioxidant electrospun materials from poly (3-hydroxybutyrate) and polyvinylpyrrolidone containing caffeic acid phenethyl ester–“in” and “on” strategies for enhanced solubility. Int. J. Pharm. 2018, 545, 342–356. [Google Scholar] [CrossRef]
- Pinho, E.; Soares, G.; Henriques, M. Evaluation of antibacterial activity of caffeic acid encapsulated by β-cyclodextrins. J. Microencapsul. 2015, 32, 804–810. [Google Scholar] [CrossRef] [Green Version]
- Lou, Z.; Wang, H.; Rao, S.; Sun, J.; Ma, C.; Li, J. p-Coumaric acid kills bacteria through dual damage mechanisms. Food Control 2012, 25, 550–554. [Google Scholar] [CrossRef]
- Monte, J.; Abreu, A.C.; Borges, A.; Simões, L.C.; Simões, M. Antimicrobial activity of selected phytochemicals against Escherichia coli and Staphylococcus aureus and their biofilms. Pathogens 2014, 3, 473–498. [Google Scholar] [CrossRef] [Green Version]
- Kabir, F.; Katayama, S.; Tanji, N.; Nakamura, S. Antimicrobial effects of chlorogenic acid and related compounds. J. Korean Soc. Appl. Biol. Chem. 2014, 57, 359–365. [Google Scholar] [CrossRef]
- Kim, J.-H.; Yu, D.; Eom, S.-H.; Kim, S.-H.; Oh, J.; Jung, W.K.; Kim, Y.-M. Synergistic antibacterial effects of chitosan-caffeic acid conjugate against antibiotic-resistant acne-related bacteria. Mar. Drugs 2017, 15, 167. [Google Scholar] [CrossRef]
- Kfoury, M.; Landy, D.; Auezova, L.; Greige-Gerges, H.; Fourmentin, S. Effect of cyclodextrin complexation on phenylpropanoids’ solubility and antioxidant activity. Beilstein J. Org. Chem. 2014, 10, 2322–2331. [Google Scholar] [CrossRef] [Green Version]
- Shiozawa, R.; Inoue, Y.; Murata, I.; Kanamoto, I. Effect of antioxidant activity of caffeic acid with cyclodextrins using ground mixture method. Asian J. Pharm. Sci. 2018, 13, 24–33. [Google Scholar] [CrossRef] [PubMed]
- Luís, Â.; Silva, F.; Sousa, S.; Duarte, A.P.; Domingues, F. Antistaphylococcal and biofilm inhibitory activities of gallic, caffeic, and chlorogenic acids. Biofouling 2014, 30, 69–79. [Google Scholar] [CrossRef]
- Evers, L.H.; Bhavsar, D.; Mailänder, P. The biology of burn injury. Exp. Dermatol. 2010, 19, 777–783. [Google Scholar] [CrossRef] [PubMed]
- Hseu, Y.-C.; Korivi, M.; Lin, F.-Y.; Li, M.-L.; Lin, R.-W.; Wu, J.-J.; Yang, H.-L. Trans-cinnamic acid attenuates UVA-induced photoaging through inhibition of AP-1 activation and induction of Nrf2-mediated antioxidant genes in human skin fibroblasts. J. Dermatol. Sci. 2018, 90, 123–134. [Google Scholar] [CrossRef] [Green Version]
- Fernando, P.M.D.J.; Piao, M.J.; Kang, K.A.; Ryu, Y.S.; Hewage, S.R.K.M.; Chae, S.W.; Hyun, J.W. Rosmarinic acid attenuates cell damage against UVB radiation-induced oxidative stress via enhancing antioxidant effects in human HaCaT cells. Biomol. Ther. 2016, 24, 75. [Google Scholar] [CrossRef] [Green Version]
- Vostálová, J.; Zdařilová, A.; Svobodová, A. Prunella vulgaris extract and rosmarinic acid prevent UVB-induced DNA damage and oxidative stress in HaCaT keratinocytes. Arch. Dermatol. Res. 2010, 302, 171–181. [Google Scholar] [CrossRef]
- Lembo, S.; Balato, A.; Di Caprio, R.; Cirillo, T.; Giannini, V.; Gasparri, F.; Monfrecola, G. The modulatory effect of ellagic acid and rosmarinic acid on ultraviolet-B-induced cytokine/chemokine gene expression in skin keratinocyte (HaCaT) cells. BioMed. Res. Int. 2014, 2014. [Google Scholar] [CrossRef]
- Hahn, H.J.; Kim, K.B.; An, I.S.; Ahn, K.J.; Han, H.J. Protective effects of rosmarinic acid against hydrogen peroxide-induced cellular senescence and the inflammatory response in normal human dermal fibroblasts. Mol. Med. Rep. 2017, 16, 9763–9769. [Google Scholar] [CrossRef] [Green Version]
- Psotova, J.; Svobodova, A.; Kolarova, H.; Walterova, D. Photoprotective properties of Prunella vulgaris and rosmarinic acid on human keratinocytes. J. Photochem. Photobiol. B Biol. 2006, 84, 167–174. [Google Scholar] [CrossRef]
- Sánchez-Campillo, M.; Gabaldon, J.; Castillo, J.; Benavente-García, O.; Del Baño, M.; Alcaraz, M.; Vicente, V.; Alvarez, N.; Lozano, J. Rosmarinic acid, a photo-protective agent against UV and other ionizing radiations. Food Chem. Toxicol. 2009, 47, 386–392. [Google Scholar] [CrossRef] [PubMed]
- Abraham, S.K.; Sarma, L.; Kesavan, P. Protective effects of chlorogenic acid, curcumin and β-carotene against γ-radiation-induced in vivo chromosomal damage. Mutat. Res. Lett. 1993, 303, 109–112. [Google Scholar] [CrossRef]
- Her, Y.; Lee, T.-K.; Kim, J.D.; Kim, B.; Sim, H.; Lee, J.-C.; Ahn, J.H.; Park, J.H.; Lee, J.-W.; Hong, J. Topical Application of Aronia melanocarpa Extract Rich in Chlorogenic Acid and Rutin Reduces UVB-Induced Skin Damage via Attenuating Collagen Disruption in Mice. Molecules 2020, 25, 4577. [Google Scholar] [CrossRef]
- Ha, S.J.; Lee, J.; Kim, H.; Song, K.-M.; Lee, N.H.; Kim, Y.E.; Lee, H.; Kim, Y.H.; Jung, S.K. Preventive effect of Rhus javanica extract on UVB-induced skin inflammation and photoaging. J. Funct. Foods 2016, 27, 589–599. [Google Scholar] [CrossRef]
- Kwon, K.-R.; Alam, M.B.; Park, J.-H.; Kim, T.-H.; Lee, S.-H. Attenuation of UVB-induced photo-aging by polyphenolic-rich Spatholobus suberectus stem extract via modulation of MAPK/AP-1/MMPs signaling in human keratinocytes. Nutrients 2019, 11, 1341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jeon, J.; Sung, J.; Lee, H.; Kim, Y.; Jeong, H.S.; Lee, J. Protective activity of caffeic acid and sinapic acid against UVB-induced photoaging in human fibroblasts. J. Food Biochem. 2019, 43, e12701. [Google Scholar] [CrossRef]
- Staniforth, V.; Huang, W.-C.; Aravindaram, K.; Yang, N.-S. Ferulic acid, a phenolic phytochemical, inhibits UVB-induced matrix metalloproteinases in mouse skin via posttranslational mechanisms. J. Nutr. Biochem. 2012, 23, 443–451. [Google Scholar] [CrossRef]
- Peres, D.D.A.; Sarruf, F.D.; de Oliveira, C.A.; Velasco, M.V.R.; Baby, A.R. Ferulic acid photoprotective properties in association with UV filters: Multifunctional sunscreen with improved SPF and UVA-PF. J. Photochem. Photobiol. B Biol. 2018, 185, 46–49. [Google Scholar] [CrossRef]
- Ha, S.J.; Lee, J.; Park, J.; Kim, Y.H.; Lee, N.H.; Kim, Y.E.; Song, K.-M.; Chang, P.-S.; Jeong, C.-H.; Jung, S.K. Syringic acid prevents skin carcinogenesis via regulation of NoX and EGFR signaling. Biochem. Pharmacol. 2018, 154, 435–445. [Google Scholar] [CrossRef]
- Choi, E.-J.; Ryu, Y.B.; Tang, Y.; Kim, B.R.; Lee, W.S.; Debnath, T.; Fan, M.; Lee, H.-S.; Kim, E.-K. Effect of cinnamamides on atopic dermatitis through regulation of IL-4 in CD4+ cells. J. Enzym. Inhib. Med. Chem. 2019, 34, 613–619. [Google Scholar] [CrossRef] [Green Version]
- Ong, P.Y.; Leung, D.Y. Bacterial and viral infections in atopic dermatitis: A comprehensive review. Clin. Rev. Allergy Immunol. 2016, 51, 329–337. [Google Scholar] [CrossRef]
- Zhou, Z.; Shi, T.; Hou, J.; Li, M. Ferulic acid alleviates atopic dermatitis-like symptoms in mice via its potent anti-inflammatory effect. Immunopharmacol. Immunotoxicol. 2020, 42, 156–164. [Google Scholar] [CrossRef]
- Lim, K.-M.; Bae, S.; Koo, J.E.; Kim, E.-S.; Bae, O.-N.; Lee, J.Y. Suppression of skin inflammation in keratinocytes and acute/chronic disease models by caffeic acid phenethyl ester. Arch. Dermatol. Res. 2015, 307, 219–227. [Google Scholar] [CrossRef] [PubMed]
- Jang, A.-H.; Kim, T.-H.; Kim, G.-D.; Kim, J.E.; Kim, H.J.; Kim, S.S.; Jin, Y.-H.; Park, Y.S.; Park, C.-S. Rosmarinic acid attenuates 2, 4-dinitrofluorobenzene-induced atopic dermatitis in NC/Nga mice. Int. Immunopharmacol. 2011, 11, 1271–1277. [Google Scholar] [CrossRef] [PubMed]
- Lee, J.; Jung, E.; Koh, J.; Kim, Y.S.; Park, D. Effect of rosmarinic acid on atopic dermatitis. J. Dermatol. 2008, 35, 768–771. [Google Scholar] [CrossRef] [PubMed]
- Cho, K.; Parveen, A.; Kang, M.C.; Subedi, L.; Lee, J.H.; Park, S.Y.; Jin, M.R.; Yoon, H.; Son, Y.K.; Kim, S.Y. Pyrus ussuriensis Maxim. leaves extract ameliorates DNCB-induced atopic dermatitis-like symptoms in NC/Nga mice. Phytomedicine 2018, 48, 76–83. [Google Scholar] [CrossRef]
- Yang, W.S.; Jeong, D.; Yi, Y.-S.; Park, J.G.; Seo, H.; Moh, S.H.; Hong, S.; Cho, J.Y. IRAK1/4-targeted anti-inflammatory action of caffeic acid. Med. Inflamm. 2013, 2013. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Búfalo, M.C.; Ferreira, I.; Costa, G.; Francisco, V.; Liberal, J.; Cruz, M.T.; Lopes, M.C.; Batista, M.T.; Sforcin, J.M. Propolis and its constituent caffeic acid suppress LPS-stimulated pro-inflammatory response by blocking NF-κB and MAPK activation in macrophages. J. Ethnopharmacol. 2013, 149, 84–92. [Google Scholar] [CrossRef] [PubMed]
- Khan, A.Q.; Khan, R.; Qamar, W.; Lateef, A.; Ali, F.; Tahir, M.; Sultana, S. Caffeic acid attenuates 12-O-tetradecanoyl-phorbol-13-acetate (TPA)-induced NF-κB and COX-2 expression in mouse skin: Abrogation of oxidative stress, inflammatory responses and proinflammatory cytokine production. Food Chem. Toxicol. 2012, 50, 175–183. [Google Scholar] [CrossRef]
- Kim, M.-C.; Kim, S.-J.; Kim, D.-S.; Jeon, Y.-D.; Park, S.J.; Lee, H.S.; Um, J.-Y.; Hong, S.-H. Vanillic acid inhibits inflammatory mediators by suppressing NF-κB in lipopolysaccharide-stimulated mouse peritoneal macrophages. Immunopharmacol. Immunotoxicol. 2011, 33, 525–532. [Google Scholar] [CrossRef]
- Calixto-Campos, C.; Carvalho, T.T.; Hohmann, M.S.; Pinho-Ribeiro, F.A.; Fattori, V.; Manchope, M.F.; Zarpelon, A.C.; Baracat, M.M.; Georgetti, S.R.; Casagrande, R. Vanillic acid inhibits inflammatory pain by inhibiting neutrophil recruitment, oxidative stress, cytokine production, and NFκB activation in mice. J. Nat. Prod. 2015, 78, 1799–1808. [Google Scholar] [CrossRef] [PubMed]
- Moustafa, S.F.; Gabr, N.M.; Zaki, J.T.; El Awdan, S.A.; Mina, S.A. The anti-inflammatory, anti-ulcer activities and phytochemical investigation of Cucumis melo L. cv. Ismailawi fruits. Nat. Prod. Res. 2020, 1–5. [Google Scholar] [CrossRef]
- Rocha, J.; Eduardo-Figueira, M.; Barateiro, A.; Fernandes, A.; Brites, D.; Bronze, R.; Duarte, C.M.; Serra, A.T.; Pinto, R.; Freitas, M. Anti-inflammatory effect of rosmarinic acid and an extract of Rosmarinus officinalis in rat models of local and systemic inflammation. Basic Clin. Pharmacol. Toxicol. 2015, 116, 398–413. [Google Scholar] [CrossRef]
- Usha, T.; Middha, S.K.; Bhattacharya, M.; Lokesh, P.; Goyal, A.K. Rosmarinic acid, a new polyphenol from Baccaurea ramiflora Lour. leaf: A probable compound for its anti-inflammatory activity. Antioxidants 2014, 3, 830–842. [Google Scholar] [CrossRef] [PubMed]
- Dos Santos, M.D.; Almeida, M.C.; Lopes, N.P.; De Souza, G.E.P. Evaluation of the anti-inflammatory, analgesic and antipyretic activities of the natural polyphenol chlorogenic acid. Biol. Pharm. Bull. 2006, 29, 2236–2240. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ha, H.; Lee, H.; Seo, C.S.; Lim, H.-S.; Lee, J.K.; Lee, M.-Y.; Shin, H. Artemisia capillaris inhibits atopic dermatitis-like skin lesions in Dermatophagoides farinae-sensitized Nc/Nga mice. BMC Complement. Altern. Med. 2014, 14, 1–10. [Google Scholar] [CrossRef] [Green Version]
- Rapalli, V.K.; Waghule, T.; Gorantla, S.; Dubey, S.K.; Saha, R.N.; Singhvi, G. Psoriasis: Pathological mechanisms, current pharmacological therapies, and emerging drug delivery systems. Drug Discov. Today 2020. [Google Scholar] [CrossRef] [PubMed]
- Saleem, S.; Iqubal, M.K.; Garg, S.; Ali, J.; Baboota, S. Trends in nanotechnology-based delivery systems for dermal targeting of drugs: An enticing approach to offset psoriasis. Expert Opin. Drug Deliv. 2020, 17, 817–838. [Google Scholar] [CrossRef] [PubMed]
- Zhou, M.-W.; Jiang, R.-H.; Kim, K.-D.; Lee, J.-H.; Kim, C.-D.; Yin, W.-T.; Lee, J.-H. Rosmarinic acid inhibits poly (I: C)-induced inflammatory reaction of epidermal keratinocytes. Life Sci. 2016, 155, 189–194. [Google Scholar] [CrossRef]
- Zhang, M.; Li, N.; Cai, R.; Gu, J.; Xie, F.; Wei, H.; Lu, C.; Wu, D. Rosmarinic acid protects mice from imiquimod induced psoriasis-like skin lesions by inhibiting the IL-23/Th17 axis via regulating Jak2/Stat3 signaling pathway. Phytother. Res. 2021. [Google Scholar] [CrossRef]
- Koycheva, I.K.; Vasileva, L.V.; Amirova, K.M.; Marchev, A.S.; Balcheva-Sivenova, Z.P.; Georgiev, M.I. Biotechnologically Produced Lavandula angustifolia Mill. Extract Rich in Rosmarinic Acid Resolves Psoriasis-Related Inflammation Through Janus Kinase/Signal Transducer and Activator of Transcription Signaling. Front. Pharmacol. 2021, 12, 940. [Google Scholar] [CrossRef] [PubMed]
- Lo, H.-Y.; Li, C.-C.; Cheng, H.-M.; Liu, I.-C.; Ho, T.-Y.; Hsiang, C.-Y. Ferulic acid altered IL-17A/IL-17RA interaction and protected against imiquimod-induced psoriasis-like skin injury in mice. Food Chem. Toxicol. 2019, 129, 365–375. [Google Scholar] [CrossRef]
- Cheng, H.-M.; Chen, F.-Y.; Li, C.-C.; Lo, H.-Y.; Liao, Y.-F.; Ho, T.-Y.; Hsiang, C.-Y. Oral administration of vanillin improves imiquimod-induced psoriatic skin inflammation in mice. J. Agric. Food Chem. 2017, 65, 10233–10242. [Google Scholar] [CrossRef]
- Lee, S.Y.; Nam, S.; Hong, I.K.; Kim, H.; Yang, H.; Cho, H.J. Antiproliferation of keratinocytes and alleviation of psoriasis by the ethanol extract of Artemisia capillaris. Phytother. Res. 2018, 32, 923–932. [Google Scholar] [CrossRef]
- Zhang, Y.-T.; Xu, Y.-M.; Zhang, S.-J.; Zhao, J.-H.; Wang, Z.; Xu, D.-Q.; Feng, N.-P. In vivo microdialysis for the evaluation of transfersomes as a novel transdermal delivery vehicle for cinnamic acid. Drug Dev. Ind. Pharm. 2014, 40, 301–307. [Google Scholar] [CrossRef]
- Aitipamula, S.; Das, S. Cocrystal formulations: A case study of topical formulations consisting of ferulic acid cocrystals. Eur. J. Pharm. Biopharm. 2020, 149, 95–104. [Google Scholar] [CrossRef]
- Suzuki, T.; Sasai, A.; Tsujimoto, H.; Yasunaga, T.; Ogawa, N.; Yamamoto, H. Promoting effect of type 17 collagen production by chlorogenic acid using PLGA nanoparticles in the human epidermal keratinocyte cell. J. Drug Deliv. Sci. Technol. 2020, 58, 101624. [Google Scholar] [CrossRef]
- Ammar, N.M.; Hassan, H.A.; Mohammed, M.A.; Serag, A.; Abd El-Alim, S.H.; Elmotasem, H.; El Raey, M.; El Gendy, A.N.; Sobeh, M.; Abdel-Hamid, A.-H.Z. Metabolomic profiling to reveal the therapeutic potency of Posidonia oceanica nanoparticles in diabetic rats. RSC Adv. 2021, 11, 8398–8410. [Google Scholar] [CrossRef]
- Chhabra, P.; Chauhan, G.; Kumar, A. Augmented healing of full thickness chronic excision wound by rosmarinic acid loaded chitosan encapsulated graphene nanopockets. Drug Dev. Ind. Pharm. 2020, 46, 878–888. [Google Scholar] [CrossRef] [PubMed]
- Alberti, T.; Coelho, D.; Voytena, A.; Iacovski, R.; Mazzarino, L.; Maraschin, M.; Veleirinho, B. Effect of propolis nanoparticles on early-stage wound healing in a diabetic noncontractile wound model. Nanotechnol. Adv. Mater. Sci. 2019, 2, 1–10. [Google Scholar]
- Sguizzato, M.; Mariani, P.; Ferrara, F.; Drechsler, M.; Hallan, S.S.; Huang, N.; Simelière, F.; Khunti, N.; Cortesi, R.; Marchetti, N. Nanoparticulate Gels for Cutaneous Administration of Caffeic Acid. Nanomaterials 2020, 10, 961. [Google Scholar] [CrossRef]
- Carbone, C.; Caddeo, C.; Grimaudo, M.A.; Manno, D.E.; Serra, A.; Musumeci, T. Ferulic Acid-NLC with Lavandula Essential Oil: A Possible Strategy for Wound-Healing? Nanomaterials 2020, 10, 898. [Google Scholar] [CrossRef] [PubMed]
- Rodríguez-Luna, A.; Talero, E.; Ávila-Román, J.; Romero, A.M.F.; Rabasco, A.M.; Motilva, V.; González-Rodríguez, M.L. Preparation and In Vivo Evaluation of Rosmarinic Acid-Loaded Transethosomes After Percutaneous Application on a Psoriasis Animal Model. AAPS Pharm.Sci. Tech. 2021, 22, 1–18. [Google Scholar] [CrossRef] [PubMed]
- Singh, A.; Srivastava, N.; Yadav, K.S.; Sinha, P.; Yadav, N.P. Preparation, optimization, characterization and bioevaluation of rosmarinic acid loaded phytovesicles for anti-inflammatory activity. J. Drug Deliv. Sci. Technol. 2020, 59, 101888. [Google Scholar] [CrossRef]
- Memic, A.; Abudula, T.; Mohammed, H.S.; Joshi Navare, K.; Colombani, T.; Bencherif, S.A. Latest progress in electrospun nanofibers for wound healing applications. ACS Appl. Bio Mater. 2019, 2, 952–969. [Google Scholar] [CrossRef]
- Suarato, G.; Contardi, M.; Perotto, G.; Jose’A, H.-G.; Fiorentini, F.; Ceseracciu, L.; Pignatelli, C.; Debellis, D.; Bertorelli, R.; Athanassiou, A. From fabric to tissue: Recovered wool keratin/polyvinylpyrrolidone biocomposite fibers as artificial scaffold platform. Mater. Sci. Eng. C 2020, 116, 111151. [Google Scholar] [CrossRef]
- Rogina, A. Electrospinning process: Versatile preparation method for biodegradable and natural polymers and biocomposite systems applied in tissue engineering and drug delivery. Appl. Surf. Sci. 2014, 296, 221–230. [Google Scholar] [CrossRef]
- Bombin, A.D.J.; Dunne, N.; McCarthy, H.O. Electrospinning of natural polymers for the production of nanofibres for wound healing applications. Mater. Sci. Eng. C 2020, 110994. [Google Scholar] [CrossRef] [PubMed]
- Kossyvaki, D.; Suarato, G.; Summa, M.; Gennari, A.; Francini, N.; Gounaki, I.; Venieri, D.; Tirelli, N.; Bertorelli, R.; Athanassiou, A. Keratin–cinnamon essential oil biocomposite fibrous patches for skin burn care. Mater. Adv. 2020, 1, 1805–1816. [Google Scholar] [CrossRef]
- Ignatova, M.G.; Manolova, N.E.; Rashkov, I.B.; Markova, N.D.; Toshkova, R.A.; Georgieva, A.K.; Nikolova, E.B. Poly (3-hydroxybutyrate)/caffeic acid electrospun fibrous materials coated with polyelectrolyte complex and their antibacterial activity and in vitro antitumor effect against HeLa cells. Mater. Sci. Eng. C 2016, 65, 379–392. [Google Scholar] [CrossRef] [PubMed]
- Narayanan, V.; Alam, M.; Ahmad, N.; Balakrishnan, S.B.; Ganesan, V.; Shanmugasundaram, E.; Rajagopal, B.; Thambusamy, S. Electrospun poly (vinyl alcohol) nanofibers incorporating caffeic acid/cyclodextrins through the supramolecular assembly for antibacterial activity. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2021, 249, 119308. [Google Scholar] [CrossRef]
- Oh, G.-W.; Ko, S.-C.; Je, J.-Y.; Kim, Y.-M.; Oh, J.; Jung, W.-K. Fabrication, characterization and determination of biological activities of poly (ε-caprolactone)/chitosan-caffeic acid composite fibrous mat for wound dressing application. Int. J. Biol. Macromol. 2016, 93, 1549–1558. [Google Scholar] [CrossRef]
- Chuysinuan, P.; Pavasant, P.; Supaphol, P. Preparation and characterization of caffeic acid-grafted electrospun poly (l-lactic acid) fiber mats for biomedical applications. ACS Appl. Mater. Interfaces 2012, 4, 3031–3040. [Google Scholar] [CrossRef] [PubMed]
- Adomavičiūtė, E.; Stanys, S.; Žilius, M.; Juškaitė, V.; Pavilonis, A.; Briedis, V. Formation and biopharmaceutical characterization of electrospun PVP mats with propolis and silver nanoparticles for fast releasing wound dressing. BioMed. Res. Int. 2016, 2016. [Google Scholar] [CrossRef] [PubMed]
- Adomavičiūtė, E.; Pupkevičiūtė, S.; Juškaitė, V.; Žilius, M.; Stanys, S.; Pavilonis, A.; Briedis, V. Formation and investigation of electrospun PLA materials with propolis extracts and silver nanoparticles for biomedical applications. J. Nanomater. 2017, 2017. [Google Scholar] [CrossRef] [Green Version]
- Vatankhah, E. Rosmarinic acid-loaded electrospun nanofibers: In vitro release kinetic study and bioactivity assessment. Eng. Life Sci. 2018, 18, 732–742. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sandova-Herrera, I.; Ledezma-Pérez, A.; De-León, A.; Alvaro-Canche, C.; Torres-Lubian, R.; Romero-García, J. Fabrication and Sustained Release of Chlorogenic Acid from Poly (vinyl alcohol)/Poly (γ-glutamic Acid) Blends Electrospun Mats. Res. Sq. 2020. [Google Scholar] [CrossRef]
- Balaji, A.; Vellayappan, M.; John, A.; Subramanian, A.; Jaganathan, S.; Supriyanto, E.; Razak, S. An insight on electrospun-nanofibers-inspired modern drug delivery system in the treatment of deadly cancers. RSC Adv. 2015, 5, 57984–58004. [Google Scholar] [CrossRef]
- Poornima, B.; Korrapati, P.S. Fabrication of chitosan-polycaprolactone composite nanofibrous scaffold for simultaneous delivery of ferulic acid and resveratrol. Carbohydr. Polym. 2017, 157, 1741–1749. [Google Scholar] [CrossRef]
- Zhao, Z.; Vizetto-Duarte, C.; Moay, Z.K.; Setyawati, M.I.; Rakshit, M.; Kathawala, M.H.; Ng, K.W. Composite hydrogels in three-dimensional in vitro models. Front. Bioeng. Biotechnol. 2020, 8, 611. [Google Scholar] [CrossRef]
- Tomatsu, I.; Peng, K.; Kros, A. Photoresponsive hydrogels for biomedical applications. Adv. Drug Deliv. Rev. 2011, 63, 1257–1266. [Google Scholar] [CrossRef]
- Debele, T.A.; Su, W.-P. Polysaccharide and protein-based functional wound dressing materials and applications. Int. J. Polym. Mater. Polym. Biomater. 2020, 1–22. [Google Scholar] [CrossRef]
- Das, S.; Wong, A.B. Stabilization of ferulic acid in topical gel formulation via nanoencapsulation and pH optimization. Sci. Rep. 2020, 10, 1–18. [Google Scholar] [CrossRef] [PubMed]
- Sivakumar, S.; Murali, R.; Arathanaikotti, D.; Gopinath, A.; Senthilkumar, C.; Kesavan, S.; Madhan, B. Ferulic acid loaded microspheres reinforced in 3D hybrid scaffold for antimicrobial wound dressing. Int. J. Biol. Macromol. 2021, 177, 463–473. [Google Scholar] [CrossRef]
- Raja, S.T.K.; Thiruselvi, T.; Aravindhan, R.; Mandal, A.B.; Gnanamani, A. In vitro and in vivo assessments of a 3-(3, 4-dihydroxyphenyl)-2-propenoic acid bioconjugated gelatin-based injectable hydrogel for biomedical applications. J. Mater. Chem. B 2015, 3, 1230–1244. [Google Scholar] [CrossRef] [PubMed]
- Rao, S.R.; Subbarayan, R.; Dinesh, M.G.; Arumugam, G.; Raja, S.T.K. Differentiation of human gingival mesenchymal stem cells into neuronal lineages in 3D bioconjugated injectable protein hydrogel construct for the management of neuronal disorder. Exp. Mol. Med. 2016, 48, e209. [Google Scholar] [CrossRef] [Green Version]
- Budhiraja, A.; Dhingra, G. Development and characterization of a novel antiacne niosomal gel of rosmarinic acid. Drug Deliv. 2015, 22, 723–730. [Google Scholar] [CrossRef] [Green Version]
- Azadmanesh, F.; Pourmadadi, M.; Zavar Reza, J.; Yazdian, F.; Omidi, M.; Haghirosadat, B.F. Synthesis of a novel nanocomposite containing chitosan as a three-dimensional printed wound dressing technique: Emphasis on gene expression. Biotechnol. Prog. 2021, e3132. [Google Scholar] [CrossRef]
- Kanitkar, A.; Smoak, M.; Chen, C.; Aita, G.; Scherr, T.; Madsen, L.; Hayes, D. Synthesis of novel polyesters for potential applications in skin tissue engineering. J. Chem. Technol. Biotechnol. 2016, 91, 733–741. [Google Scholar] [CrossRef]
- Moreira, J.; Vale, A.C.; Alves, N.M. Spin-coated freestanding films for biomedical applications. J. Mater. Chem. B 2021, 9, 3778–3799. [Google Scholar] [CrossRef]
- Kathe, K.; Kathpalia, H. Film forming systems for topical and transdermal drug delivery. Asian J. Pharm. Sci. 2017, 12, 487–497. [Google Scholar] [CrossRef]
- Contardi, M.; Russo, D.; Suarato, G.; Heredia-Guerrero, J.A.; Ceseracciu, L.; Penna, I.; Margaroli, N.; Summa, M.; Spanò, R.; Tassistro, G. Polyvinylpyrrolidone/hyaluronic acid-based bilayer constructs for sequential delivery of cutaneous antiseptic and antibiotic. Chem. Eng. J. 2019, 358, 912–923. [Google Scholar] [CrossRef]
- Fiorentini, F.; Suarato, G.; Grisoli, P.; Zych, A.; Bertorelli, R.; Athanassiou, A. Plant-based biocomposite films as potential antibacterial patches for skin wound healing. Eur. Polym. J. 2021, 150, 110414. [Google Scholar] [CrossRef]
- Liakos, I.; Rizzello, L.; Scurr, D.J.; Pompa, P.P.; Bayer, I.S.; Athanassiou, A. All-natural composite wound dressing films of essential oils encapsulated in sodium alginate with antimicrobial properties. Int. J. Pharm. 2014, 463, 137–145. [Google Scholar] [CrossRef] [PubMed]
- Neto, R.J.G.; Genevro, G.M.; de Almeida Paulo, L.; Lopes, P.S.; de Moraes, M.A.; Beppu, M.M. Characterization and in vitro evaluation of chitosan/konjac glucomannan bilayer film as a wound dressing. Carbohydr. Polym. 2019, 212, 59–66. [Google Scholar] [CrossRef] [PubMed]
- Contardi, M.; Heredia-Guerrero, J.A.; Perotto, G.; Valentini, P.; Pompa, P.P.; Spanò, R.; Goldoni, L.; Bertorelli, R.; Athanassiou, A.; Bayer, I.S. Transparent ciprofloxacin-povidone antibiotic films and nanofiber mats as potential skin and wound care dressings. Eur. J. Pharm. Sci. 2017, 104, 133–144. [Google Scholar] [CrossRef]
- Bouthillette, M.; Beccati, D.; Akthakul, A.; Ramadurai, N.; Nashat, A.; Langer, R.; Anderson, R.R.; Sakamoto, F.H. A crosslinked polymer skin barrier film for moderate to severe atopic dermatitis: A pilot study in adults. J. Am. Acad. Dermatol 2020, 82, 895–901. [Google Scholar] [CrossRef] [PubMed]
- Kang, N.-W.; Kim, M.-H.; Sohn, S.-Y.; Kim, K.-T.; Park, J.-H.; Lee, S.-Y.; Lee, J.-Y.; Kim, D.-D. Curcumin-loaded lipid-hybridized cellulose nanofiber film ameliorates imiquimod-induced psoriasis-like dermatitis in mice. Biomaterials 2018, 182, 245–258. [Google Scholar] [CrossRef] [PubMed]
- Contardi, M.; Heredia-Guerrero, J.A.; Guzman-Puyol, S.; Summa, M.; BENITEZ, J.J.; Goldoni, L.; Caputo, G.; Cusimano, G.; Picone, P.; Di Carlo, M. Combining Dietary Phenolic Antioxidants with Polyvinylpyrrolidone: Transparent Biopolymer Films based on p-Coumaric Acid for Controlled Release. J. Mater. Chem. B 2019, 7, 1384–1396. [Google Scholar] [CrossRef] [PubMed]
- Jones, J.I.; Nguyen, T.T.; Peng, Z.; Chang, M. Targeting MMP-9 in diabetic foot ulcers. Pharmaceuticals 2019, 12, 79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Contardi, M.; Alfaro-Pulido, A.; Picone, P.; Guzman-Puyol, S.; Goldoni, L.; Benítez, J.J.; Heredia, A.; Barthel, M.J.; Ceseracciu, L.; Cusimano, G. Low molecular weight ε-caprolactone-p-coumaric acid copolymers as potential biomaterials for skin regeneration applications. PLoS ONE 2019, 14, e0214956. [Google Scholar] [CrossRef]
- Žilius, M.; Ramanauskienė, K.; Briedis, V. Release of propolis phenolic acids from semisolid formulations and their penetration into the human skin in vitro. Evid. Based Complement. Altern. Med. 2013, 2013. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Seo, Y.; Kim, S.; Boo, Y.; Baek, J.; Lee, S.; Koh, J. Effects of p-coumaric acid on erythema and pigmentation of human skin exposed to ultraviolet radiation. Clin. Exp. Dermatol. Clin. Dermatol. 2011, 36, 260–266. [Google Scholar] [CrossRef]
- Küba, M.; Türkoğlu, A.; Oğuz, A.; Tuncer, M.; Kaya, Ş.; Başol, Ö.; Bilge, H.; Tatlı, F. Comparison of local rosmarinic acid and topical dexpanthenol applications on wound healing in a rat experimental wound model. Folia Morphol. 2020. [Google Scholar] [CrossRef]
- Kitagawa, S.; Yoshii, K.; Morita, S.-Y.; Teraoka, R. Efficient topical delivery of chlorogenic acid by an oil-in-water microemulsion to protect skin against UV-induced damage. Chem. Pharm. Bull. 2011, 59, 793–796. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Qi, W.; Zhao, T.; Yang, W.-W.; Wang, G.-H.; Yu, H.; Zhao, H.-X.; Yang, C.; Sun, L.-X. Comparative pharmacokinetics of chlorogenic acid after oral administration in rats. J. Pharm. Anal. 2011, 1, 270–274. [Google Scholar] [CrossRef] [Green Version]
- Bhattacharyya, S.; Majhi, S.; Saha, B.P.; Mukherjee, P.K. Chlorogenic acid–phospholipid complex improve protection against UVA induced oxidative stress. J. Photochem. Photobiol. B Biol. 2014, 130, 293–298. [Google Scholar] [CrossRef] [PubMed]
- Yutani, R.; Kikuchi, T.; Teraoka, R.; Kitagawa, S. Efficient delivery and distribution in skin of chlorogenic acid and resveratrol induced by microemulsion using sucrose laurate. Chem. Pharm. Bull. 2014, 62, 274–280. [Google Scholar] [CrossRef] [Green Version]
- Chen, W.-C.; Liou, S.-S.; Tzeng, T.-F.; Lee, S.-L.; Liu, I.-M. Effect of topical application of chlorogenic acid on excision wound healing in rats. Planta Med. 2013, 79, 616–621. [Google Scholar] [CrossRef] [PubMed]
- Waghule, T.; Singhvi, G.; Dubey, S.K.; Pandey, M.M.; Gupta, G.; Singh, M.; Dua, K. Microneedles: A smart approach and increasing potential for transdermal drug delivery system. Biomed. Pharmacother. 2019, 109, 1249–1258. [Google Scholar] [CrossRef] [PubMed]
- Zhu, J.; Zhang, Q.; Yang, T.; Liu, Y.; Liu, R. 3D printing of multi-scalable structures via high penetration near-infrared photopolymerization. Nat. Commun. 2020, 11, 3462. [Google Scholar] [CrossRef] [PubMed]
- Xie, C.-y.; Gu, Z.-x.; You, X.; Liu, G.; Tan, Y.; Zhang, H. Screening of edible mushrooms for release of ferulic acid from wheat bran by fermentation. Enzym. Microb. Technol. 2010, 46, 125–128. [Google Scholar] [CrossRef]
- Antinori, M.E.; Contardi, M.; Suarato, G.; Armirotti, A.; Bertorelli, R.; Mancini, G.; Debellis, D.; Athanassiou, A. Advanced mycelium materials as potential self-growing biomedical scaffolds. Sci. Rep. 2021, 11, 12630. [Google Scholar] [CrossRef] [PubMed]
- Narayanan, K.B.; Zo, S.M.; Han, S.S. Novel biomimetic chitin-glucan polysaccharide nano/microfibrous fungal-scaffolds for tissue engineering applications. Int. J. Biol. Macromol. 2020, 149, 724–731. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Contardi, M.; Lenzuni, M.; Fiorentini, F.; Summa, M.; Bertorelli, R.; Suarato, G.; Athanassiou, A. Hydroxycinnamic Acids and Derivatives Formulations for Skin Damages and Disorders: A Review. Pharmaceutics 2021, 13, 999. https://doi.org/10.3390/pharmaceutics13070999
Contardi M, Lenzuni M, Fiorentini F, Summa M, Bertorelli R, Suarato G, Athanassiou A. Hydroxycinnamic Acids and Derivatives Formulations for Skin Damages and Disorders: A Review. Pharmaceutics. 2021; 13(7):999. https://doi.org/10.3390/pharmaceutics13070999
Chicago/Turabian StyleContardi, Marco, Martina Lenzuni, Fabrizio Fiorentini, Maria Summa, Rosalia Bertorelli, Giulia Suarato, and Athanassia Athanassiou. 2021. "Hydroxycinnamic Acids and Derivatives Formulations for Skin Damages and Disorders: A Review" Pharmaceutics 13, no. 7: 999. https://doi.org/10.3390/pharmaceutics13070999
APA StyleContardi, M., Lenzuni, M., Fiorentini, F., Summa, M., Bertorelli, R., Suarato, G., & Athanassiou, A. (2021). Hydroxycinnamic Acids and Derivatives Formulations for Skin Damages and Disorders: A Review. Pharmaceutics, 13(7), 999. https://doi.org/10.3390/pharmaceutics13070999