Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 7032 word(s) 7032 2021-07-09 05:10:41 |
2 format change -1 word(s) 7031 2021-07-23 03:09:31 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Trigo-Gutierrez, J.K.; Mima, E.G.D.O.; Vega Chacón, Y. Antimicrobial Activity of Curcumin. Encyclopedia. Available online: https://encyclopedia.pub/entry/12312 (accessed on 25 April 2024).
Trigo-Gutierrez JK, Mima EGDO, Vega Chacón Y. Antimicrobial Activity of Curcumin. Encyclopedia. Available at: https://encyclopedia.pub/entry/12312. Accessed April 25, 2024.
Trigo-Gutierrez, Jeffersson Krishan, Ewerton Garcia De Oliveira Mima, Yuliana Vega Chacón. "Antimicrobial Activity of Curcumin" Encyclopedia, https://encyclopedia.pub/entry/12312 (accessed April 25, 2024).
Trigo-Gutierrez, J.K., Mima, E.G.D.O., & Vega Chacón, Y. (2021, July 22). Antimicrobial Activity of Curcumin. In Encyclopedia. https://encyclopedia.pub/entry/12312
Trigo-Gutierrez, Jeffersson Krishan, et al. "Antimicrobial Activity of Curcumin." Encyclopedia. Web. 22 July, 2021.
Antimicrobial Activity of Curcumin
Edit

Curcumin (CUR) is a natural substance extracted from turmeric that has antimicrobial properties. Due to its ability to absorb light in the blue spectrum, CUR is also used as a photosensitizer (PS) in antimicrobial Photodynamic Therapy (aPDT). 

curcumin drug delivery systems antimicrobial agents microbial drug resistance viruses bacteria fungi photochemotherapy

1. Introduction

The global changes arising from globalization and climate change have a profound impact on human health, including infectious diseases [1][2]. The increased mobility of people, urbanization, greenhouse-gas emissions, pollution, deforestation, global warming, loss of sea ice, sea-level rise, extreme weather events with droughts and flood, etc., have all contributed to affect the transmission, prevalence, and spread of existing infections, such as vector-borne diseases, and the emergence of new pathogens [1][2] . In some cases, these infections have resulted in epidemics such as dengue and pandemics such as COVID-19, which the world is currently facing [3].
Notwithstanding the existence of anti-infective medications, other current concerns are the drug resistance arising from the misuse of antimicrobial agents and the emergence of multidrug-resistant species [4]. These problems are a challenge for humanity, especially when considering that the development of new drugs demands time and money. Thus, the repurposing of existing medications and alternative therapies, such as natural substances, has been investigated [5][6][7].
Curcumin (CUR) is a yellow dye (diferuloylmethane—a natural polyphenol) found in turmeric (Curcuma longa), which is a plant native to India and Southeast Asia. Beyond its culinary use as food flavoring and coloring, CUR also has a potential application in medicine due to its therapeutic properties, which include antioxidant, anticancer, anti-inflammatory, and antimicrobial effects [8]. CUR is not toxic and, according to the Food and Drug Administration, it is “Generally Recognized as Safe” [9]. The literature shows a plethora of studies reporting the biological and pharmacological features of CUR on health. Comprehensive reviews are available on the anticancer [10], anti-inflammatory [8], and antimicrobial [11] effects of CUR.
Nonetheless, CUR is not soluble in water, unstable in solutions, and shows low bioavailability, poor absorption, and rapid elimination from the body [11]. For these reasons, organic solvents such as ethanol, methanol, acetone, and dimethyl sulfoxide (DMSO) have been used to solubilize CUR [12]. These drawbacks hinder the in vivo use of CUR as a therapeutic agent. Thus, some approaches have been used to overcome the problems of CUR, such as the use of adjuvants and drug delivery systems. Piperine, a substance derived from black pepper, and lecithin, a phospholipid, have been associated with CUR to improve its bioavailability by blocking the metabolism of CUR and enhancing its gastrointestinal absorption [11]. Additionally, drug delivery systems have been used to solubilize CUR and protect it from degradation until it reaches the target tissue, where CUR is sustainably released [13].
Nanotechnology has been a promising field in medicine (nanomedicine). Nanoscale structures show intrinsic physical and chemical properties, which have been exploited as diagnostic and therapeutic tools [13][14]. The present study reviews the drug delivery systems (DDS) used for CUR, aiming at its antimicrobial effect. Although comprehensive reviews about the antimicrobial effect of CUR (encapsulated or not) are found elsewhere [15][16][17][18], they describe only the antibacterial and antifungal activities of CUR in DDSs . Our review summarizes the DDSs used for CUR as an antiviral, antibacterial, and antifungal agent, encompassing different nanosystems (colloids and metals) and the relevant issues of antimicrobial resistance and the emergence of new pathogens.

2. Free CUR

The broad-spectrum activity of CUR as an antibacterial, antifungal, and antivirus agent was reviewed previously [15][16]. Thus, this section reviews recent studies not covered by these reviews about the antimicrobial activity of free (non-encapsulated) CUR (Table 1) before reporting the DDS used for CUR.
Table 1. In vitro and in vivo studies using free CUR and curcuminoids as antimicrobial.
Solvent Microorganism Culture Antimicrobial Method CUR
Concentration
Light/Ultrasonic Parameters Reference
DMSO (0.4%) ZIKV Cell infection IC50 5.62–16.57 µM - [19]
>DGEV >IC90
N/R HPVA Cell infection Viral survival 0.015 mg/mL - [20]
Tulane V
N/R KSPV Infected cells EC50 Up to 6.68 µM - [21]
Aqueous Piper nigrum seed extract SARS-CoV-2 Cell infection IC50 Plaque reduction 0.4 µg/mL - [22]
DMSO (<0.4%) SARS-CoV-1 Cell infection Inhibiton of viral replication 20 µM - [23]
N/R SARS-CoV In vitro Viral inhibition 23.5 µM - [24]
N/R SARS-CoV In vitro papain-like inhibition 5.7 µM - [25]
DMSO (1 w/v) S. aureus Planktonic Inhibition zone MIC 600 and - [26]
E. coli 400 µg/mL
DMSO MRSA Planktonic MIC
FICI
15.5 µg/mL - [27]
N/R S. aureus Planktonic Colony count 100 µg/mL 8 or 20 J/cm2 [28]
MSSA
MRSA
DMSO (10%) S. aureus Biofilm aPDT 20, 40, and 80 µM 5.28 J/cm2 [29]
DMSO VRSA Biofilm/animal infection model MIC
MBC
156.25 µg/mL 20 J/cm2 [30]
N/R S. aureus Animal infection model aPDT 78 µg/mL 60 J/cm2 [31]
DMSO S. aureus Infected fruit Survival fraction 100 nM 1.5 and 9 J/cm2 [32]
N/R S. aureus Planktonic PDI 40 and 80 µM 15 J/cm2 [33]
E. coli
Tween 80 (0.5%) S. aureus Planktonic CFU/mL 300 and 500 µM 0.03–0.05 W/cm2 [34]
N/R S. aureus Biofilm Confocal microscope N/R 170 µmol m2 s1 [35]
DMSO (0.5%) S. aureus Biofilm SDT
aPDT
SPDT
80 µM 100 Hz
15 and 70 J/cm2
100 Hz, 15 and 70 J/cm2
[36]
DMSO E. coli Planktonic MIC
Inhibition zone
110, 220 and 330 µg/mL - [37]
DMSO E. coli Planktonic OD600nm 8,16, 32, and 64 µg/mL - [38]
N/R S. dysenteriae Planktonic MIC/MBC 256 and - [39]
C. jejuni 512 µg/mL
Edible alcohol E. coli Planktonic aPDT 5, 10, and 20 µM 3.6 J/cm2 [40]
DMSO H. pylori Planktonic biofilm MIC
MBC
aPDT
50 µg/mL 10 mW/cm2 [41]
DMSO P. aeruginosa Biofilm aPDT
CFU/mL
N/R 5 and 10 J/cm2 [42]
DMSO Imipenem-resistant
A. baumannii
Planktonic aPDT 25, 50, 100, and 200 µM 5.4 J/cm2 [43]
DMSO (2%) P. aeruginosa, A. baumannii, K. pneumoniae, E. coli, E. faecalis Planktonic MIC/FICI 128-256 µg/mL - [44]
N/R C. difficile, C. sticklandii, B. fragilis, P. bryantii Planktonic Viable cell number 10 µg/mL - [45]
N/R B. subtillis, E. coli, S. carnosus, M. smegmatis Planktonic MIC/MBC Up to 25 µM - [46]
N/R MRSA Planktonic/animal infection model MIC 4–16 μg/mL - [47]
MSSA 2–8 μg/mL
E. coli 8–32 μg/mL
N/R E. faecalis, S. aureus, B. subtillis, P. aeruginosa, E. coli Planktonic MIC 156 μg/mL - [48]
DMSO (0.5%) A. hydrophila, E. coli
E. faecalis, K. pneumoniae, P. aeruginosa, S. aureus, C. albicans
Planktonic MIC/MBC/
FICI/aPDT
37.5–150 µg/mL N/C [49]
N/R E. faecalis Infection model CFU/mL 1 µg/mL - [50]
Commercial solution E. faecalis Biofilm aPDT 1.5 g/mL 20.1 J/cm2 [51]
Ethanol 99% A. hydrophila, V. parahaemolyticus Planktonic aPDT/SDT Up to 15 mg/L N/C [52]
DMSO (10%) E. faecalis Biofilm MIC/MBC 120 mg/mL - [53]
N/R S. mutans Planktonic aPDT 10 g/100cc N/C [54]
DMSO: ethyl alcohol S. mutans, S. pyogenes Planktonic aPDT 3 mg/mL 28.8 J/cm2 [55]
DMSO (0.8%) Caries isolated Biofilm aPDT 600 µg/mL 75 J/cm2 [56]
DMSO S. mutans, C. albicans Biofilm single/dual MBEC 0.5 mM - [57]
DMSO (0.05 M) A. actinomycetemcomitans Planktonic aPDT 40 µg/mL 300–420 J/cm2 [58]
DMSO (<1%) P. gingivalis, A. actinomycetemcomitans Planktonic aPDT 20 µg/mL 6, 12 or 18 J/cm2 [59]
DMSO (0.5%) P. gingivalis, A. actinomycetemcomitans, C. rectus, E. corrodens, F. nucleatum, P. intermedia, P. micra, T. denticola, T. forsythis Biofilm aPDT 100 mg/L - [60]
N/R Subgingival plaque Biofilm aPDT 100 µg/mL 30 J/cm2 [61]
DMSO P. gingivalis Planktonic MIC 12.5 µg/mL - [62]
Ethanol: DMSO
(99.9%: 0.1%)
Periodontal pocket - aPDT 100 mg/mL 7.69 J/cm2 [63]
Tween 80 Streptococcus spp, Staphylococcus spp, Enterobacteriaceae, C. albicans Clinical trail aPDT 0.75 mg/mL 20.1 J/cm2 [64]
Sodium hydroxide: PBS C. albicans, C. parapsilosis, C. glabrata, C.dubliniensis Planktonic/biofilm MIC 0.1–0.5 mg/mL - [65]
N/R C. albicans, S. aureus Planktonic
Biofilm
MIC/Biofilm percentag 200 µg/mL - [66]
N/R C. albicans Biofilm aPDT 1.5 g/mL 20.1 J/cm2 [67]
DMSO (10%) C. albicans Biofilm aPDT 20, 40, 60 and 80 µM 2.64, 5.28, 7.92, 10.56, and 13.2 J/cm2 [68]
DMSO (1%) C. albicans Biofilm aPDT 40 and 80 mM 37.5 and 50 J/cm2 [69]
N/R C. albicans Biofilm aPDT 100 µM 10 J/cm2 [70]
DMSO (2.5%) Fluconazole-resistant C. albicans Planktonic/biofilm/infection model MIC/
aPDT
40 µM 5.28 J/cm2 [71]
Fluconazole-susceptible C. albicans 80 µM 40.3 J/cm2
DMSO C. albicans, F. oxysporum, A. flavus, A. niger, C. neoformans Planktonic MIC 137.5–200 μg/mL - [72]

2.1. Antiviral Activity

The antiviral activity of CUR has been described against enveloped and non-enveloped DNA and RNA viruses, such as HIV, Zika, chikungunya, dengue, influenza, hepatitis, respiratory syncytial viruses, herpesviruses, papillomavirus, arboviruses, and, noroviruses [11][73][74]. The action mechanism of CUR involves the inhibition of viral attachmentand penetration into the host cell and interference with viral replication machinery and the host cell signaling pathways used for viral replication. Moreover, CUR works as a virucidal substance, acting on the viral envelope or proteins [11][73][74]. CUR in 0.4%vol/vol DMSO was able to inhibit several strains of the Zika virus, including those causing human epidemics, inhibiting the viral attachment to the host cell [19]. The inhibitory effect was potentiated when CUR was combined with gossypol, which is another natural product. CUR also inhibited human strains of the dengue virus [19]. The combination of CUR with heat treatment reduced the time and temperature needed for inactivating the foodborne enteric virus (hepatitis A virus and Tulane virus—a cultivable surrogate of the human norovirus) [20]. CUR was able to inhibit the lytic replication of Kaposi’s sarcoma-associated herpesvirus (KSHV) as well as reduce its pathogenesis (neoangiogenesis and cell invasionof KSHV-infected mesenchymal stem cell from the periodontal ligament) [21].
While the antiviral effect of CUR has been experimentally demonstrated, the effect of CUR against the new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the etiologic agent of COVID-19, has been predicted by in silico studies using computational techniques, such as molecular docking [75][76][77][78][79][80][81][82]. These in silico studies showed the binding affinity of CUR to the spike protein of SARS-CoV-2 and the human receptors of the host cell, which could inhibit the viral infection into the human cells. The targets to which CUR may bind are the viral non-structural protein 9 (Nsp9) [77] and 15 (Nsp15) [81], main proteases of SARS-CoV-2 (important for viral replication) [75][80], receptor-binding domains (RBD) of the viral spike protein [76,78,82], human cell receptors angiotensin-converting enzyme2 (ACE2) [76][82], and glucose-regulating protein 78 (GRP78) [79], as well as the RBD/ACE complex [76]. Nevertheless, a virtual screening evaluated the interaction between potential functional foods and the main protease of SARS-CoV-2 and found that CUR showed lower docking affinity than flavonoids, vitamin, and β-sitosterol [83]. Omics approaches have been studied to identify infection pathways and propose drugs that could target these pathways. Thus, an integrative multiomics (interactome, proteome, transcriptome, andbibliome) analysis identified biological processes and SARS-CoV-2 infection pathways and proposed CUR as a potential prophylactic agent for blocking the SARS-CoV-2 infection [84].Although most investigations have evaluated the potential of CUR against SARS-CoV-2 by computational simulations, an in vitro study showed that an immunomodulatory herbal extract composed of CUR and piperine presented a virucidal effect (viral inhibitionof up to 92%) on SARS-CoV-2 [22]. Other in vitro studies showed the ability of CUR to inhibit its viral predecessor—the SARS-CoV-1 [23][24]. CUR in DMSO (<0.4%) inhibited by 25–50% the cytopathogenic effect of SARS-CoV-1 on Vero E6 cells and by 50% the viral replication and 3CL protease (main protease) [23]. Another study used CUR as a positive control for 3CL protease inhibition [24]. CUR also inhibited the papain-like protease, which is another protease used for SARS-CoV replication [25].

2.2. Antibacterial Activity

The antibacterial effect of CUR has been demonstrated against Gram-positive and Gram-negative species, including strains responsible for human infections and showing antibiotic resistance [11][16][85][86]. CUR also inhibits bacterial biofilms, which are communities of cells embedded in a self-produced polymeric matrix tolerant to antimicrobial treatments [11][16][85][86]. The antibacterial mechanism of action of CUR involves damage to the cell wall or cell membrane, interference on cellular processes by targeting DNA and proteins, and inhibition of bacterial quorum sensing (communication process mediated by biochemical signals that regulate cell density and microbial behavior) [85]. Moreover, CUR affected the L-tryptophan metabolism in Staphylococcus aureus (Gram-positive) but not in Escherichia coli (Gram-negative), produced lipid peroxidation, and increased DNA fragmentation in both bacteria [26]. These results, along with the increased levels of total thiol and antioxidant capacity observed after bacterial cells were treated with CUR, suggested that oxidative stress may be the mechanism of antibacterial action of CUR [26].Therefore, these multiple targets make CUR an interesting option for antibiotic-resistant strains. CUR is effective in killing methicillin-resistant S. aureus (MRSA), which is a concerning pathogen responsible for nosocomial and community-associated infections [86]. CUR and another polyphenol, quercetin, inhibited the growth of MRSA and their combinationwas synergistic [27]. Moreover, CUR absorbs blue light (400–500 nm) and is used as anatural photosensitizer (PS) in antimicrobial Photodynamic Therapy (aPDT) [87]. CUR-mediated aPDT reduced the viability of reference strain of S. aureus and clinical isolates of methicillin-sensitive S. aureus (MSSA) and MRSA by 4 log10, while CUR alone reduced their survival by 2 log10 [28]. The aPDT mediated by CUR in 10% DMSO reduced the biofilm viability of S. aureus and MRSA by 3 and 2 log10, respectively, and their metabolicactivity by 94% and 89%, respectively [29]. The antibiofilm activity of CUR-mediated aPDT was also observed against clinical isolates of vancomycin-resistant S. aureus (VRSA),with reductions of 3.05 log10 in biofilm viability, 67.73% in biofilm biomass, and 47.94% in biofilm matrix [30]. Additionally, aPDT resulted in the eradication of VRSA in a rat model of skin infection [30]. The association of CUR-mediated aPDT with artificial skin resulted in a 4.14 log10 reduction in S. aureus from infected wounds in rats [31].
The combination of CUR and another natural PS, hypocrellin B, increased the photoinactivation of S. aureus compared with the photodynamic effect of each PS alone [32]. Bacterial cells showed alteration in their membrane integrity and the dual-PS-mediatedaPDT also decontaminated apples with S. aureus [32]. The CUR-mediated aPDT was also effective in decontaminating food, reducing the number of S. aureus recovered from meatand fruit [33]. Compared to another natural PS, aloe-emodin, CUR was less effective inphotokilling S. aureus and E. coli [34]. Three-dimensional _cages fabricated with CURand resin monomer (pentaerythritol triacrylate) polymerized by infrared light were usedto entrap and kill S. aureus. Irradiation of µcages for 10 min with visible light resulted in a bacteria mortality rate of 95% [35]. Following the principles of aPDT, SonodynamicTherapy (SDT) associates a PS (also called sonosensitizer) with ultrasound (US) instead of light for the treatment of deeper lesions and infections, where light cannot reach [88].Both aPDT and SDT mediated by CUR, as well as the combination of both (SPDT, when the PS is activated by light and US simultaneously), reduced the viability of S. aureus biofilmS. SPDT promoted the highest reduction (3.48 log10), which was potentiated when CUR was combined with sodium dodecyl sulfate (7.43 log10) [36]. Regarding Gram-negative species, CUR alone was not able to inhibit the growth of an Enterotoxigenic E. coli, which is a strain that causes severe diarrhea and is resistant to antibiotics [37]. However, synergism was observed between CUR at 330 µg/mL and antibiotics (Ceftazidime, Amoxicillin/Clavulanic acid, Cefotaxime, and Ampicillin) [37]. CUR did not affect the growth of enteroaggregative (EAEC) and enteropathogenic (EPEC) diarrheagenic E. coli but inhibited the secretion and release of their virulence factors, Pet,and EspC, which are toxins produced by these strains [38]. Conversely, CUR alone and with ampicillin inhibited the growth of other species that caused diarrhea—Shigella dysenteriae and Campylobacter jejuni, including multidrug-resistant strains [39]. The aPDT mediated byCUR and light reduced the viability of E. coli by 3.5 log, increased membrane permeabilityof bacteria, and decontaminated oysters [40]. CUR-mediated aPDT reduced the viability of Helicobacter pylori and its virulence factors (motility, urease production, adhesion toerythrocytes, and biofilm formation) [41]. On Pseudomonas aeruginosa, aPDT potentiated the inhibitory effect of CUR, inhibited biofilm formation and matrix production, reduced biofilm thickness, and downregulated quorum sensing genes [42]. The photoinactivation of imipenem-resistant Acinetobacter baumannii reduced bacterial viability by 97.5% and shotgun proteomics analysis identified 70 carbonylated proteins modified after CUR mediated aPDT related to the membrane, translation, and response to oxidative stress [43].CUR inhibited the growth of antibiotic-resistant P. aeruginosa, A. baumannii, and Klebsiella pneumoniae isolated from burn wound infections and showed synergism with meropenem [44]. On gastrointestinal bacteria of human and bovine origin, CUR inhibited Firmicutes (Clostridioides difficile and Acetoanaerobium (Clostridium) sticklandii) but did not affect Bacteroidetes (Bacteroides fragilis and Prevotella bryantii) [45]. CUR was conjugated to triphenyl phosphonium resulting in a compound named Mitocurcumin, which inhibited the growth of Bacillus subtilis, E. coli, Staphylococcus carnosus, and Mycobacterium smegmatis, and induced morphological changes in B. subtilis [46]. Seventeen synthesized monocarbonyl curcuminoids showed high antibacterial activity against MSSA and MRSA and moderate activity against E. coli [47]. The four most effective curcuminoids were bacteriostatic at low concentrations and bactericidal at high concentrations against MRSA, which showed membrane damage. In an ex vivo mammalian co-culture infection model,two curcuminoids decreased the viability of MSSA internalized in the fibroblasts [47]. One of thirteen synthesized curcuminoids, 3,30-dihydroxycurcumin, showed antibacterial activity against S. aureus, B. subtilis, Enterococcus faecalis, and Mycobacterium tuberculosis, and produced membrane damage on B. subtilis [48]. Nonetheless, all the synthesized curcuminoids were not effective against Gram-negative species (P. aeruginosa and E. coli) [48]. CUR analogs (monocurcuminoids, MC) were synthesized and showed higher, lower, or similar antimicrobial activity than CUR against Aeromonas hydrophila, E. coli, E. faecalis, K. pneumoniae, P. aeruginosa, S. aureus, and the yeast Candida albicans [49]. Two MC and turmeric powder presented synergism against A. hydrophila, P. aeruginosa, and C. albicans. When aPDT was performed with UV light, two MC-mediated aPDT decreased the growth of E. faecalis, E. coli, and S. aureus, while aPDT with another MC and CUR increased the growth of A. hydrophila, E. faecalis, S. aureus, C. albicans, and P. aeruginosa [49]. CUR was more effective than other natural biomolecules (quercetin and resveratrol) in inhibiting thegrowth of E. faecalis in spermatozoa from rabbits, but less effective than antibiotics [50]. CUR-mediated aPDT also reduced the viability of E. faecalis biofilms grown in bovine bone cavities for 14 days by 1.92 log10 [51]. The aPDT and the combination of a nanobubble solution and the US reduced the viability of the aquatic pathogens Aeromonas hydrophila and Vibrio parahaemolyticus [52].
CUR and aPDT have been used for dental infections and oral diseases. The Curcuma longa extract decreased the viability of 3-week-old E. faecalis biofilms formed on the root canal surface of human teeth [53]. The aPDT mediated by CUR and continuous laser irradiationeradicated planktonic cultures of Streptococcus mutans, which is the main etiologic factor of dental caries [54]. A formulation of syrup with curcuminoids and 30% sucrose wasused as a PS in aPDT, which reduced the viability of S. mutans, Streptococcus pyogenes, and aclinical isolate from a patient with pharyngotonsillitis [55]. Microbial samples from carious dentin were grown as microcosm biofilm and submitted to CUR-mediated aPDT, which reduced the vitality of 3-  and 5-day-old biofilms [56]. CUR alone decreased the biomass and the viability of mono- and dual-species biofilms of S. mutans and C. albicans, as well as the production of biofilm matrix and the expression of genes related to glucosyltransferaseand quorum sensing of S. mutans, and the adherence of C. albicans [57].The therapeutic effect of CUR on periodontal diseases was extensively investigated in animal models and clinical trials, which were reviewed [89]. Beyond its antibacterial activity, CUR-mediated aPDT also produced a bystander effect (behavior change of cellsexposed to treated target cells) on the periodontal pathogen Aggregatibacter actinomycetemcomitans, reducing its survival, metabolic activity, and the production of quorum sensing molecule [58]. The aPDT with CUR decreased the growth of both A. actinomycetemcomitans and Porphyromonas gingivalis, which is another pathogenic periodontal bacterium [59].Antimicrobial photothermal treatment promoted higher reduction than CUR-mediated aPDT in the viability of mixed biofilms of periodontal pathogens (P. gingivalis, A. actinomycetemcomitans, Campylobacter rectus, Eikenella corrodens, Fusobacterium nucleatum, Prevotella intermedia, Parvimonas micra, Treponema denticola, and Tannerella forsythia) grown on a titanium surface inside artificial periodontal pockets [60]. The aPDT mediated by different PS (methylene blue, CUR, and chlorin e6) eradicated the planktonic growth and reducedthe biofilm viability of metronidazole-resistant bacteria from the subgingival plaque [61].CUR alone inhibited the growth of P. gingivalis and CUR in gel was biocompatible when evaluated subcutaneously in rats [62].
A randomized clinical trial showed that CUR-mediated aPDT associated with scaling and root planing improved the clinical attachment level gain of periodontal pockets in type-2 diabetic patients after three and six months [63]. The aPDT with CUR and LED applied in the mouth of 30 patients with acquired immune deficiency syndrome (AIDS) reduced the counts of Streptococcus spp., Staphylococcus spp., and total microorganismsfrom saliva, but not the number of Enterobacteriaceae and Candida spp. [64]. Additionally,there was no reduction in patients with CD8 lymphocytes lower than 25% [64].

2.3. Antifungal Activity

The antifungal activity of CUR has been demonstrated mostly against Candida spp. by many in vitro and few in vivo studies [90]. CUR inhibited the growth of a reference strainand a clinical isolate of C. albicans, as well as reference strains of Candida parapsilosis, Candida glabrata, and Candida dubliniensis [65]. When biofilms of both C. albicans strains were evaluated, CUR reduced only the viability of the standard strain in a concentration-dependent effect, while the antifungal fluconazole did not inhibit the viability of either strain [65]. CUR and 2-aminobenzimidazole (2-ABI) inhibited the growth and adhesion of C. albicans and S. aureus to medical-grade silicone [66]. The combination of CUR and 2-ABI enhanced the inhibition of biofilm formation and reduced the viability of 48 h-old single and dual-species biofilms [66]. The aPDT mediated by CUR reduced the survival of 14-day-old biofilm of C. albicans in bone cavities, confirmed by fluorescence spectroscopy [67]. CUR-mediated aPDT reduced the metabolic activity of biofilms of C. albicans reference strain and clinical isolates from the oral cavity of patients with HIV and lichen planus [68]. Moreover, genes related to hyphae and biofilm formation were downregulated [68]. The aPDT mediatedby CUR and another PS, Photodithazine®, also resulted in the downregulation of genes involved in adhesion and oxidative stress response in C. albicans biofilms [69]. CUR alone and CUR-mediated aPDT, combined or not with an antibody-derived killer decapeptide,reduced the metabolic activity of an 18 h biofilm of C. albicans [70]. CUR showed synergism with fluconazole and CUR-mediated aPDT inhibited the planktonic growth and reduced the biofilm viability of fluconazole-resistant C. albicans [71]. CUR-mediated aPDT also increased the survival of Galleria mellonella infected with fluconazole-susceptible C. albicans, but did not affect the survival of larvae infected with fluconazole-resistant strain [71]. A library of 2-chloroquinoline incorporated monocarbonyl curcuminoids (MACs) was synthetized and most of the MACs exhibited strong or moderate antifungal activity compared with miconazole against C. albicans, Fusarium oxysporum, Aspergillus flavus, Aspergillus niger, and Cryptococcus neoformans [72]. To suggest a possible antifungal mechanism, a moleculardocking analysis showed that MACs had binding affinity to sterol 14α-demethylase(CYP51), leading to impaired fungal growth [72].

3. Curcumin in DDSs (Colloidal, Metal, and Hybrid Nanosystems)

3.1. CUR in Micelles

Micelles are aggregates of surfactants or block polymers self-assembled in water solution. They are used as DDSs and formed by a hydrophilic domain named corona and a hydrophobic domain called core (Figure 1) [91], which stays in contact with hydrophobic drugs such as CUR [91]. Micelles have low toxicity, biocompatibility, and sustained release, which makes them an attractive DDS to carry CUR and to be used in medical applications [91]. Antimicrobial studies with CUR-loaded micelles are summarized in Table 2.
Figure 1. Schematic representation of: (A) an amphiphilic molecule and (B) an assembled micelle.
Table 2. Antimicrobial studies performed with CUR in micelles.
Type of Micelles [CUR] Formulation Microorganism Type of
Culture
Antimicrobial Method Antimicrobial [CUR] Light/Ultrasonic Parameters Reference
Mixed polymer micelles 1000 ppm E. coli, S. aureus, A. niger Planktonic MIC 350 and 275 µg/mL - [92]
PCL-b-PAsp and Ag 2 mg/mL P. aeruginosa, S. aureus Planktonic OD600nm 8–500 µg/mL - [93]
mPEG-OA 1:10 P. aeruginosa Planktonic MIC 400 µg/mL - [94]
PEG-PCL 10 mg C. albicans Planktonic MIC 256 µg/mL - [95]
PEG-PE 50 mM S. mutans Planktonic SACT 50 mM 1.56 W/cm2 [96]
DAPMA, SPD, SPM 0.32 mg/mL P. aeruginosa Planktonic OD600nm and aPDT 250, 500 nM, 1 µM and 50, 100 nM 18 and 30 J/cm2 [97]
P123 0.5% w/V S. aureus Planktonic aPDT 7.80 μmol/L 6.5 J/cm2 [98]
PCL-b-PHMG-b-PCL, STES 10 mg S. aureus,E. coli Planktonic MIC 16 and 32 μg/mL * - [99]
CUR-PLGA-DEX 1 mg/mL P. fluorescens, P. putida Planktonic biofilm OD600nm antibiofilm 0.625–5 mg/mL - [100]

3.2. CUR in Liposomes

Liposomes are biodegradable and biocompatible systems, which consist of hydrophobic and hydrophilic groups (Figure 2) [101]. The hydrophobic layer is mainly composed of phospholipids and cholesterol molecules. This lipid-based carrier is suitable for administering water-insoluble drugs, such as CUR [102]. Liposomes are classified into three groups: single unilamellar vesicles, large unilamellar vesicles, and multilamellar vesicles [103]. Drugs encapsulated in liposomes are protected from chemical degradation and show increased drug solubility [101]. Additionally, liposomes have advantageous properties such as better penetration into the skin, deposition, anti-melanoma, and antimicrobial activity [102]. Antimicrobial studies with CUR-loaded liposomes are summarized in Table 3.

Figure 2. Schematic representation of the liposome structure.

Table 3. Antimicrobial studies performed with CUR in liposomes and solid lipid nanoparticles (SLN).

Type of Liposomes or SLN [CUR] Formulation Microorganism Type of Culture Antimicrobial Method Antimicrobial [CUR] Reference
Lecithin and cholesterol 0.5 mg/mL A. sobria, C. violaceum, A. tumefaciens Planktonic biofilm MIC, antibiofilm 420, 400, and 460 μg/mL       [104]
PCNL 60.65 ± 1.68 µg/µl B. subtilis, K. pneumoniae, C. violaceum, E. coli, M. smegmatis, A. niger, C. albicans, F. oxysporum Planktonic Disk diffusion assay N/R [105]
Phosphocolines 100:1 M S. aureus Planktonic MIC 7 μg/mL [106]
PLGA: triglycerides: F68 0.8 mg/mL E. coli, S. typhimurium, P. aeruginosa, S. aureus, B. sonorensis, B. licheniformis Planktonic MIC 75 and 100 μg/mL [107]
Soya lecithin and menthol 0.5 mg/mL MRSA Planktonic, Biolfim MIC, microscopy, biomass 10 and 125 µg/mL [108]
CurSLN 60 mg/500 mg lipid S. aureus, S. mutans, V. streptococci, L. acidophilus, E. coli, C. albicans Planktonic MIC, MBC 0.09375–3 and 1.5–6 mg/mL [109]

[CUR]: CUR concentration. N/R: not reported.

3.3. CUR in Solid Lipid Nanoparticles

Solid lipid nanoparticles (SLN, Figure 3, Table 3) are a modern type of lipid-based carrier composed by solid biodegradable lipids and spherical solid lipid particles. SLNs are water colloidal or aqueous surfactant solution systems [102]. SLNs have advantages such as biocompatibility, biodegradability, greater drug absorption, and drug retention [18][102], thus they are an interesting system to carry CUR [14]. Currently, SLNs have become popular because they are used as carriers for COVID-19 vaccines based on RNA vaccine technology (Moderna and Pfizer–BioNTech).

Figure 3. Schematic representation of solid lipid nanoparticle.

3.4. CUR in Nanoemulsions

Nanoemulsions (NE) are thermodynamically stable dispersions of oil and water (Figure 4) [110]. They are formed by a phospholipid monolayer composed of a surfactant and co-surfactant, which are important for nanoemulsion stabilization [110][111]. This system has thermodynamic stability and high solubilization characteristics, with improved drug release kinetics [112]. NE systems can be manufactured through emulsification, which can control the size of the drops and increase the drug solubility and efficacy. Moreover, the main disadvantage of NE is the high amount of surfactants in the formulation, which can lead to a potential toxic effect [111]. Antimicrobials studies with CUR-loaded NE are summarized in Table 4.

Figure 4. Schematic diagram of oil-in-water nanoemulsion (A) and water-in-oil nanoemulsion (B), stabilized by surfactants.

Table 4. Antimicrobial studies performed with curcumin/curcuminoid in emulsions.

Type of

Emulsion

[CUR] Formulation Microorganisms  Type of culture Antimicrobial method  Antimicrobial Concentration  Light/Ultrasonic Paramet Reference 
THC ME 5% HIV-1 Cell infection  IC50 0.9357 μM - [113]
CUR-NE N/R HPV - aPDT 80 µM 50 J/cm2 [114]
CUR-NE N/R DENV-1 to 4 Cell infection  Cell viability 1, 5, 10 µg/mL - [115]
P60-CUR 4 mg/L E. coli Planktonic  OD595 nm N/R - [116]
PE:CUR 0.566 mg/mL S. aureus, S. epidermidis, S. faecalis, C. albicans, E. coli Planktonic  Inhibition zone 1 mg/mL* - [117]
cu-SEDDS 1% E. aureus, E. coli, P. aeruginosa, K. pneumonia  Planktonic  MIC 45–62 µg/mL - [118]
CUR:NE in microbeads 0.5 mg/mL E. coli, S. typhmerium,Y. enterocolitica, P. aeruginosa, S, aureus, B. cereus, L. monocytogenes Planktonic  Inhibition zone 90 and 180 mg/mL* - [119]
Lignin sulfomethylated 0.3 mg/mL S. aureus  Planktonic  OD600 nm 2.4 mg/mL* - [120]
C14-EDA/GM/WC14-MEDA/GM/W N/R C. albicans  Planktonic, biofilm  Microdilution assay, antibiofilm 100 µg/mL, 20 µg - [121]

[CUR]: CUR concentration. -: not performed. N/R: not reported. *: formulation concentration.

3.5. CUR in Cyclodextrin

Cyclodextrins (CDs) have revolutionized the pharmaceutical industry in recent years [122]. CDs consist of three naturally occurring oligosaccharides in a cyclic structure produced from starch [123][124][125]. The natural CDs have their nomenclature system and their chemical structure based on the number of glucose residues in their structure: 6, 7, or 8 glucose units, which are denominated α-CD, β-CD, and γ-CD, respectively [126][127]. Although the entire CD molecule is soluble in water, the interior is relatively non-polar and creates a hydrophobic microenvironment. Therefore, CDs are cup-shaped, hollow structures with an outer hydrophilic layer and an internal hydrophobic cavity (Figure 5) [126]. They can sequester insoluble compounds within their hydrophobic cavity, resulting in better solubility and consequently better chemical and enzymatic stability [124]. Due to the cavity size, β-CD forms appropriate inclusion complexes with molecules with aromatic rings [128], such as CUR [129]. Antimicrobial studies with CUR in CDs are summarized in Table 5.

Figure 5. Schematic representation of CUR in CD.

Table 5. Antimicrobial studies performed with CUR in CDs.

Type of CD [CUR] Formulation Microorganism Type of Culture Antimicrobial Method Antimicrobial [CUR] Light/Ultrasonic Parameters Reference
PEG-based β-CD or γ-CD 10 µM E. coli, E. faecalis Planktonic aPDT 10 µM 4.8, 29 J/cm2 [130]
HPMC-stabilized hydroxypro pyl-β-CD 7.64 × 10−3 M E. coli Planktonic aPDT 10, 25 µM 5, 14, 28 J/cm2 [131]
methyl-β-CD hyaluronic acid HPMC 7.64 × 10−3 M E. faecalis, E. coli Planktonic aPDT 0.5–25 µM 11, 16, 32 J/cm2 [132]
carboxymethyl-β-CD 20 µM E. coli Planktonic aPDT 0.7 ± 0.1 to 4.1 ± 1.6  nmole cm−2 1050 ± 250 lx [133]
hydrogel with CUR in hydroxypropyl-β-CD 15.8 mg/mL S. aureus Planktonic Inhibition zone 2% (w/v) - [134]
α- and β-CD 1 mol/L E. coli, S. aureus Planktonic MIC, OD600 nm 0.25 and 0.31 mg/mL - [135]
β-CD or γ-CD in CS 0.06 mM E. coli, S. aureus Planktonic MIC, Zone of inhibition 64 and 32 µg/mL - [136]
γ-CD 25 mg/L T. rubrum Planktonic MIC, aPDT N/R 45 J/cm2 [137]
hydroxypropyl-β-CD 1:1 B. subtillis, S. aureus, S. pyrogenes, P. aeruginosa, C. difficile, C. butyricum, L. monocytogenes, E. faecalis, E. coli, K. pneumoniae, P. mirabilis, S. typhimurium, E. aerogens, C. kusei, C. albicans Planktonic Inhibition zone 25 mg/mL - [138]
methyl-β-CD 20 mM E. coli Planktonic MIC, MBC, aPDT 500, 90 µM 9 J/cm2 [139]

[CUR]: CUR concentration. -: not performed. N/R: not reported.

3.6. CUR in Chitosan

Chitin is a natural polysaccharide commonly found in the exoskeleton of marine crustaceans such as shrimps, prawns, lobsters, and crabs. Chitosan (CS) derives from the acetylation of chitin and has a linear structure of D-glucosamine (deacetylated monomer) linked to N-acetyl-D-glucosamine (acetylated monomer) through β-1,4 bonds [140]. The main advantages that make CS a promising drug carrier include biocompatibility, biodegradability, non-toxicity, controlled release system, mucoadhesive properties, and low cost [140][141]. Moreover, CS is soluble in aqueous solutions and is the only pseudo-natural polymer with a positive charge (cationic) [142], which can interact with negatively-charged DNA, membranes of microbial cells, and biofilm matrix [143]. Antimicrobial studies with CUR in CS are summarized in Table 6.

Table 6. Antimicrobial studies performed with CUR in CS.

Type ofCS [CUR] Formulation Microorganism Type of Culture Antimicrobial Method Antimicrobial [CUR] Reference
PEG-CS 4.4%, 5 mg/mL MRSA, P. aeruginosa Planktonic, Animal model OD600nm, CFU 5 and 10 mg/mL * [144]
CCS microspheres 12.27 mg/mL, 1 mol S. aureus, E. coli Planktonic Zone of inhibition, MIC N/R [145]
CS nanoparticles 1.06 mg/mL S. mutans Planktonic, Biofilm MIC 0.114 mg/mL [146]
CS-CMS-MMT 0.0004–0.004 g S. mutans Planktonic, Biofilm MIC 0.101 mg/mL [147]
CS-GP-CUR 148.09 ± 5.01 µg S. aureus Planktonic Zone of inhibition, tissue bacteria count N/C [148]
PVA-CS-CUR N/C E. coli, P. aeruginosa, S. aureus, B. subtilis Planktonic Zone of inhibition N/R [149]
PVA-CS-CUR 10, 20, 30 mg P. multocida, S. aureus, E. coli, B. subtilis Planktonic Zone of inhibition 10, 20, 30 mg [150]
CS NPs 2, 4, 8, 16% C. albicans, S. aureus Planktonic, Biofilm MIC, Colony count 400 mg/mL [151]
CS NPs 4 mg/mL HCV-4 N/R Antiviral assay 15 µg/mL [152]
CS/milk protein nanocomposites 100 mg PVY Plant infection Antiviral activity 500, 1000, 1500 mg/100 mL [153]
[CUR]: CUR concentration. N/R: not reported. N/C: not clear. *: formulation concentration.

3.7. CUR in Other Polymeric DDS

Antimicrobial studies with CUR loaded in other polymeric DDSs are summarized in Table 7.

Table 7. Antimicrobial studies performed with curcumin in polymeric drug delivery systems.

Type of Polymeric DDS [CUR] Formulation Microorganism Type of Culture Antimicrobial Method Antimicrobial [CUR] Light/Ultrasonic Parameters Reference
PEG 400γ-CD and PEG + β-CD 0.18% E. faecalis, E. coli Planktonic CFU/mL aPDT N/R 9.7 J/cm2 29 J/cm2 [154]
CUR-NP without polymer 100 mg S. aureus, B. subtillis, E. coli, P. aeruginosa, P. notatum, A. niger Planktonic MIC Inhibition zone 100 mg, 0.27 mmol - [155]
CUR-NP without polymer 100 mg M. lutues, S. aureus, E. coli, P. aeruginosa Planktonic MBC N/R - [156]
Mixed polymer NP 5 mM E. coli Planktonic MIC 400–500 μM - [157]
CTABTween 20Sodium dodecylsulfate 100 mg/mL L. monicytogenes Planktonic Inhibition zone N/R - [158]
PLA/dextran sulfate 4 mg/mL MRSA, C. albicans, S. mutans Planktonic/mono- and –mixed biofilm aPDT 260 μM 43.2 J/cm2 [159]
PLA/dextran sulfate 0.4% C. albicans Animal model aPDT 260 μM 37.5 J/cm2 [160]
Nanocurcumin N/R P. aeruginosa (isolates) and standard strain Planktonic MIC 128 µg/mL - [161]
PLGA 5 mg S. saprophyticus subsp. Bovis, E. coli Planktonic aPDT 50 µg/mL 13.2 J/cm2 [162]
Eudragit L-100 N/C L. monocytogenes Planktonic Animal model infection N/R - [163]
nCUR N/R S. mutans PlanktonicBiofilm Inhibition zoneaPDT N/R 300–420 J/cm2 [164]
nCUR combined with indocyanine 100 mg E. faecalis Biofilm Metabolic activity N/R 500 mW/cm2 [165]
PVAc-CUR-PET-PVDC 0.02 g S. aureus, S. tiphimurium Planktonic aPDT N/R 24, 48, and 72 J/cm2 [166]
MOA.CUR-PLGA-NP Up to 10% S. mutans Biofilm aPDT 7% wt 45 J/cm2 [167]
CS- β-CD N/C S. aureus, E. coli Planktonic Colony count Up to 0.03% - [168]
[CUR]: CUR concentration. -: not performed. N/R: not reported. N/C: not clear.

3.8. CUR with Metallic Nanoparticles

Metal complexation plays an important role in the therapeutic properties of CUR. The β-diketone moiety in the CUR chemical structure enables it to form complexes with metal ions [169]. A previous review summarized the antimicrobial activity of CUR and curcuminoid complexes with metals, such as boron, Ca2+, Cd2+, Cr3+, Co2+, Cu2+, Fe3+, Ga3+, Hg2+, In3+, Mn2+, Ni2+, Pd2+, Sn2+, Y3+, and Zn2+ against viruses, bacteria, and fungi [169]. Metals have also been combined with polymers to improve the biological effects of CUR and to be used as films, hydrogels, dressings, and other pharmaceutical formulations [170][171]. In this context, silver NPs (AgNPs) have been extensively used due to their antimicrobial activity (Figure 6) [172]. Antimicrobial studies with CUR complexes with metals are summarized in Table 8.

Figure 6. Schematic representation of CUR in silver nanoparticles.

Table 8. Antimicrobial studies performed with CUR complexes with metallic NPs.

Type of Metallic Material [CUR] Formulation Microorganism Type of Culture Antimicrobial Method Antimicrobial [CUR] Reference
CUR-AgNPs 20 mg/mL P. aeruginosa, E. coli, B. subtilis, S. aureus Planktonic MIC 20 mg/mL [173]
Ag-CUR-nanoconjugates 0.1 mM E. coli, Salmonella spp., Fusarium spp., S. aureus Planktonic Zone of Inhibition 0.1 mM [174]
AgCURNPs 500 mg P. aeruginosaS. aureus Biofilm CLSM SEM Up to 400 μg/mL [175]
AgNPs 7 mg E. coli Planktonic Turbidimetric Assay 0.005 µM [176]
cAgNPs 7 mg E. coliB. subtilis Planktonic MIC, CFU/mL 7 mg [177]
Ru II complex 0.092 g E. coli, S. aureus, K. pneumoniae, A. baumannii, P. aeruginosa, Enterococcus sp. Plakntonic MIC/FICI >64 µg/mL [178]
SCMC SNCF nanocomposites with CUR 0.25 mg/mL E. coli Planktonic Disc Method Count Method 2 mg/mL [179]
CSCL CUR-AgNP 0.092 g E. coli, B. subtilis Planktonic Zone of Inhibition 10 and 20 μM [180]
nSnH 10% S. aureus, E. coli. Planktonic CFU/mL N/R [181]
Nanocomposite of CUR and ZnO NPs N/C S. epidermidis, S. hemolyticus, S. saprophyticus Planktonic Zone of Inhibition 1000, 750, 500, 250 μg/mL [182]
Thermo-responsive hydrogels N/C S. aureusP. aeruginosaE. coli Planktonic MIC 400 μg/mL [183]
CUR-AgNPs 5 mg/mL C. albicans, C. glabrata, C. tropicalis, C. parapsilosis, C. krusei, C. kefyr Planktonic Zone of Inhibition, MIC 32.2–250 μg/mL [184]
Gel-CUR-Ag 20 mg P. aeruginosa, S. aureus Planktonic MIC, MBC 20 mg [185]
HGZ-CUR N/C S. aureus, T. rubrum Planktonic Zone of Inhibition N/C [186]
CHG-ZnO-CUR N/C S. aureus, T. rubrum Planktonic Zone of Inhibition N/C [187]
Copper (II) oxide NPs 1 g E. faecalis, P. aeruginosa Planktonic Zone of Inhibition CFU/mL 1 mg/mL [188]
OA-Ag-C 1 g P. aeruginosa, S. aureus Planktonic OD600nm 2.5 mg/mL [189]
Ag-NP-β-CD-BC 0.79 g P. aeruginosa, S. aureus, C. auris Planktonic Zone of Inhibition N/R [190]
Cotton fabrics coated ZnO-NP 2.71 × 10−3 M S. aureus, E. coli Planktonic Bacterial Count N/R [191]
CS-ZnO-CUR 0.2 g S. aureus, E. coli Planktonic MIC, MBC Up to 50 μg/mL [192]
CUR-TiO2 -CS 100–300 mg S. aureus, E. coli Planktonic Animal infection MIC 10 mg [193]
CUR-Au-NPs 1 mg/mL E. coli, B. subtilis, S. aureus, P. aeruginosa Planktonic Zone of Inhibition 100, 200, 300 μg/mL [194]
[CUR]: CUR concentration. N/R: not reported. N/C: not clear.

3.9. CUR in Mesoporous Particles

Porous materials are structures with ordered pores ranging from nanometer to micrometers, which are classified as microporous (less 2 nm), mesoporous (from 2 up to 50 nm), and macroporous (above 50 nm) [195]. Porous materials can be synthesized using carbon, silica, and metal oxides [196]. Mesoporous silica nanoparticles (MSN, Figure 7) are inorganic scaffolds [197], which seemed ideal carriers for hydrophobic drugs due to their well-defined structure, large specific surface area, and versatile chemistry for functionalization [198]. The pore size and volume and the surface area, as well as the surface functionalization of the mesoporous material, determine the drug load and release [199]. Moreover, mesoporous materials can be modified or functionalized to control drug release under environmental stimuli, such as pH, temperature, or light. These stimuli-responsive DDS, or smart DDS, prevent undesirable drug release before reaching the target tissue (“zero premature release”) [199]. Antimicrobial studies with CUR in porous DDSs are summarized in Table 9.

Figure 7. Schematic representation of a porous particle.

Table 9. Antimicrobial studies performed with CUR in porous DDSs.

Porous DDS [CUR] Formulation Microorganism Type of Culture Antimicrobial Method Antimicrobial [CUR] Light/Ultrasonic Parameters Reference
Cu-SNP/Ag 1.0 mmol E. coli Planktonic aPDT N/R 72 J/cm2 [200]
Bionanocomposite silica/chitosan 100 mg E. coli, S. aureus Planktonic Zone of inhibition N/R - [201]
NCIP 1 mg HIV-1 Transfected cells Immuno fluorescent staining 5–8 mg/mL - [202]
Lollipop-like MSN 30 mg L−1 E. coli, S. aureus Planktonic OD600nm N/R - [203]
SBA-15/PDA/Ag 2 mg E. coli, S. aureus Planktonic CFU/mL 50 mM - [204]
[CUR]: CUR concentration. -: not performed. N/R: not reported.

3.10. CUR in Quantum Dots

Quantum dots (QDs) are semiconductor particles at nanosize (up to 10 nm) with electrical and photoluminescence properties of biotechnological and biomedical applications, such as bioimaging and DDS [205]. Carbon dots are divided into carbon QD and graphene QD and are produced by top-down and bottom-up methods using bulk carbon material and molecular precursors, respectively [205]. Antimicrobial studies with CUR in QDs are summarized in Table 10.

Table 10. Antimicrobial studies performed with CUR in quantum dots (QDs).

Type of
Material
[CUR]
Formulation
Microorganism Type of
Culture
Antimicrobial
Method
Antimicrobial
[CUR]
Light/Ultrasonic
Parameters
Reference
CUR-cQDs 0.6 S. aureus
MRSA
E. faecalis
E. coli
K. pneumoniae
P. aeruginosa
Planktonic
Biofilm
Grown
inhibition
Biomass
evaluation
Confocal
microscopy
3.91–
7.825 µg/mL
- [206]
CUR-cQDs 200 mg EV-71 Cell infection
Animal
infection
MIC
Plaque assay
TC IC50 assay
Western blot
PCR
5 µg/mL - [207]
CUR-MQD 2:1 wt% K. pneumoniae
P. aeruginosa
S. aureus
Planktonic MIC
MBC
Confocal
microscopy
Fluorescence
microscopy
Flow
cytometry
<0.00625–
0.125 µg/mL
- [208]
CUR-GQDs N/C A. actinomycetemcomitans
P. gingivalis
P. intermedia
Mixed biofilm aPDT 100 µg/mL 60–80 J/cm2 [209]

3.11. CUR in Films, Hydrogels, and Other Nanomaterials

Antimicrobial studies with CUR in films, hydrogels, and other nanomaterials are summarized in Table 11.

Table 11. Antimicrobial studies performed with CUR in films, hydrogels, and other nanomaterials.

Type of Material [CUR] Formulation Microorganism Type of Culture Antimicrobial Method Antimicrobial [CUR] Light/Ultrasonic Parameters Reference
CuR-SiNPs 20 mg S. aureus, P. aeruginosa Planktonic, Biofilm aPDT 50 μg/mL, 1 mg/mL 20 J/cm2 [210]
CUR-HNT-DX 10 mg S. marcescens, E. coli Planktonic, Infection model Grown inhibition, Confocal microscopy Up to 0.5 mg/mL - [211]
Exosomes N/R HIV-1 infection - Flow cytometry N/R - [212]
Electrospun nanofibers 100 mg/mL Actinomyces naeslundii Biofilm aPDT 2.5 and 5 mg/mL 1200 mW/cm2 [213]
Ga NFCD-GO NF 0.1 mol B. cereus, E. coli Planktonic Zone of inhibition, MIC Up to 63.25 µg/mL - [214]
Multinanofibers-film 1, 2.5, and 5 mg/mL S. aureusE. coli Planktonic UFC/mL, Confocal microscopy 1 mg/mL - [215]
Nanofibers scaffolds 4.0 wt% S. aureus Pseudomonassp. Planktonic Colony count N/R ,- [216]
Nanofibrous scaffold 5% S. aureus, E. coli Planktonic Colony count 20 mg - [217]
Nanofibers 5 and 10%wt S. aureusE. coli Planktonic OD600nm Up to 212.5 µg/mL - [218]
CSDG 1 w/w S. aureus, E. coli Planktonic, Infection model Colony count, Microscopy N/R - [219]
Gelatin film 0, 0.25, 0.5, 1.0, and 1.5 wt% E. coli, L. monocytogenes Planktonic UFC/mL 0.25 and 1.5 wt% - [220]
ZnO-CMC film 0.5 and 1.0 wt% E. coli, L. monocytogenes Planktonic UFC/mL 1 wt% - [221]
Pectin film 40 mg E. coli, L. monocytogenes Planktonic UFC/mL N/R - [222]
Edible film 0.4% (w/v) E. coli, B. subtilis Planktonic Zone of inhibition 1% wt. - [223]
[CUR]: CUR concentration. -: not performed. N/R: not reported. N/C: not clear

4. Conclusions and Future Perspectives

CUR has a broad-spectrum antimicrobial activity against viruses, bacteria, and fungi, including resistant and emergent pathogens. However, some species such as Gram-negative bacteria are less susceptible to CUR and aPDT. For those, the combination of CUR with antibiotics has been suggested, especially for antibiotic-resistant strains [220]. CUR showed synergism with polymyxin and protection against the side effects of polymyxin treatment, nephrotoxicity, and neurotoxicity [224]. Nonetheless, the evaluation of synergism requires accurate methods to study drug interaction, considering potential differences between the dose–response relationship of individual drugs and avoiding over- or under-estimation of interactions. For example, while the time–kill curve of C. jejuni treated with both cinnamon oil and ZnO NPs resulted in the over-estimation of synergism between the antimicrobials, the fractional inhibitory concentration index (FICI) method showed no synergism but only an additive effect [225]. The FICI method was not able to detect the synergism between binary combinations of antimicrobials (cinnamon oil, ZnO NPs, and CUR encapsulated in starch) at sub-MIC, which resulted in the non-turbidity of C. jejuni. In turn, mathematical modeling using isobolograms and median-effect curves showed synergism when CUR in starch was combined with other antimicrobials against C. jejuni, with bacterial reductions of 3 log for the binary combination and over 8 log for the tertiary combination. The mathematical modeling suggested that CUR in starch was the main antimicrobial responsible for the synergistic interaction [225].

In addition to the antimicrobial evaluation, in vitro and in vivo studies have demonstrated the cytocompatibility and biocompatibility of CUR in DDSs [108][113][114][115][118][121][144][145][152][156][168][185][192][202][209], suggesting that CUR-loaded DDSs might be safe. Although a plethora of DDSs has been developed to circumvent the hydrophobicity, instability in solution, and low bioavailability of CUR, several studies are still performed with free CUR dissolved in organic solvents [19][20][21][22][23][24][25][26][27][28][29][30][31][32][33][34][35][36][37][38][39][40][41][42][43][44][45][46][47][48][49][50][51][52][53][54][55][56][57][58][59][60][61][62][63][64][65][66][67][68][69][70][71][72][85][86][87][88][89][90]. Furthermore, compared to several in vitro investigations, few in vivo studies using animal infection models and scarce clinical trials have been reported. A randomized clinical trial showed that aPDT mediated by free CUR improved gingivitis in adolescents under fixed orthodontic treatment but did not reduce dental plaque accumulation after 1 month [226]. Clinical improvements after CUR-mediated aPDT were also observed for periodontal diseases, although few studies have evaluated the microbiological parameters [63][89]. Therefore, the improvement of clinical parameters might be due to the anti-inflammatory effect of CUR/aPDT instead of their in vivo antimicrobial activity. Nonetheless, randomized clinical trials evaluating CUR in DDSs against infections are required.

As a note on the future use of CUR, the incorporation of CUR in DDS and other pharmaceutical formulations allows its clinical use especially as an adjuvant agent to conventional antimicrobial agents. Such a combination can be an important weapon in the battle against resistant strains and emergent pathogens. The use of stimuli-responsive (or smart) DDS can also improve CUR delivery and its therapeutic effect on the target tissue. The combination of polymeric and metallic carriers may also enhance the therapeutic activity of CUR. Nonetheless, the degradation of DDS and its clearance from the body are other issues that require further investigation [18]. The evidence produced so far about the antimicrobial activity of CUR in DDSs supports future in vivo and clinical studies, which may pave the way for industrial production.

References

  1. Charles E Davis; Antonella Rossati; Olivia Bargiacchi; Pietro Luigi Garavelli; Anthony J McMichael; Gunnar Juliusson; Globalization, Climate Change, and Human Health. New England Journal of Medicine 2013, 369, 94-96, 10.1056/nejmc1305749.
  2. Lance Saker; Kelley Lee; Barbara Cannito; Anna Gilmore; Diarmid Campbell-Lendrum; Globalization and infectious diseases: a review of the linkages. WHO Special Programme on Tropical Diseases Research 2004, 3, 3-12.
  3. Shaghayegh Gorji; Ali Gorji; COVID-19 pandemic: the possible influence of the long-term ignorance about climate change. Environmental Science and Pollution Research 2021, 28, 15575-15579, 10.1007/s11356-020-12167-z.
  4. Jyoti Tanwar; Shrayanee Das; Zeeshan Fatima; Saif Hameed; Multidrug Resistance: An Emerging Crisis. Interdisciplinary Perspectives on Infectious Diseases 2014, 2014, 1-7, 10.1155/2014/541340.
  5. Maya A. Farha; Eric D. Brown; Drug repurposing for antimicrobial discovery. Nature Microbiology 2019, 4, 565-577, 10.1038/s41564-019-0357-1.
  6. Bradley S. Moore; Guy T. Carter; Mark Brönstrup; Editorial: Are natural products the solution to antimicrobial resistance?. Natural Product Reports 2017, 34, 685-686, 10.1039/c7np90026k.
  7. Thaís Soares Bezerra Santos Nunes; Leticia Matheus Rosa; Yuliana Vega-Chacón; Ewerton Garcia De Oliveira Mima; Fungistatic Action of N-Acetylcysteine on Candida albicans Biofilms and Its Interaction with Antifungal Agents. Microorganisms 2020, 8, 980, 10.3390/microorganisms8070980.
  8. Susan J. Hewlings; Douglas S. Kalman; Curcumin: A Review of Its Effects on Human Health. Foods 2017, 6, 92, 10.3390/foods6100092.
  9. Subash C. Gupta; Sridevi Patchva; Bharat B. Aggarwal; Therapeutic Roles of Curcumin: Lessons Learned from Clinical Trials. The AAPS Journal 2012, 15, 195-218, 10.1208/s12248-012-9432-8.
  10. Mhd Anas Tomeh; Roja Hadianamrei; Xiubo Zhao; A Review of Curcumin and Its Derivatives as Anticancer Agents. International Journal of Molecular Sciences 2019, 20, 1033, 10.3390/ijms20051033.
  11. Michele Dei Cas; Riccardo Ghidoni; Dietary Curcumin: Correlation between Bioavailability and Health Potential. Nutrients 2019, 11, 2147, 10.3390/nu11092147.
  12. Kavirayani Indira Priyadarsini; The Chemistry of Curcumin: From Extraction to Therapeutic Agent. Molecules 2014, 19, 20091-20112, 10.3390/molecules191220091.
  13. Deljoo Somayeh; Rabiee Navid; Rabiee Mohammad; Curcumin-hybrid Nanoparticles in Drug Delivery System (Review). Asian J. Nanosci. Mater 2019, 2, 66–91, 10.26655/AJNANOMAT.2019.1.5.
  14. Javad Safari; Zohre Zarnegar; Advanced drug delivery systems: Nanotechnology of health design A review. Journal of Saudi Chemical Society 2014, 18, 85-99, 10.1016/j.jscs.2012.12.009.
  15. Dimas Praditya; Lisa Kirchhoff; Janina Brüning; Heni Rachmawati; Joerg Steinmann; Eike Steinmann; Anti-infective Properties of the Golden Spice Curcumin. Frontiers in Microbiology 2019, 10, 912, 10.3389/fmicb.2019.00912.
  16. Soheil Zorofchian Moghadamtousi; Habsah Abdul Kadir; Pouya Hassandarvish; Hassan Tajik; Sazaly Abubakar; Keivan Zandi; A Review on Antibacterial, Antiviral, and Antifungal Activity of Curcumin. BioMed Research International 2014, 2014, 1-12, 10.1155/2014/186864.
  17. Anderson Clayton da Silva; Priscila Dayane De Freitas Santos; Jéssica Thais Do Prado Silva; Fernanda Vitória Leimann; Lívia Bracht; Odinei Hess Gonçalves; Impact of curcumin nanoformulation on its antimicrobial activity. Trends in Food Science & Technology 2018, 72, 74-82, 10.1016/j.tifs.2017.12.004.
  18. Simin Sharifi; Nazanin Fathi; Mohammad Yousef Memar; Seyed Mahdi Hosseiniyan Khatibi; Rovshan Khalilov; Ramin Negahdari; Sepideh Zununi Vahed; Solmaz Maleki Dizaj; Anti‐microbial activity of curcumin nanoformulations: New trends and future perspectives. Phytotherapy Research 2020, 34, 1926-1946, 10.1002/ptr.6658.
  19. Yaning Gao; WanBo Tai; Ning Wang; Xiang Li; Shibo Jiang; Asim K. Debnath; Lanying Du; Shizhong Chen; Gao; Tai; et al.WangLiDuChen Identification of Novel Natural Products as Effective and Broad-Spectrum Anti-Zika Virus Inhibitors. Viruses 2019, 11, 1019, 10.3390/v11111019.
  20. Mayuri Patwardhan; Mark T. Morgan; Vermont Dia; Doris H. D'Souza; Heat sensitization of hepatitis A virus and Tulane virus using grape seed extract, gingerol and curcumin. Food Microbiology 2020, 90, 103461, 10.1016/j.fm.2020.103461.
  21. He Li; Canrong Zhong; Qian Wang; Weikang Chen; Yan Yuan; Curcumin is an APE1 redox inhibitor and exhibits an antiviral activity against KSHV replication and pathogenesis. Antiviral Research 2019, 167, 98-103, 10.1016/j.antiviral.2019.04.011.
  22. Wael H. Roshdy; Helmy A. Rashed; Ahmed Kandeil; Ahmed Mostafa; Yassmin Moatasim; Omnia Kutkat; Noura M. Abo Shama; Mokhtar R. Gomaa; Ibrahim H. El-Sayed; Nancy M. El Guindy; et al.Amal NaguibGhazi KayaliMohamed A. Ali EGYVIR: An immunomodulatory herbal extract with potent antiviral activity against SARS-CoV-2. PLOS ONE 2020, 15, e0241739, 10.1371/journal.pone.0241739.
  23. Chih-Chun Wen; Yueh-Hsiung Kuo; Jia-Tsrong Jan; Po-Huang Liang; Sheng-Yang Wang; Hong-Gi Liu; Ching-Kuo Lee; Shang-Tzen Chang; Chih-Jung Kuo; Shoei-Sheng Lee; et al.Chia-Chung HouPei-Wen HsiaoShih-Chang ChienLie-Fen ShyurNing-Sun Yang Specific Plant Terpenoids and Lignoids Possess Potent Antiviral Activities against Severe Acute Respiratory Syndrome Coronavirus. Journal of Medicinal Chemistry 2007, 50, 4087-4095, 10.1021/jm070295s.
  24. Young Bae Ryu; Su-Jin Park; Young Min Kim; Ju-Yeon Lee; Woo Duck Seo; Jong Sun Chang; Ki Hun Park; Mun-Chual Rho; Woo Song Lee; SARS-CoV 3CLpro inhibitory effects of quinone-methide triterpenes from Tripterygium regelii. Bioorganic & Medicinal Chemistry Letters 2010, 20, 1873-1876, 10.1016/j.bmcl.2010.01.152.
  25. Ji-Young Park; Hyung Jae Jeong; Jang Hoon Kim; Young Min Kim; Su-Jin Park; Doman Kim; Ki Hun Park; Woo Song Lee; Young Bae Ryu; Diarylheptanoids from Alnus japonica Inhibit Papain-Like Protease of Severe Acute Respiratory Syndrome Coronavirus. Biological and Pharmaceutical Bulletin 2011, 35, 2036-2042, 10.1248/bpb.b12-00623.
  26. Oluyomi Stephen Adeyemi; Joy Ihuoma Obeme-Imom; Benjamin Oghenerobor Akpor; Damilare Rotimi; Gaber El-Saber Batiha; Akinyomade Owolabi; Altered redox status, DNA damage and modulation of L-tryptophan metabolism contribute to antimicrobial action of curcumin. Heliyon 2020, 6, e03495, 10.1016/j.heliyon.2020.e03495.
  27. Mümtaz Güran; Gizem Şanlıtürk; Namık Refik Kerküklü; Ergül Mutlu Altundag; A. Süha Yalçın; Combined effects of quercetin and curcumin on anti-inflammatory and antimicrobial parameters in vitro. European Journal of Pharmacology 2019, 859, 172486, 10.1016/j.ejphar.2019.172486.
  28. Mirian Aa Freitas; André Hc Pereira; Juliana G Pinto; Adriana Casas; Juliana Ferreira-Strixino; Bacterial viability after antimicrobial photodynamic therapy with curcumin on multiresistant Staphylococcus aureus. Future Microbiology 2019, 14, 739-748, 10.2217/fmb-2019-0042.
  29. Camilo Geraldo De Souza Teixeira; Paula Volpato Sanitá; Ana Paula Dias Ribeiro; Luana Mendonça Dias; Janaina Habib Jorge; Ana Cláudia Pavarina; Antimicrobial photodynamic therapy effectiveness against susceptible and methicillin-resistant Staphylococcus aureus biofilms. Photodiagnosis and Photodynamic Therapy 2020, 30, 101760, 10.1016/j.pdpdt.2020.101760.
  30. Farheen Akhtar; Asad U. Khan; Lama Misba; Kafil Akhtar; Asif Ali; Antimicrobial and antibiofilm photodynamic therapy against vancomycin resistant Staphylococcus aureus (VRSA) induced infection in vitro and in vivo. European Journal of Pharmaceutics and Biopharmaceutics 2021, 160, 65-76, 10.1016/j.ejpb.2021.01.012.
  31. Fernanda Rossi Paolillo; Phamilla Gracielli Sousa Rodrigues; Vanderlei Salvador Bagnato; Fernanda Alves; Layla Pires; Adalberto Vieira Corazza; The effect of combined curcumin-mediated photodynamic therapy and artificial skin on Staphylococcus aureus–infected wounds in rats. Lasers in Medical Science 2020, 20, 1-8, 10.1007/s10103-020-03160-6.
  32. Yali Li; Yi Xu; Qiaoming Liao; Mengmeng Xie; Han Tao; Hui‐Li Wang; Synergistic effect of hypocrellin B and curcumin on photodynamic inactivation of Staphylococcus aureus. Microbial Biotechnology 2021, 14, 692-707, 10.1111/1751-7915.13734.
  33. Thaila Quatrini Corrêa; Kate Cristina Blanco; Érica Boer Garcia; Shirly Marleny Lara Perez; Daniel José Chianfrone; Vinicius Sigari Morais; Vanderlei Salvador Bagnato; Effects of ultraviolet light and curcumin-mediated photodynamic inactivation on microbiological food safety: A study in meat and fruit. Photodiagnosis and Photodynamic Therapy 2020, 30, 101678, 10.1016/j.pdpdt.2020.101678.
  34. Truong Dang Le; Pimonpan Phasupan; Loc Thai Nguyen; Antimicrobial photodynamic efficacy of selected natural photosensitizers against food pathogens: Impacts and interrelationship of process parameters. Photodiagnosis and Photodynamic Therapy 2020, 32, 102024, 10.1016/j.pdpdt.2020.102024.
  35. Davy-Louis Versace; Gabriela Moran; Mehdi Belqat; Arnaud Spangenberg; Rachel Meallet-Renault; Samir Abbad-Andaloussi; Vlasta Brezova; Jean-Pierre Malval; Highly Virulent Bactericidal Effects of Curcumin-Based μ-Cages Fabricated by Two-Photon Polymerization. ACS Applied Materials & Interfaces 2020, 12, 5050-5057, 10.1021/acsami.9b18693.
  36. Fernanda Alves; Gabriela Gomes Guimarães; Natália Mayumi Inada; Sebastião Pratavieira; Vanderlei Salvador Bagnato; Cristina Kurachi; Strategies to Improve the Antimicrobial Efficacy of Photodynamic, Sonodynamic, and Sonophotodynamic Therapies. Lasers in Surgery and Medicine 2021, null, 1-9, 10.1002/lsm.23383.
  37. Rangel-Castañeda Itzia Azucena; Cruz-Lozano José Roberto; Zermeño-Ruiz Martin; Cortes-Zarate Rafael; Hernández-Hernández Leonardo; Tapia-Pastrana Gabriela; Castillo-Romero Araceli; Drug Susceptibility Testing and Synergistic Antibacterial Activity of Curcumin with Antibiotics against Enterotoxigenic Escherichia coli. Antibiotics 2019, 8, 43, 10.3390/antibiotics8020043.
  38. Javier I. Sanchez-Villamil; Fernando Navarro-Garcia; Araceli Castillo-Romero; Filiberto Gutiérrez-Gutiérrez; Daniel Tapia; Gabriela Tapia-Pastrana; Curcumin Blocks Cytotoxicity of Enteroaggregative and Enteropathogenic Escherichia coli by Blocking Pet and EspC Proteolytic Release From Bacterial Outer Membrane. Frontiers in Cellular and Infection Microbiology 2019, 9, 334, 10.3389/fcimb.2019.00334.
  39. Sawsan Kareem; Suhad Saad Mahmood; Nada Hindi; Effects of Curcumin and Silymarin on the Shigella dysenteriae and Campylobacter jejuni In vitro. Journal of Gastrointestinal Cancer 2019, 51, 824-828, 10.1007/s12029-019-00301-1.
  40. Yuan Gao; Juan Wu; Zhaojie Li; Xu Zhang; Na Lu; Changhu Xue; Albert Wingnang Leung; Chuanshan Xu; Qingjuan Tang; Curcumin-mediated photodynamic inactivation (PDI) against DH5α contaminated in oysters and cellular toxicological evaluation of PDI-treated oysters. Photodiagnosis and Photodynamic Therapy 2019, 26, 244-251, 10.1016/j.pdpdt.2019.04.002.
  41. Homa Darmani; Ehda A.M. Smadi; Sereen M.B. Bataineh; Blue light emitting diodes enhance the antivirulence effects of Curcumin against Helicobacter pylori. Journal of Medical Microbiology 2020, 69, 617-624, 10.1099/jmm.0.001168.
  42. Hayder Abdulrahman; Lama Misba; Shabbir Ahmad; Asad U. Khan; Curcumin induced photodynamic therapy mediated suppression of quorum sensing pathway of Pseudomonas aeruginosa: An approach to inhibit biofilm in vitro. Photodiagnosis and Photodynamic Therapy 2020, 30, 101645, 10.1016/j.pdpdt.2019.101645.
  43. Kai‐Chih Chang; Ya‐Yun Cheng; Meng‐Jiun Lai; Anren Hu; Identification of carbonylated proteins in a bactericidal process induced by curcumin with blue light irradiation on imipenem‐resistant Acinetobacter baumannii. Rapid Communications in Mass Spectrometry 2019, 34, e8548, 10.1002/rcm.8548.
  44. Javad Yasbolaghi Sharahi; Zahra Aliakbar Ahovan; Donya Taghizadeh Maleki; Zahra Riahi Rad; Zohreh Riahi Rad; Mehdi Goudarzi; Aref Shariati; Narjess Bostanghadiri; Elham Abbasi; Ali Hashemi; et al. In vitro antibacterial activity of curcumin-meropenem combination against extensively drug-resistant (XDR) bacteria isolated from burn wound infections.. Avicenna J Phytomed 2020, 10, 3-10.
  45. Jourdan E. Lakes; Christopher I. Richards; Michael D. Flythe; Inhibition of Bacteroidetes and Firmicutes by select phytochemicals. Anaerobe 2020, 61, 102145-102145, 10.1016/j.anaerobe.2019.102145.
  46. Shweta Kumari; Sundarraj Jayakumar; Gagan D. Gupta; Subhash C. Bihani; Deepak Sharma; Vijay Kumar Kutala; Santosh K. Sandur; Vinay Kumar; Antibacterial activity of new structural class of semisynthetic molecule, triphenyl-phosphonium conjugated diarylheptanoid. Free Radical Biology and Medicine 2019, 143, 140-145, 10.1016/j.freeradbiomed.2019.08.003.
  47. Prince Kumar; Shamseer Kulangara Kandi; Kasturi Mukhopadhyay; Diwan S. Rawat; Gagandeep; Synthesis of novel monocarbonyl curcuminoids, evaluation of their efficacy against MRSA, including ex vivo infection model and their mechanistic studies. European Journal of Medicinal Chemistry 2020, 195, 112276, 10.1016/j.ejmech.2020.112276.
  48. Carlos R. Polaquini; Luana G. Morão; Ana C. Nazaré; Guilherme S. Torrezan; Guilherme Dilarri; Lúcia B. Cavalca; Débora L. Campos; Isabel Silva; Jesse Augusto Pereira; Dirk-Jan Scheffers; et al.Cristiane DuqueFernando PavanHenrique FerreiraLuis O. Regasini Antibacterial activity of 3,3′-dihydroxycurcumin (DHC) is associated with membrane perturbation. Bioorganic Chemistry 2019, 90, 103031, 10.1016/j.bioorg.2019.103031.
  49. Milena Mattes Cerveira; Helena Silveira Vianna; Edila Maria Kickhofel Ferrer; Bruno Nunes da Rosa; Claudio Martin Pereira de Pereira; Matheus Dellaméa Baldissera; Leonardo Quintana Soares Lopes; Virginia Cielo Rech; Janice Luehring Giongo; Rodrigo De Almeida Vaucher; et al. Bioprospection of novel synthetic monocurcuminoids: Antioxidant, antimicrobial, and in vitro cytotoxic activities. Biomedicine & Pharmacotherapy 2020, 133, 111052, 10.1016/j.biopha.2020.111052.
  50. Michal Duracka; Norbert Lukac; Miroslava Kacaniova; Attila Kantor; Lukas Hleba; Lubomir Ondruska; Eva Tvrda; Antibiotics Versus Natural Biomolecules: The Case of In Vitro Induced Bacteriospermia by Enterococcus Faecalis in Rabbit Semen. Molecules 2019, 24, 4329, 10.3390/molecules24234329.
  51. Marisol Porto Rocha; Mariana Sousa Santos; Paôlla Layanna Fernandes Rodrigues; Thalita Santos Dantas Araújo; Janeide Muritiba de Oliveira; Luciano Pereira Rosa; Vanderlei Salvador Bagnato; Francine Cristina da Silva; Photodynamic therapy with curcumin in the reduction of enterococcus faecalis biofilm in bone cavity: rMicrobiological and spectral fluorescense analysis. Photodiagnosis and Photodynamic Therapy 2021, 33, 102084, 10.1016/j.pdpdt.2020.102084.
  52. Shamil Rafeeq; Setareh Shiroodi; Michael H. Schwarz; Nitin Nitin; Reza Ovissipour; Inactivation of Aeromonas hydrophila and Vibrio parahaemolyticus by Curcumin-Mediated Photosensitization and Nanobubble-Ultrasonication Approaches. Foods 2020, 9, 1306, 10.3390/foods9091306.
  53. Sivadas Ganapathy; Shan Sainudeen; Veena S Nair; Mohammad Zarbah; Anshad Mohamed Abdulla; Chawre Mustufa Najeeb; Can herbal extracts serve as antibacterial root canal irrigating solutions? Antimicrobial efficacy of Tylophora indica, Curcumin longa, Phyllanthus amarus, and sodium hypochlorite on Enterococcus faecalis biofilms formed on tooth substrate: In vitro study. Journal of Pharmacy And Bioallied Sciences 2019, 12, 423-S429, 10.4103/jpbs.jpbs_127_20.
  54. Arash Azizi; Parastoo Shohrati; Mehdi Goudarzi; Shirin Lawaf; Arash Rahimi; Comparison of the effect of photodynamic therapy with curcumin and methylene Blue on streptococcus mutans bacterial colonies. Photodiagnosis and Photodynamic Therapy 2019, 27, 203-209, 10.1016/j.pdpdt.2019.06.002.
  55. Jennifer Machado Soares; Karoliny Oliveira Ozias Silva; Natalia Mayumi Inada; Vanderlei Salvador Bagnato; Kate Cristina Blanco; Optimization for microbial incorporation and efficiency of photodynamic therapy using variation on curcumin formulation. Photodiagnosis and Photodynamic Therapy 2020, 29, 101652, 10.1016/j.pdpdt.2020.101652.
  56. Daniela Alejandra Cusicanqui Méndez; Eliezer Gutierrez; Giuliana Campos Chaves Lamarque; Veridiana Lopes Rizzato; Marília Afonso Rabelo Buzalaf; Maria Aparecida Andrade Moreira Machado; Thiago Cruvinel; The effectiveness of curcumin-mediated antimicrobial photodynamic therapy depends on pre-irradiation and biofilm growth times. Photodiagnosis and Photodynamic Therapy 2019, 27, 474-480, 10.1016/j.pdpdt.2019.07.011.
  57. Xinlong Li; Luoping Yin; Gordon Ramage; Bingchun Li; Ye Tao; Qinghui Zhi; Huancai Lin; Yan Zhou; Assessing the impact of curcumin on dual‐species biofilms formed by Streptococcus mutans and Candida albicans. MicrobiologyOpen 2019, 8, e937, 10.1002/mbo3.937.
  58. Marzie Mahdizade-Ari; Maryam Pourhajibagher; Abbas Bahador; Changes of microbial cell survival, metabolic activity, efflux capacity, and quorum sensing ability of Aggregatibacter actinomycetemcomitans due to antimicrobial photodynamic therapy-induced bystander effects. Photodiagnosis and Photodynamic Therapy 2019, 26, 287-294, 10.1016/j.pdpdt.2019.04.021.
  59. Hui Pan; Dongqing Wang; Fengqiu Zhang; In vitro antimicrobial effect of curcumin-based photodynamic therapy on Porphyromonas gingivalis and Aggregatibacter actinomycetemcomitans. Photodiagnosis and Photodynamic Therapy 2020, 32, 102055, 10.1016/j.pdpdt.2020.102055.
  60. Sarah Böcher; Johannes-Simon Wenzler; Wolfgang Falk; Andreas Braun; Comparison of different laser-based photochemical systems for periodontal treatment. Photodiagnosis and Photodynamic Therapy 2019, 27, 433-439, 10.1016/j.pdpdt.2019.06.009.
  61. Lucas Henrique De Paula Zago; Sarah Raquel de Annunzio; Kleber Thiago de Oliveira; Paula Aboud Barbugli; Belen Retamal Valdes; Magda Feres; Carla Raquel Fontana; Antimicrobial photodynamic therapy against metronidazole-resistant dental plaque bactéria. Journal of Photochemistry and Photobiology B: Biology 2020, 209, 111903, 10.1016/j.jphotobiol.2020.111903.
  62. Aram Mohammed Sha; Balkees Taha Garib; Aram Sha; Antibacterial Effect of Curcumin against Clinically Isolated Porphyromonas gingivalis and Connective Tissue Reactions to Curcumin Gel in the Subcutaneous Tissue of Rats.. BioMed Research International 2019, 2019, 6810936-14, 10.1155/2019/6810936.
  63. Camila Ayumi Ivanaga; Daniela Maria Janjacomo Miessi; Marta Aparecida Alberton Nuernberg; Marina Módolo Claudio; Valdir Gouveia Garcia; Leticia Helena Theodoro; Antimicrobial photodynamic therapy (aPDT) with curcumin and LED, as an enhancement to scaling and root planing in the treatment of residual pockets in diabetic patients: A randomized and controlled split-mouth clinical trial. Photodiagnosis and Photodynamic Therapy 2019, 27, 388-395, 10.1016/j.pdpdt.2019.07.005.
  64. Francine Cristina Da Silva; Luciano Pereira Rosa; Gabriel Pinto De Oliveira Santos; Natália Mayumi Inada; Kate Cristina Blanco; Thalita Santos Dantas Araújo; Vanderlei Salvador Bagnato; Total mouth photodynamic therapy mediated by blue led and curcumin in individuals with AIDS. Expert Review of Anti-infective Therapy 2020, 18, 689-696, 10.1080/14787210.2020.1756774.
  65. Veena S Narayanan; Sunil Muddaiah; R Shashidara; U S Sudheendra; N C Deepthi; Lakshman Samaranayake; Variable antifungal activity of curcumin against planktonic and biofilm phase of different candida species.. Indian Journal of Dental Research 2019, 31, 145-148, 10.4103/ijdr.IJDR_521_17.
  66. Yulong Tan; Matthias Leonhard; Doris Moser; Su Ma; Berit Schneider-Stickler; Antibiofilm efficacy of curcumin in combination with 2-aminobenzimidazole against single- and mixed-species biofilms of Candida albicans and Staphylococcus aureus. Colloids and Surfaces B: Biointerfaces 2019, 174, 28-34, 10.1016/j.colsurfb.2018.10.079.
  67. Francine Cristina da Silva; Paôlla Layanna Fernandes Rodrigues; Thalita Santos Dantas Araújo; Mariana Sousa Santos; Janeide Muritiba de Oliveira; Luciano Pereira Rosa; Gabriel Pinto De Oliveira Santos; Bruno Pereira de Araújo; Vanderlei Salvador Bagnato; Fluorescence spectroscopy of Candida albicans biofilms in bone cavities treated with photodynamic therapy using blue LED (450 nm) and curcumin. Photodiagnosis and Photodynamic Therapy 2019, 26, 366-370, 10.1016/j.pdpdt.2019.05.002.
  68. Jing Ma; Hang Shi; Hongying Sun; Jiyang Li; Yu Bai; Antifungal effect of photodynamic therapy mediated by curcumin on Candida albicans biofilms in vitro. Photodiagnosis and Photodynamic Therapy 2019, 27, 280-287, 10.1016/j.pdpdt.2019.06.015.
  69. Cláudia Carolina Jordão; Tábata Viana de Sousa; Marlise Inêz Klein; Luana Mendonça Dias; Ana Cláudia Pavarina; Juliana Cabrini Carmello; Antimicrobial photodynamic therapy reduces gene expression of Candida albicans in biofilms. Photodiagnosis and Photodynamic Therapy 2020, 31, 101825, 10.1016/j.pdpdt.2020.101825.
  70. Elisabetta Merigo; Marlène Chevalier; Stefania Conti; Tecla Ciociola; Carlo Fornaini; Maddalena Manfredi; Paolo Vescovi; Alain Doglio; Antimicrobial effect on Candida albicans biofilm by application of different wavelengths and dyes and the synthetic killer decapeptide KP. LASER THERAPY 2018, 28, 180-186, 10.5978/islsm.28_19-OR-14.
  71. Yuliana Vega-Chacón; Maria Carolina de Albuquerque; Ana Cláudia Pavarina; Gustavo Henrique Goldman; Ewerton Garcia De Oliveira Mima; Verapamil inhibits efflux pumps in Candida albicans, exhibits synergism with fluconazole, and increases survival of Galleria mellonella. Virulence 2020, 12, 231-243, 10.1080/21505594.2020.1868814.
  72. Amol A. Nagargoje; Satish Akolkar; Madiha M. Siddiqui; Dnyaneshwar D. Subhedar; Jaiprakash N. Sangshetti; Vijay M. Khedkar; Bapurao Babruwan Shingate; Quinoline Based Monocarbonyl Curcumin Analogs as Potential Antifungal and Antioxidant Agents: Synthesis, Bioevaluation and Molecular Docking Study. Chemistry & Biodiversity 2019, 17, null, 10.1002/cbdv.201900624.
  73. Morgan Jennings; Robin Parks; Curcumin as an Antiviral Agent. Viruses 2020, 12, 1242, 10.3390/v12111242.
  74. Dony Mathew; Wei-Li Hsu; Antiviral potential of curcumin. Journal of Functional Foods 2017, 40, 692-699, 10.1016/j.jff.2017.12.017.
  75. Divya M. Teli; Mamta B. Shah; Mahesh T. Chhabria; In silico Screening of Natural Compounds as Potential Inhibitors of SARS-CoV-2 Main Protease and Spike RBD: Targets for COVID-19. Frontiers in Molecular Biosciences 2021, 7, 599079, 10.3389/fmolb.2020.599079.
  76. Atala B. Jena; Namrata Kanungo; Vinayak Nayak; G.B.N. Chainy; Jagneshwar Dandapat; Catechin and Curcumin interact with corona (2019-nCoV/SARS-CoV2) viral S protein and ACE2 of human cell membrane: insights from Computational study and implication for intervention. Scientific Reports 2020, 11, 1-14, 10.21203/rs.3.rs-22057/v1.
  77. Mohit Kumar; Kushneet Kaur Sodhi; Dileep Kumar Singh; Addressing the potential role of curcumin in the prevention of COVID-19 by targeting the Nsp9 replicase protein through molecular docking. Archives of Microbiology 2021, 203, 1691-1696, 10.1007/s00203-020-02163-9.
  78. Ashish Patel; Malathi Rajendran; Ashish Shah; Harnisha Patel; Suresh B. Pakala; Prashanthi Karyala; Virtual screening of curcumin and its analogs against the spike surface glycoprotein of SARS-CoV-2 and SARS-CoV. Journal of Biomolecular Structure and Dynamics 2021, 5, 1-9, 10.1080/07391102.2020.1868338.
  79. Loubna Allam; Fatima Ghrifi; Hakmi Mohammed; Naima El Hafidi; Rachid El Jaoudi; Jaouad El Harti; Badreddine Lmimouni; Lahcen Belyamani; Azeddine Ibrahimi; Targeting the GRP78-Dependant SARS-CoV-2 Cell Entry by Peptides and Small Molecules. Bioinformatics and Biology Insights 2019, 14, 1177932220965505, 10.1177/1177932220965505.
  80. Mahmoud A.A. Ibrahim; Alaa H.M. Abdelrahman; Taha A. Hussien; Esraa A.A. Badr; Tarik A. Mohamed; Hesham R. El-Seedi; Paul W. Pare; Thomas Efferth; Mohamed-Elamir F. Hegazy; In silico drug discovery of major metabolites from spices as SARS-CoV-2 main protease inhibitors. Computers in Biology and Medicine 2020, 126, 104046-104046, 10.1016/j.compbiomed.2020.104046.
  81. Suresh Kumar; Priya Kashyap; Suman Chowdhury; Shivani Kumar; Anil Panwar; Ashok Kumar; Identification of phytochemicals as potential therapeutic agents that binds to Nsp15 protein target of coronavirus (SARS-CoV-2) that are capable of inhibiting virus replication. Phytomedicine 2021, 85, 153317-153317, 10.1016/j.phymed.2020.153317.
  82. Vimal K. Maurya; Swatantra Kumar; Anil K. Prasad; Madan L. B. Bhatt; Shailendra K. Saxena; Structure-based drug designing for potential antiviral activity of selected natural products from Ayurveda against SARS-CoV-2 spike glycoprotein and its cellular receptor. VirusDisease 2020, 31, 179-193, 10.1007/s13337-020-00598-8.
  83. Jiao Wang; Xiaoli Zhang; Alejandra B. Omarini; Binglin Li; Virtual screening for functional foods against the main protease of SARS‐CoV‐2. Journal of Food Biochemistry 2020, 44, 13481, 10.1111/jfbc.13481.
  84. Debmalya Barh; Sandeep Tiwari; Marianna E. Weener; Vasco Azevedo; Aristóteles Góes-Neto; M. Michael Gromiha; Preetam Ghosh; Multi-omics-based identification of SARS-CoV-2 infection biology and candidate drugs against COVID-19. Computers in Biology and Medicine 2020, 126, 104051-104051, 10.1016/j.compbiomed.2020.104051.
  85. Dantong Zheng; Chongxing Huang; Haohe Huang; Yuan Zhao; Muhammad Rafi Ullah Khan; Hui Zhao; Lijie Huang; Antibacterial Mechanism of Curcumin: A Review. Chemistry & Biodiversity 2020, 17, 2000171, 10.1002/cbdv.202000171.
  86. Sin-Yeang Teow; Kitson Liew; Syed A. Ali; Alan Soo-Beng Khoo; Suat-Cheng Peh; Antibacterial Action of Curcumin against Staphylococcus aureus: A Brief Review. Journal of Tropical Medicine 2016, 2016, 1-10, 10.1155/2016/2853045.
  87. Carolina Santezi; Bárbara Donadon Reina; Lívia Nordi Dovigo; Curcumin-mediated Photodynamic Therapy for the treatment of oral infections—A review. Photodiagnosis and Photodynamic Therapy 2018, 21, 409-415, 10.1016/j.pdpdt.2018.01.016.
  88. Fernanda Alves; Ana Cláudia Pavarina; Ewerton Garcia De Oliveira Mima; Anthony P McHale; John Francis Callan; Antimicrobial sonodynamic and photodynamic therapies against Candida albicans. Biofouling 2018, 34, 357-367, 10.1080/08927014.2018.1439935.
  89. Fatemeh Forouzanfar; Ali Forouzanfar; Thozhukat Sathyapalan; Hossein M. Orafai; Amirhossein Sahebkar; Curcumin for the Management of Periodontal Diseases: A Review. Current Pharmaceutical Design 2020, 26, 4277-4284, 10.2174/1381612826666200513112607.
  90. Kourosh Cheraghipour; Behrouz Ezatpour; Leila Masoori; Abdolrazagh Marzban; Asghar Sepahvand; Arian Karimi Rouzbahani; Abbas Moridnia; Sayyad Khanizadeh; Hossein Mahmoudvand; Anti-Candida Activity of Curcumin: A Systematic Review. Current Drug Discovery Technologies 2021, 18, 379-390, 10.2174/1570163817666200518074629.
  91. Kazunori Kataoka; Atsushi Harada; Yukio Nagasaki; Block copolymer micelles for drug delivery: design, characterization and biological significance. Advanced Drug Delivery Reviews 2001, 47, 113-131, 10.1016/s0169-409x(00)00124-1.
  92. Muhammad Usman Akbar; Khalid Mahmood Zia; Ahsan Nazir; Jamshed Iqbal; Syeda Abida Ejaz; Muhammad Sajid Hamid Akash; Pluronic-Based Mixed Polymeric Micelles Enhance the Therapeutic Potential of Curcumin. AAPS PharmSciTech 2018, 19, 2719-2739, 10.1208/s12249-018-1098-9.
  93. Fan Huang; Yang Gao; Yumin Zhang; Tangjian Cheng; Hanlin Ou; Lijun Yang; Jinjian Liu; Linqi Shi; Jianfeng Liu; Silver-Decorated Polymeric Micelles Combined with Curcumin for Enhanced Antibacterial Activity. ACS Applied Materials & Interfaces 2017, 9, 16880-16889, 10.1021/acsami.7b03347.
  94. Saeid Rahbar Takrami; Najmeh Ranji; Majid Sadeghizadeh; Antibacterial effects of curcumin encapsulated in nanoparticles on clinical isolates of Pseudomonas aeruginosa through downregulation of efflux pumps. Molecular Biology Reports 2019, 46, 2395-2404, 10.1007/s11033-019-04700-2.
  95. Fangfang Teng; Peizong Deng; Zhimei Song; Feilong Zhou; Runliang Feng; Enhanced effect in combination of curcumin- and ketoconazole-loaded methoxy poly (ethylene glycol)-poly (ε-caprolactone) micelles. Biomedicine & Pharmacotherapy 2017, 88, 43-51, 10.1016/j.biopha.2017.01.033.
  96. Maryam Pourhajibaghera; BahmanRahimi esboeibc; Mahshid Hodjata; Abbas Bahadord; [Email Protected]; Sonodynamic excitation of nanomicelle curcumin for eradication of Streptococcus mutans under sonodynamic antimicrobial chemotherapy: Enhanced anti-caries activity of nanomicelle curcumin. Photodiagnosis and Photodynamic Therapy 2020, 30, 101780, 10.1016/j.pdpdt.2020.101780.
  97. Katia Rupel; Luisa Zupin; Silvia Brich; Mario Mardirossian; Giulia Ottaviani; Margherita Gobbo; Roberto Di Lenarda; Sabrina Pricl; Sergio Crovella; Serena Zacchigna; et al.Matteo Biasotto Antimicrobial activity of amphiphilic nanomicelles loaded with curcumin against Pseudomonas aeruginosa alone and activated by blue laser light. Journal of Biophotonics 2020, 14, null, 10.1002/jbio.202000350.
  98. Victor Hugo Cortez Dias; Amanda Milene Malacrida; Adriele Rodrigues dos Santos; Andreia Farias Pereira Batista; Paula Aline Zanetti Campanerut-Sá; Gustavo Braga; Evandro Bona; Wilker Caetano; Jane Martha Graton Mikcha; pH interferes in photoinhibitory activity of curcumin nanoencapsulated with pluronic® P123 against Staphylococcus aureus. Photodiagnosis and Photodynamic Therapy 2021, 33, 102085, 10.1016/j.pdpdt.2020.102085.
  99. Yanhui Zhu; Qiaojie Luo; Hongjie Zhang; Qiuquan Cai; Xiaodong Li; Zhiquan Shen; Weipu Zhu; A shear-thinning electrostatic hydrogel with antibacterial activity by nanoengineering of polyelectrolytes. Biomaterials Science 2019, 8, 1394-1404, 10.1039/c9bm01386e.
  100. Caio H N Barros; Dishon W Hiebner; Stephanie Fulaz; Stefania Vitale; Laura Quinn; Eoin Casey; Synthesis and self-assembly of curcumin-modified amphiphilic polymeric micelles with antibacterial activity.. Journal of Nanobiotechnology 2021, 19, 104.
  101. N.P. Aditya; Geetanjali Chimote; Karthigayan Gunalan; Rinti Banerjee; Swati Patankar; Basavaraj Madhusudhan; Curcuminoids-loaded liposomes in combination with arteether protects against Plasmodium berghei infection in mice. Experimental Parasitology 2012, 131, 292-299, 10.1016/j.exppara.2012.04.010.
  102. Soumitra Shome; Anupam Das Talukdar; Manabendra Dutta Choudhury; Mrinal Kanti Bhattacharya; Hrishikesh Upadhyaya; Curcumin as potential therapeutic natural product: a nanobiotechnological perspective. Journal of Pharmacy and Pharmacology 2016, 68, 1481-1500, 10.1111/jphp.12611.
  103. Yan Chen; Yao Lu; Robert J Lee; Guangya Xiang; Nano Encapsulated Curcumin: And Its Potential for Biomedical Applications. International Journal of Nanomedicine 2020, ume 15, 3099-3120, 10.2147/ijn.s210320.
  104. Ting Ding; Tingting Li; Zhi Wang; Jianrong Li; Curcumin liposomes interfere with quorum sensing system of Aeromonas sobria and in silico analysis. Scientific Reports 2017, 7, 1-16, 10.1038/s41598-017-08986-9.
  105. Anuj Mittal; Naveen Kumar; Nar Singh Chauhan; Curcumin Encapsulated PEGylated Nanoliposomes: A Potential Anti-Infective Therapeutic Agent. Indian Journal of Microbiology 2019, 59, 336-343, 10.1007/s12088-019-00811-3.
  106. Sara Battista; Maria Anna Maggi; Pierangelo Bellio; Luciano Galantini; Angelo Antonio D’Archivio; Giuseppe Celenza; Roberta Colaiezzi; Luisa Giansanti; Curcuminoids-loaded liposomes: influence of lipid composition on their physicochemical properties and efficacy as delivery systems. Colloids and Surfaces A: Physicochemical and Engineering Aspects 2020, 597, 124759, 10.1016/j.colsurfa.2020.124759.
  107. Mengqian Gao; Xue Long; Jing Du; Mengting Teng; Weichen Zhang; Yuting Wang; Xingqi Wang; Ziyuan Wang; Peng Zhang; Jun Li; et al. Enhanced curcumin solubility and antibacterial activity by encapsulation in PLGA oily core nanocapsules. Food & Function 2019, 11, 448-455, 10.1039/c9fo00901a.
  108. Eshant Bhatia; Shivam Sharma; Kiran Jadhav; Rinti Banerjee; Combinatorial liposomes of berberine and curcumin inhibit biofilm formation and intracellular methicillin resistant Staphylococcus aureus infections and associated inflammation. Journal of Materials Chemistry B 2020, 9, 864-875, 10.1039/d0tb02036b.
  109. Heba A. Hazzah; Ragwa M. Farid; Maha M.A. Nasra; Walaa A. Hazzah; Magda A. El-Massik; Ossama Y. Abdallah; Gelucire-Based Nanoparticles for Curcumin Targeting to Oral Mucosa: Preparation, Characterization, and Antimicrobial Activity Assessment. Journal of Pharmaceutical Sciences 2015, 104, 3913-3924, 10.1002/jps.24590.
  110. Min Sun; Xun Su; Buyun Ding; Xiuli He; Xiuju Liu; Aihua Yu; Hongxiang Lou; Guangxi Zhai; Advances in nanotechnology-based delivery systems for curcumin. Nanomedicine 2012, 7, 1085-1100, 10.2217/nnm.12.80.
  111. Ankur Gupta; Huseyin Burak Eral; Trevor Alan Hatton; Patrick S. Doyle; Nanoemulsions: formation, properties and applications. Soft Matter 2016, 12, 2826-2841, 10.1039/c5sm02958a.
  112. Gagan Flora; Deepesh Gupta; Archana Tiwari; Nanocurcumin: A Promising Therapeutic Advancement over Native Curcumin. Critical Reviews in Therapeutic Drug Carrier Systems 2012, 30, 331-368, 10.1615/critrevtherdrugcarriersyst.2013007236.
  113. Amit Mirani; Harish Kundaikar; Shilpa Velhal; Vainav Patel; Atmaram Bandivdekar; Mariam Degani; Vandana Patravale; Tetrahydrocurcumin-loaded vaginal nanomicrobicide for prophylaxis of HIV/AIDS: in silico study, formulation development, and in vitro evaluation. Drug Delivery and Translational Research 2019, 9, 828-847, 10.1007/s13346-019-00633-2.
  114. Caroline Measso Do Bonfim; Letícia Figueiredo Monteleoni; Marília De Freitas Calmon; Natália Maria Cândido; Paola Jocelan Scarin Provazzi; Vanesca De Souza Lino; Tatiana Rabachini; Laura Sichero; Luisa Lina Villa; Silvana Maria Quintana; et al.Patrícia Pereira Dos Santos MelliFernando Lucas PrimoCamila AmantinoAntonio TedescoEnrique BoccardoPaula Rahal Antiviral activity of curcumin-nanoemulsion associated with photodynamic therapy in vulvar cell lines transducing different variants of HPV-16. Artificial Cells, Nanomedicine, and Biotechnology 2019, 48, 515-524, 10.1080/21691401.2020.1725023.
  115. Najwa Nabila; Nadia Khansa Suada; Dionisius Denis; Benediktus Yohan; Annis Catur Adi; Anna Surgean Veterini; Atsarina Larasati Anindya; R. Tedjo Sasmono; Heni Rachmawati; Antiviral Action of Curcumin Encapsulated in Nanoemulsion against Four Serotypes of Dengue Virus. Pharmaceutical Nanotechnology 2020, 8, 54-62, 10.2174/2211738507666191210163408.
  116. Atinderpal Kaur; Yashaswee Saxena; Rakhi Bansal; Sonal Gupta; Amit Tyagi; Rakesh Kumar Sharma; Javed Ali; Amulya Kumar Panda; Reema Gabrani; Shweta Dang; et al. Intravaginal Delivery of Polyphenon 60 and Curcumin Nanoemulsion Gel.. AAPS PharmSciTech 2017, 18, 2188-2202, 10.1208/s12249-016-0652-6.
  117. Fahanwi Asabuwa Ngwabebhoh; Sevinc Ilkar Erdagi; Ufuk Yildiz; Pickering emulsions stabilized nanocellulosic-based nanoparticles for coumarin and curcumin nanoencapsulations: In vitro release, anticancer and antimicrobial activities. Carbohydrate Polymers 2018, 201, 317-328, 10.1016/j.carbpol.2018.08.079.
  118. Momin Khan, Muhammad Ali, Walayat Shah, Akram Shah & Muhammad Masoom Yasinzai; Curcumin-loaded self-emulsifying drug delivery system (cu-SEDDS): A promising approach for the control of primary pathogen and secondary bacterial infections in cutaneous leishmaniasis. Applied Microbiology and Biotechnology volume 2019, 103, 7481.
  119. Ayat F. Hashim; Said Hamed; Hoda A. Abdel Hamid; Kamel A. Abd-Elsalam; Iwona Golonka; Witold Musiał; Ibrahim M. El-Sherbiny; Antioxidant and antibacterial activities of omega-3 rich oils/curcumin nanoemulsions loaded in chitosan and alginate-based microbeads. International Journal of Biological Macromolecules 2019, 140, 682-696, 10.1016/j.ijbiomac.2019.08.085.
  120. Kai Chen; Yong Qian; Senyi Wu; Xueqing Qiu; Dongjie Yang; Lei Lei; Neutral fabrication of UV-blocking and antioxidation lignin-stabilized high internal phase emulsion encapsulates for high efficient antibacterium of natural curcumin. Food & Function 2019, 10, 3543-3555, 10.1039/c9fo00320g.
  121. Agnieszka Lewińska; Anna Jaromin; Julia Jezierska; Role of architecture of N-oxide surfactants in the design of nanoemulsions for Candida skin infection. Colloids and Surfaces B: Biointerfaces 2020, 187, 110639, 10.1016/j.colsurfb.2019.110639.
  122. Jaya Lakkakula; Rui Werner Maçedo Krause; A vision for cyclodextrin nanoparticles in drug delivery systems and pharmaceutical applications. Nanomedicine 2014, 9, 877-894, 10.2217/nnm.14.41.
  123. Fliur Macaev; Veaceslav Boldescu; Athina Geronikaki; Natalia Sucman; Recent Advances in the Use of Cyclodextrins in Antifungal Formulations. Current Topics in Medicinal Chemistry 2013, 13, 2677-2683, 10.2174/15680266113136660194.
  124. Takuro Kurita; Yuji Makino; Novel curcumin oral delivery systems.. Anticancer Research 2013, 33, 2807–2821.
  125. E.M.Martin Del Valle; Cyclodextrins and their uses: a review. Process Biochemistry 2004, 39, 1033-1046, 10.1016/s0032-9592(03)00258-9.
  126. Atwood Jerry L.. Comprehensive Supramolecular Chemistry; Gokel George, Eds.; .: New York, USA, 1996; pp. 1.
  127. Phennapha Saokham; Chutimon Muankaew; Phatsawee Jansook; Thorsteinn Loftsson; Solubility of Cyclodextrins and Drug/Cyclodextrin Complexes. Molecules 2018, 23, 1161, 10.3390/molecules23051161.
  128. Mark E. Davis; Marcus E. Brewster; Cyclodextrin-based pharmaceutics: past, present and future. Nature Reviews Drug Discovery 2004, 3, 1023-1035, 10.1038/nrd1576.
  129. Aaron J. Smith; John Oertle; Dino Prato; Multiple Actions of Curcumin Including Anticancer, Anti-Inflammatory, Antimicrobial and Enhancement via Cyclodextrin. Journal of Cancer Therapy 2014, 06, 257-272, 10.4236/jct.2015.63029.
  130. Anne Bee Hegge; Thorbjørn T. Nielsen; Kim L. Larsen; Ellen Bruzell; Hanne H. Tønnesen; Impact of Curcumin Supersaturation in Antibacterial Photodynamic Therapy—Effect of Cyclodextrin Type and Amount: Studies on Curcumin and Curcuminoides XLV. Journal of Pharmaceutical Sciences 2012, 101, 1524-1537, 10.1002/jps.23046.
  131. Anne Bee Hegge; M. Vukicevic; E. Bruzell; S. Kristensen; H.H. Tønnesen; Solid dispersions for preparation of phototoxic supersaturated solutions for antimicrobial photodynamic therapy (aPDT). European Journal of Pharmaceutics and Biopharmaceutics 2012, 83, 95-105, 10.1016/j.ejpb.2012.09.011.
  132. Kristine Wikene; Anne Bee Hegge; Ellen Bruzell; Hanne Hjorth Tønnesen; Formulation and characterization of lyophilized curcumin solid dispersions for antimicrobial photodynamic therapy (aPDT): studies on curcumin and curcuminoids LII. Drug Development and Industrial Pharmacy 2014, 41, 969-977, 10.3109/03639045.2014.919315.
  133. Ilya Shlar; Samir Droby; Victor Rodov; Antimicrobial coatings on polyethylene terephthalate based on curcumin/cyclodextrin complex embedded in a multilayer polyelectrolyte architecture. Colloids and Surfaces B: Biointerfaces 2018, 164, 379-387, 10.1016/j.colsurfb.2018.02.008.
  134. A. Gupta; D.J. Keddie; V. Kannappan; H. Gibson; I.R. Khalil; M. Kowalczuk; C. Martin; X. Shuai; I. Radecka; Production and characterisation of bacterial cellulose hydrogels loaded with curcumin encapsulated in cyclodextrins as wound dressings. European Polymer Journal 2019, 118, 437-450, 10.1016/j.eurpolymj.2019.06.018.
  135. Shao-Pin Wang And Fu-Yung Huang Desu Naveen Kumar Reddy; Ramya Kumar; Shao-Pin Wang; Fu-Yung Huang; Curcumin-C3 Complexed with α-, β-cyclodextrin Exhibits Antibacterial and Antioxidant Properties Suitable for Cancer Treatments. Current Drug Metabolism 2020, 20, 988-1001, 10.2174/1389200220666191001104834.
  136. Nina Alizadeh; Shokufeh Malakzadeh; Antioxidant, antibacterial and anti-cancer activities of β-and γ-CDs/curcumin loaded in chitosan nanoparticles. International Journal of Biological Macromolecules 2020, 147, 778-791, 10.1016/j.ijbiomac.2020.01.206.
  137. Jochen Brasch; Vera Beck‐Jendroschek; Grit Walther; Darian Rubbel; Clinical isolates of Trichophyton rubrum are completely inhibited by photochemical treatment with a γ‐cyclodextrin formulation of curcuminoids. Mycoses 2020, 63, 369-375, 10.1111/myc.13051.
  138. Anna Stasiłowicz; Ewa Tykarska; Kornelia Lewandowska; Maciej Kozak; Andrzej Miklaszewski; Joanna Kobus-Cisowska; Daria Szymanowska; Tomasz Plech; Jacek Jenczyk; Judyta Cielecka-Piontek; et al. Hydroxypropyl-β-cyclodextrin as an effective carrier of curcumin – piperine nutraceutical system with improved enzyme inhibition properties. Journal of Enzyme Inhibition and Medicinal Chemistry 2019, 35, 1811-1821, 10.1080/14756366.2020.1801670.
  139. Ilya Shlar; Samir Droby; Ruplal Choudhary; Victor Rodov; The mode of antimicrobial action of curcumin depends on the delivery system: monolithic nanoparticles vs. supramolecular inclusion complex. RSC Advances 2017, 7, 42559-42569, 10.1039/c7ra07303h.
  140. Mahsa Saheb; Narges Fereydouni; Saeideh Nemati; George E. Barreto; Thomas P. Johnston; Amirhossein Sahebkar; Chitosan‐based delivery systems for curcumin: A review of pharmacodynamic and pharmacokinetic aspects. Journal of Cellular Physiology 2019, 234, 12325-12340, 10.1002/jcp.28024.
  141. Michelly Pellá; Michele K. Lima-Tenório; Ernandes Tenório-Neto; Marcos Rogério Guilherme; Edvani C. Muniz; Adley F. Rubira; Chitosan-based hydrogels: From preparation to biomedical applications. Carbohydrate Polymers 2018, 196, 233-245, 10.1016/j.carbpol.2018.05.033.
  142. Marguerite Rinaudo; Chitin and chitosan: Properties and applications. Progress in Polymer Science 2006, 31, 603-632, 10.1016/j.progpolymsci.2006.06.001.
  143. Ying-Chien Chung; Ya-Ping Su; Chiing-Chang Chen; Guang Jia; Huey-Lan Wang; J C Gaston Wu; Jaung-Geng Lin; Relationship between antibacterial activity of chitosan and surface characteristics of cell wall.. Acta Pharmacologica Sinica 2004, 25, 932–936.
  144. Aimee E. Krausz; Brandon L. Adler; Vitor Cabral; Mahantesh Navati; Jessica Doerner; Rabab Charafeddine; Dinesh Chandra; Hongying Liang; Leslie Gunther; Alicea Clendaniel; et al.Stacey HarperJoel M. FriedmanJoshua D. NosanchukAdam J. Friedman Curcumin-encapsulated nanoparticles as innovative antimicrobial and wound healing agent. Nanomedicine: Nanotechnology, Biology and Medicine 2014, 11, 195-206, 10.1016/j.nano.2014.09.004.
  145. T.S. Saranya; V.K. Rajan; Raja Biswas; R. Jayakumar; S. Sathianarayanan; Synthesis, characterisation and biomedical applications of curcumin conjugated chitosan microspheres. International Journal of Biological Macromolecules 2018, 110, 227-233, 10.1016/j.ijbiomac.2017.12.044.
  146. Amir Maghsoudi; Fatemeh Yazdian; Saleheh Shahmoradi; Leila Ghaderi; Mehran Hemati; Ghassem Amoabediny; Curcumin-loaded polysaccharide nanoparticles: Optimization and anticariogenic activity against Streptococcus mutans. Materials Science and Engineering: C 2017, 75, 1259-1267, 10.1016/j.msec.2017.03.032.
  147. Shabnam Jahanizadeh; Fatemeh Yazdian; Azam Marjani; Meisam Omidi; Hamid Rashedi; Curcumin-loaded chitosan/carboxymethyl starch/montmorillonite bio-nanocomposite for reduction of dental bacterial biofilm formation. International Journal of Biological Macromolecules 2017, 105, 757-763, 10.1016/j.ijbiomac.2017.07.101.
  148. Yong Zhao; Jia‑Guo Liu; Wei‑Min Chen; Ai‑Xi Yu; Efficacy of thermosensitive chitosan/β‑glycerophosphate hydrogel loaded with β‑cyclodextrin‑curcumin for the treatment of cutaneous wound infection in rats. Experimental and Therapeutic Medicine 2017, 15, 1304-1313, 10.3892/etm.2017.5552.
  149. Niranjan R.; Kaushik M.; Prakash J.; Venkataprasanna K.S.; Christy Arpana; Pannerselvam Balashanmugam; G. Devanand Venkatasubbu; Enhanced wound healing by PVA/Chitosan/Curcumin patches: In vitro and in vivo study. Colloids and Surfaces B: Biointerfaces 2019, 182, 110339, 10.1016/j.colsurfb.2019.06.068.
  150. Mazhar Abbas; Tariq Hussain; Muhammad Arshad; Abdur Rahman Ansari; Asma Irshad; Jan Nisar; Fida Hussain; Nasir Masood; Arif Nazir; Munawar Iqbal; et al. Wound healing potential of curcumin cross-linked chitosan/polyvinyl alcohol. International Journal of Biological Macromolecules 2019, 140, 871-876, 10.1016/j.ijbiomac.2019.08.153.
  151. Su Ma; Doris Moser; Feng Han; Matthias Leonhard; Berit Schneider-Stickler; Yulong Tan; Preparation and antibiofilm studies of curcumin loaded chitosan nanoparticles against polymicrobial biofilms of Candida albicans and Staphylococcus aureus. Carbohydrate Polymers 2020, 241, 116254, 10.1016/j.carbpol.2020.116254.
  152. Samah A Loutfy; Mostafa H Elberry; Khaled Yehia Farroh; Hossam Taha Mohamed; Aya A Mohamed; ElChaimaa B Mohamed; Ahmed Hassan Ibrahim Faraag; Shaker A Mousa; Antiviral Activity of Chitosan Nanoparticles Encapsulating Curcumin Against Hepatitis C Virus Genotype 4a in Human Hepatoma Cell Lines. International Journal of Nanomedicine 2020, ume 15, 2699-2715, 10.2147/IJN.S241702.
  153. Soad Hassan Taha; Ibrahim Mohamed El-Sherbiny; Aida Soliman Salem; Mahmoud Abdel-Hamid; Ali Hussein Hamed; Ghada Abady Ahmed; Antiviral Activity of Curcumin Loaded Milk Proteins Nanoparticles on Potato Virus Y. Pakistan Journal of Biological Sciences 2019, 22, 614-622, 10.3923/pjbs.2019.614.622.
  154. Anne Bee Hegge; T. Andersen; J.E. Melvik; E. Bruzell; S. Kristensen; H.H. Tønnesen; Formulation and Bacterial Phototoxicity of Curcumin Loaded Alginate Foams for Wound Treatment Applications: Studies on Curcumin and Curcuminoides XLII. Journal of Pharmaceutical Sciences 2010, 100, 174-185, 10.1002/jps.22263.
  155. Bhawana; Rupesh Kumar Basniwal; Harpreet Singh Buttar; V. K. Jain; Nidhi Jain; Curcumin Nanoparticles: Preparation, Characterization, and Antimicrobial Study. Journal of Agricultural and Food Chemistry 2011, 59, 2056-2061, 10.1021/jf104402t.
  156. Mo’Ath Ahmad Adahoun; M-Ali Al-Akhras; Mohamad Suhaimi Jaafar; Mohamed Bououdina; Enhanced anti-cancer and antimicrobial activities of curcumin nanoparticles. Artificial Cells, Nanomedicine, and Biotechnology 2016, 45, 98-107, 10.3109/21691401.2015.1129628.
  157. Ilya Shlar; Elena Poverenov; Yakov Vinokur; Batia Horev; Samir Droby; Victor Rodov; High-Throughput Screening of Nanoparticle-Stabilizing Ligands: Application to Preparing Antimicrobial Curcumin Nanoparticles by Antisolvent Precipitation. Nano-Micro Letters 2014, 7, 68-79, 10.1007/s40820-014-0020-6.
  158. Da Som No; Ammar AlGburi; Phong Huynh; Aubry Moret; Marion Ringard; Nicole Comito; Djamel Drider; Paul Takhistov; Michael L. Chikindas; Antimicrobial efficacy of curcumin nanoparticles against Listeria monocytogenes is mediated by surface charge. Journal of Food Safety 2017, 37, e12353, 10.1111/jfs.12353.
  159. Jeffersson Krishan Trigo Gutierrez; Gabriela Cristina Zanatta; Ana Laura Mira Ortega; Maria Isabella Cuba Balastegui; Paula Volpato Sanitá; Ana Cláudia Pavarina; Paula Barbugli; Ewerton Garcia De Oliveira Mima; Encapsulation of curcumin in polymeric nanoparticles for antimicrobial Photodynamic Therapy. PLOS ONE 2017, 12, e0187418, 10.1371/journal.pone.0187418.
  160. Vinicius Tatsuyuji Sakima; Paula Aboud Barbugli; Paulo Sérgio Cerri; Marlus Chorilli; Juliana Cabrini Carmello; Ana Cláudia Pavarina; Ewerton Garcia De Oliveira Mima; Antimicrobial Photodynamic Therapy Mediated by Curcumin-Loaded Polymeric Nanoparticles in a Murine Model of Oral Candidiasis. Molecules 2018, 23, 2075, 10.3390/molecules23082075.
  161. Aref Shariati; Elham Asadian; Fatemeh Fallah; Taher Azimi; Ali Hashemi; Javad Yasbolaghi Sharahi; Majid Taati Moghadam; Evaluation of Nano-curcumin effects on expression levels of virulence genes and biofilm production of multidrug-resistant Pseudomonas aeruginosa isolated from burn wound infection in Tehran, Iran. Infection and Drug Resistance 2019, ume 12, 2223-2235, 10.2147/idr.s213200.
  162. Eduard Preis; Elias Baghdan; Michael R. Agel; Thomas Anders; Marcel Pourasghar; Marc Schneider; Udo Bakowsky; Spray dried curcumin loaded nanoparticles for antimicrobial photodynamic therapy. European Journal of Pharmaceutics and Biopharmaceutics 2019, 142, 531-539, 10.1016/j.ejpb.2019.07.023.
  163. Antonise M. Jaguezeski; Carine F. Souza; Gessica Perin; João H. Reis; Teane M.A. Gomes; Matheus Dellaméa Baldissera; Rodrigo A. Vaucher; Cinthia M. de Andrade; Lenita M. Stefani; Samanta Gündel; et al.Aline OuriqueAleksandro S. Da Silva Effect of free and nano-encapsulated curcumin on treatment and energetic metabolism of gerbils infected by Listeria monocytogenes. Microbial Pathogenesis 2019, 134, 103564, 10.1016/j.micpath.2019.103564.
  164. Maryam Pourhajibagher; Ladan Ranjbar Omrani; Mohammad Noroozian; Zahra Ghorbanzadeh; Abbas Bahador; In vitro antibacterial activity and durability of a nano-curcumin-containing pulp capping agent combined with antimicrobial photodynamic therapy. Photodiagnosis and Photodynamic Therapy 2021, 33, 102150, 10.1016/j.pdpdt.2020.102150.
  165. Maryam Pourhajibagher; Gianluca Plotino; Nasim Chiniforush; Abbas Bahador; Dual wavelength irradiation antimicrobial photodynamic therapy using indocyanine green and metformin doped with nano-curcumin as an efficient adjunctive endodontic treatment modality. Photodiagnosis and Photodynamic Therapy 2020, 29, 101628, 10.1016/j.pdpdt.2019.101628.
  166. Liwei Chen; Ziyue Song; Xiujuan Zhi; Bin Du; Photoinduced Antimicrobial Activity of Curcumin-Containing Coatings: Molecular Interaction, Stability and Potential Application in Food Decontamination. ACS Omega 2020, 5, 31044-31054, 10.1021/acsomega.0c04065.
  167. Hanie Ahmadi; Vahid Haddadi-Asl; Hassan-Ali Ghafari; Roghayeh Ghorbanzadeh; Yasaman Mazlum; Abbas Bahador; Shear bond strength, adhesive remnant index, and anti-biofilm effects of a photoexcited modified orthodontic adhesive containing curcumin doped poly lactic-co-glycolic acid nanoparticles: An ex-vivo biofilm model of S. mutans on the enamel slab bonded brackets. Photodiagnosis and Photodynamic Therapy 2020, 30, 101674, 10.1016/j.pdpdt.2020.101674.
  168. Kitipong Kiti; Orawan Suwantong; The potential use of curcumin-β-cyclodextrin inclusion complex/chitosan-loaded cellulose sponges for the treatment of chronic wound. International Journal of Biological Macromolecules 2020, 164, 3250-3258, 10.1016/j.ijbiomac.2020.08.190.
  169. Abolfazl Shakeri; Yunes Panahi; Thomas P. Johnston; Amirhossein Sahebkar; Biological properties of metal complexes of curcumin. BioFactors 2019, 45, 304-317, 10.1002/biof.1504.
  170. S.K. Bajpai; Sonam Ahuja; N. Chand; M. Bajpai; Nano cellulose dispersed chitosan film with Ag NPs/Curcumin: An in vivo study on Albino Rats for wound dressing. International Journal of Biological Macromolecules 2017, 104, 1012-1019, 10.1016/j.ijbiomac.2017.06.096.
  171. Camila Fabiano de Freitas; Elza Kimura; Adley Forti Rubira; Edvani Curti Muniz; Curcumin and silver nanoparticles carried out from polysaccharide-based hydrogels improved the photodynamic properties of curcumin through metal-enhanced singlet oxygen effect. Materials Science and Engineering: C 2020, 112, 110853, 10.1016/j.msec.2020.110853.
  172. Nelson Durán; Marcela Durán; Marcelo Bispo de Jesus; Amedea B. Seabra; Wagner Fávaro; Gerson Nakazato; Silver nanoparticles: A new view on mechanistic aspects on antimicrobial activity. Nanomedicine: Nanotechnology, Biology and Medicine 2016, 12, 789-799, 10.1016/j.nano.2015.11.016.
  173. Swati Jaiswal; Prashant Mishra; Antimicrobial and antibiofilm activity of curcumin-silver nanoparticles with improved stability and selective toxicity to bacteria over mammalian cells. Medical Microbiology and Immunology 2017, 207, 39-53, 10.1007/s00430-017-0525-y.
  174. Elsy El Khoury; Mohamad Abiad; Zeina G. Kassaify; Digambara Patra; Green synthesis of curcumin conjugated nanosilver for the applications in nucleic acid sensing and anti-bacterial activity. Colloids and Surfaces B: Biointerfaces 2015, 127, 274-280, 10.1016/j.colsurfb.2015.01.050.
  175. Judy Ching Yee Loo; Ramin Rohanizadeh; Paul Young; Daniela Traini; Rosalia Cavaliere; Cynthia Whitchurch; Wing Hin Lee; Combination of Silver Nanoparticles and Curcumin Nanoparticles for Enhanced Anti-biofilm Activities. Journal of Agricultural and Food Chemistry 2015, 64, 2513-2522, 10.1021/acs.jafc.5b04559.
  176. Ahmed M Abdellah; Mahmoud A Sliem; Mona Bakr‡; Rehab M Amin; Green synthesis and biological activity of silver–curcumin nanoconjugates. Future Medicinal Chemistry 2018, 10, 2577-2588, 10.4155/fmc-2018-0152.
  177. Zhiyong Song; Yang Wu; Huajuan Wang; Heyou Han; Synergistic antibacterial effects of curcumin modified silver nanoparticles through ROS-mediated pathways. Materials Science and Engineering: C 2019, 99, 255-263, 10.1016/j.msec.2018.12.053.
  178. Payal Srivastava; Manjulika Shukla; Grace Kaul; Sidharth Chopra; Ashis K. Patra; Rationally designed curcumin based ruthenium(ii) antimicrobials effective against drug-resistant Staphylococcus aureus. Dalton Transactions 2019, 48, 11822-11828, 10.1039/c9dt01650c.
  179. K. Varaprasad; K. Vimala; S. Ravindra; N. Narayana Reddy; G. Venkata Subba Reddy; K. Mohana Raju; Fabrication of silver nanocomposite films impregnated with curcumin for superior antibacterial applications. Journal of Materials Science: Materials in Medicine 2011, 22, 1863-1872, 10.1007/s10856-011-4369-5.
  180. Kunnavakkam Vinjimur Srivatsan; N. Duraipandy; Shajitha Begum; Rachita Lakra; Usha Ramamurthy; Purna Sai Korrapati; Manikantan Syamala Kiran; Effect of curcumin caged silver nanoparticle on collagen stabilization for biomedical applications. International Journal of Biological Macromolecules 2015, 75, 306-315, 10.1016/j.ijbiomac.2015.01.050.
  181. Sadiya Anjum; Amlan Gupta; Deepika Sharma; Deepti Gautam; Surya Bhan; Anupama Sharma; Arti Kapil; Bhuvanesh Gupta; Development of novel wound care systems based on nanosilver nanohydrogels of polymethacrylic acid with Aloe vera and curcumin. Materials Science and Engineering: C 2016, 64, 157-166, 10.1016/j.msec.2016.03.069.
  182. K. R. Soumya; S. Snigdha; Sheela Sugathan; Jyothis Mathew; E. K. Radhakrishnan; Zinc oxide–curcumin nanocomposite loaded collagen membrane as an effective material against methicillin-resistant coagulase-negative Staphylococci. 3 Biotech 2017, 7, 238, 10.1007/s13205-017-0861-z.
  183. Thais Alves; Marco Chaud; Denise Grotto; Angela Faustino Jozala; Raksha Pandit; Mahendra Rai; Carolina Alves Dos Santos; Association of Silver Nanoparticles and Curcumin Solid Dispersion: Antimicrobial and Antioxidant Properties. AAPS PharmSciTech 2017, 19, 225-231, 10.1208/s12249-017-0832-z.
  184. Sony Paul; Kalyani Mohanram; Iyanar Kannan; Antifungal activity of curcumin-silver nanoparticles against fluconazole-resistant clinical isolates of Candida species. AYU (An international quarterly journal of research in Ayurveda) 2017, 39, 182-186, 10.4103/ayu.ayu_24_18.
  185. Ly Loan Khanh; Nguyen Thanh Truc; Nguyen Tan Dat; Nguyen Thi Phuong Nghi; Vo van Toi; Nguyen Thi Thu Hoai; Tran Ngoc Quyen; Tran Thi Thanh Loan; Nguyen Thi Hiep; Gelatin-stabilized composites of silver nanoparticles and curcumin: characterization, antibacterial and antioxidant study. Science and Technology of Advanced Materials 2019, 20, 276-290, 10.1080/14686996.2019.1585131.
  186. B. Anagha; Dhanya George; P. Uma Maheswari; K. M. Meera Sheriffa Begum; Biomass Derived Antimicrobial Hybrid Cellulose Hydrogel with Green ZnO Nanoparticles for Curcumin Delivery and its Kinetic Modelling. Journal of Polymers and the Environment 2019, 27, 2054-2067, 10.1007/s10924-019-01495-y.
  187. Dhanya George; Palanisamy Uma Maheswari; Khadar Mohamed Meera Sheriffa Begum; Gangasalam Arthanareeswaran; G Arthanaresswarn; Biomass-Derived Dialdehyde Cellulose Cross-linked Chitosan-Based Nanocomposite Hydrogel with Phytosynthesized Zinc Oxide Nanoparticles for Enhanced Curcumin Delivery and Bioactivity. Journal of Agricultural and Food Chemistry 2019, 67, 10880-10890, 10.1021/acs.jafc.9b01933.
  188. Shabnam Farkhonde Masoule; Maryam Pourhajibagher; Javad Safari; Mehdi Khoobi; Base-free green synthesis of copper(II) oxide nanoparticles using highly cross-linked poly(curcumin) nanospheres: synergistically improved antimicrobial activity. Research on Chemical Intermediates 2019, 45, 4449-4462, 10.1007/s11164-019-03841-0.
  189. Yongbo Lyu; Mengchao Yu; Qisong Liu; Qingmei Zhang; Zhanhong Liu; Ye Tian; Defu Li; Mu Changdao; Synthesis of silver nanoparticles using oxidized amylose and combination with curcumin for enhanced antibacterial activity. Carbohydrate Polymers 2020, 230, 115573, 10.1016/j.carbpol.2019.115573.
  190. Abhishek Gupta; Sophie Marie Briffa; Sam Swingler; Hazel Gibson; Vinodh Kannappan; Grazyna Adamus; Marek M. Kowalczuk; Claire Martin; Iza Radecka; Synthesis of Silver Nanoparticles Using Curcumin-Cyclodextrins Loaded into Bacterial Cellulose-Based Hydrogels for Wound Dressing Applications. Biomacromolecules 2020, 21, 1802-1811, 10.1021/acs.biomac.9b01724.
  191. Issa M. El-Nahhal; Jamil Salem; Rawan Anbar; Fawzi S. Kodeh; Abedelraouf Elmanama; Preparation and antimicrobial activity of ZnO-NPs coated cotton/starch and their functionalized ZnO-Ag/cotton and Zn(II) curcumin/cotton materials. Scientific Reports 2020, 10, 1-10, 10.1038/s41598-020-61306-6.
  192. Seyed-Behnam Ghaffari; Mohammad-Hossein Sarrafzadeh; Maryam Salami; M.Reza Khorramizadeh; A pH-sensitive delivery system based on N-succinyl chitosan-ZnO nanoparticles for improving antibacterial and anticancer activities of curcumin. International Journal of Biological Macromolecules 2020, 151, 428-440, 10.1016/j.ijbiomac.2020.02.141.
  193. Roopesh Marulasiddeshwara; M.S. Jyothi; Khantong Soontarapa; Rangappa S. Keri; Rajendran Velmurugan; Nonwoven fabric supported, chitosan membrane anchored with curcumin/TiO2 complex: Scaffolds for MRSA infected wound skin reconstruction. International Journal of Biological Macromolecules 2020, 144, 85-93, 10.1016/j.ijbiomac.2019.12.077.
  194. N. Muniyappan; M. Pandeeswaran; Augustine Amalraj; Green synthesis of gold nanoparticles using Curcuma pseudomontana isolated curcumin: Its characterization, antimicrobial, antioxidant and anti- inflammatory activities. Environmental Chemistry and Ecotoxicology 2020, 3, 117-124, 10.1016/j.enceco.2021.01.002.
  195. Mark E. Davis; Ordered porous materials for emerging applications. Nature 2002, 417, 813-821, 10.1038/nature00785.
  196. Xiao-Yu Yang; Li-Hua Chen; Yu Li; Joanna Claire Rooke; Clément Sanchez; Bao-Lian Su; Hierarchically porous materials: synthesis strategies and structure design. Chemical Society Reviews 2016, 46, 481-558, 10.1039/c6cs00829a.
  197. Alessandro Agostini; Félix Sancenón; Ramón Martínez-Máñez; María D. Marcos; Juan Soto; Pedro Amorós; A Photoactivated Molecular Gate. Chemistry – A European Journal 2012, 18, 12218-12221, 10.1002/chem.201201127.
  198. Ying Wang; Qinfu Zhao; Ning Han; Ling Bai; Jia Li; Erxi Che; Liang Hu; Qiang Zhang; Tongying Jiang; Siling Wang; et al. Mesoporous silica nanoparticles in drug delivery and biomedical applications. Nanomedicine: Nanotechnology, Biology and Medicine 2015, 11, 313-327, 10.1016/j.nano.2014.09.014.
  199. María Vallet-Regí; Francisco Balas; Daniel Arcos; Mesoporous Materials for Drug Delivery. Angewandte Chemie International Edition 2007, 46, 7548-7558, 10.1002/anie.200604488.
  200. Yaswanth Kuthati; Ranjith Kumar Kankala; Prabhakar Busa; Shi-Xiang Lin; Jin-Pei Deng; Chung-Yuan Mou; Chia-Hung Lee; Phototherapeutic spectrum expansion through synergistic effect of mesoporous silica trio-nanohybrids against antibiotic-resistant gram-negative bacterium. Journal of Photochemistry and Photobiology B: Biology 2017, 169, 124-133, 10.1016/j.jphotobiol.2017.03.003.
  201. Chunhua Wu; Yang Zhu; Tiantian Wu; Lin Wang; Yi Yuan; Jicheng Chen; Yaqin Hu; Jie Pang; Enhanced functional properties of biopolymer film incorporated with curcurmin-loaded mesoporous silica nanoparticles for food packaging. Food Chemistry 2019, 288, 139-145, 10.1016/j.foodchem.2019.03.010.
  202. Anu Sharma; Anita Yadav; Nikesh Gupta; Sandeep Sharma; Rita Kakkar; Katherine Cwiklinski; Elizabeth Quaye; Supriya D. Mahajan; Stanley A. Schwartz; Rakesh Kumar Sharma; et al. Multifunctional mesoporous curcumin encapsulated iron-phenanthroline nanocluster: A new Anti-HIV agent. Colloids and Surfaces B: Biointerfaces 2019, 180, 289-297, 10.1016/j.colsurfb.2019.04.057.
  203. Yajun Cheng; Yudi Zhang; Weijun Deng; Jing Hu; Antibacterial and anticancer activities of asymmetric lollipop-like mesoporous silica nanoparticles loaded with curcumin and gentamicin sulfate. Colloids and Surfaces B: Biointerfaces 2020, 186, 110744, 10.1016/j.colsurfb.2019.110744.
  204. Yiyan Song; Ling Cai; Zhongcheng Tian; Yuan Wu; Jin Chen; Phytochemical Curcumin-Coformulated, Silver-Decorated Melanin-like Polydopamine/Mesoporous Silica Composites with Improved Antibacterial and Chemotherapeutic Effects against Drug-Resistant Cancer Cells. ACS Omega 2020, 5, 15083-15094, 10.1021/acsomega.0c00912.
  205. Siavash Iravani; Rajender S. Varma; Green synthesis, biomedical and biotechnological applications of carbon and graphene quantum dots. A review. Environmental Chemistry Letters 2020, 18, 703-727, 10.1007/s10311-020-00984-0.
  206. Ashish Singh; Pradyot Prakash; Ranjana Singh; Nabarun Nandy; Zeba Firdaus; Monika Bansal; Ranjan K. Singh; Anchal Srivastava; Jagat K. Roy; Brahmeshwar Mishra; et al.Rakesh K. Singh Curcumin Quantum Dots Mediated Degradation of Bacterial Biofilms. Frontiers in Microbiology 2017, 8, 1517, 10.3389/fmicb.2017.01517.
  207. Chin‐Jung Lin; Lung Chang; Han‐Wei Chu; Han‐Jia Lin; Pei‐Ching Chang; Robert Y. L. Wang; Binesh Unnikrishnan; Ju‐Yi Mao; Shiow‐Yi Chen; Chih‐Ching Huang; et al. High Amplification of the Antiviral Activity of Curcumin through Transformation into Carbon Quantum Dots. Small 2019, 15, e1902641, 10.1002/smll.201902641.
  208. Ashish Kumar Singh; Himanshu Mishra; Zeba Firdaus; Shivangi Yadav; Prerana Aditi; Nabarun Nandy; Kavyanjali Sharma; Priyanka Bose; Akhilesh Kumar Pandey; Brijesh Singh Chauhan; et al.Kaushik NeogiKunwar VikramAnchal SrivastavaAmrita Ghosh KarPradyot Prakash MoS2-Modified Curcumin Nanostructures: The Novel Theranostic Hybrid Having Potent Antibacterial and Antibiofilm Activities against Multidrug-Resistant Hypervirulent Klebsiella pneumoniae. Chemical Research in Toxicology 2019, 32, 1599-1618, 10.1021/acs.chemrestox.9b00135.
  209. Maryam Pourhajibagher; Steven Parker; Nasim Chiniforush; Abbas Bahador; Photoexcitation triggering via semiconductor Graphene Quantum Dots by photochemical doping with Curcumin versus perio-pathogens mixed biofilms. Photodiagnosis and Photodynamic Therapy 2019, 28, 125-131, 10.1016/j.pdpdt.2019.08.025.
  210. Mahtab Mirzahosseinipour; Khatereh Khorsandi; Reza Hosseinzadeh; Mehrgan Ghazaeian; Fedora Khatibi Shahidi; Antimicrobial photodynamic and wound healing activity of curcumin encapsulated in silica nanoparticles. Photodiagnosis and Photodynamic Therapy 2020, 29, 101639, 10.1016/j.pdpdt.2019.101639.
  211. Gölnur Fakhrullina; Elvira Khakimova; Farida Akhatova; Giuseppe Lazzara; Filippo Parisi; Rawil F. Fakhrullin; Selective Antimicrobial Effects of Curcumin@Halloysite Nanoformulation: A Caenorhabditis elegans Study. ACS Applied Materials & Interfaces 2019, 11, 23050-23064, 10.1021/acsami.9b07499.
  212. Xue Zou; Meng Yuan; Tongyu Zhang; Hongxia Wei; Shijie Xu; Na Jiang; Nan Zheng; Zhiwei Wu; Extracellular vesicles expressing a single-chain variable fragment of an HIV-1 specific antibody selectively target Env+ tissues. Theranostics 2018, 9, 5657-5671, 10.7150/thno.33925.
  213. Julian M. Sotomil; Eliseu A. Münchow; Divya Pankajakshan; Kenneth J. Spolnik; Jessica A. Ferreira; Richard L. Gregory; Marco C. Bottino; Curcumin—A Natural Medicament for Root Canal Disinfection: Effects of Irrigation, Drug Release, and Photoactivation. Journal of Endodontics 2019, 45, 1371-1377, 10.1016/j.joen.2019.08.004.
  214. Shabnam Sattari; Abbas Dadkhah Tehrani; Mohsen Adeli; Khadijeh Soleimani; Marzieh Rashidipour; Fabrication of new generation of co-delivery systems based on graphene-g-cyclodextrin/chitosan nanofiber. International Journal of Biological Macromolecules 2020, 156, 1126-1134, 10.1016/j.ijbiomac.2019.11.144.
  215. Yu-Ning Yang; Kun-Ying Lu; Pan Wang; Yi-Cheng Ho; Min-Lang Tsai; Fwu-Long Mi; Development of bacterial cellulose/chitin multi-nanofibers based smart films containing natural active microspheres and nanoparticles formed in situ. Carbohydrate Polymers 2020, 228, 115370, 10.1016/j.carbpol.2019.115370.
  216. Elaheh Esmaeili; Tarlan Eslami-Arshaghi; Simzar Hosseinzadeh; Elnaz Elahirad; Zahra Jamalpoor; Shadie Hatamie; Masoud Soleimani; The biomedical potential of cellulose acetate/polyurethane nanofibrous mats containing reduced graphene oxide/silver nanocomposites and curcumin: Antimicrobial performance and cutaneous wound healing. International Journal of Biological Macromolecules 2020, 152, 418-427, 10.1016/j.ijbiomac.2020.02.295.
  217. Yuewei Xi; Juan Ge; Min Wang; Mi Chen; Wen Niu; Wei Cheng; Yumeng Xue; Cai Lin; Bo Lei; Bioactive Anti-inflammatory, Antibacterial, Antioxidative Silicon-Based Nanofibrous Dressing Enables Cutaneous Tumor Photothermo-Chemo Therapy and Infection-Induced Wound Healing. ACS Nano 2020, 14, 2904-2916, 10.1021/acsnano.9b07173.
  218. Ali Shababdoust; Mojgan Zandi; Morteza Ehsani; Parvin Shokrollahi; Reza Foudazi; Controlled curcumin release from nanofibers based on amphiphilic-block segmented polyurethanes. International Journal of Pharmaceutics 2020, 575, 118947, 10.1016/j.ijpharm.2019.118947.
  219. Mengmeng Zhang; Bo Zhuang; Gangjun Du; Guang Han; Yiguang Jin; Curcumin solid dispersion-loaded in situ hydrogels for local treatment of injured vaginal bacterial infection and improvement of vaginal wound healing. Journal of Pharmacy and Pharmacology 2019, 71, 1044-1054, 10.1111/jphp.13088.
  220. Swarup Roy; Jong-Whan Rhim; Preparation of antimicrobial and antioxidant gelatin/curcumin composite films for active food packaging application. Colloids and Surfaces B: Biointerfaces 2020, 188, 110761, 10.1016/j.colsurfb.2019.110761.
  221. Swarup Roy; Jong-Whan Rhim; Carboxymethyl cellulose-based antioxidant and antimicrobial active packaging film incorporated with curcumin and zinc oxide. International Journal of Biological Macromolecules 2020, 148, 666-676, 10.1016/j.ijbiomac.2020.01.204.
  222. Parya Ezati; Jong-Whan Rhim; pH-responsive pectin-based multifunctional films incorporated with curcumin and sulfur nanoparticles. Carbohydrate Polymers 2020, 230, 115638, 10.1016/j.carbpol.2019.115638.
  223. Ayca Aydogdu; Clayton J. Radke; Semih Bezci; Emrah Kirtil; Characterization of curcumin incorporated guar gum/orange oil antimicrobial emulsion films. International Journal of Biological Macromolecules 2020, 148, 110-120, 10.1016/j.ijbiomac.2019.12.255.
  224. Chongshan Dai; Yang Wang; Gaurav Sharma; Jianzhong Shen; Tony Velkov; Xilong Xiao; Polymyxins–Curcumin Combination Antimicrobial Therapy: Safety Implications and Efficacy for Infection Treatment. Antioxidants 2020, 9, 506, 10.3390/antiox9060506.
  225. Mohammed J. Hakeem; Khalid A. Asseri; Luyao Ma; Keng C. Chou; Michael E. Konkel; Xiaonan Lu; A Novel Mathematical Model for Studying Antimicrobial Interactions Against Campylobacter jejuni. Frontiers in Microbiology 2019, 10, 1038, 10.3389/fmicb.2019.01038.
  226. Marco Aurélio Paschoal; Cíntia Maria Zanin Moura; Fabiano Jeremias; Juliana Feltrin Souza; Vanderlei S. Bagnato; Juçaíra S. M. Giusti; Lourdes Santos-Pinto; Longitudinal effect of curcumin-photodynamic antimicrobial chemotherapy in adolescents during fixed orthodontic treatment: a single-blind randomized clinical trial study. Lasers in Medical Science 2014, 30, 2059-2065, 10.1007/s10103-014-1700-7.
More
Information
Subjects: Microbiology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 807
Revisions: 2 times (View History)
Update Date: 23 Jul 2021
1000/1000