You are on page 1of 10

Critical ReviewsTM in Eukaryotic Gene Expression, 29(5):377 – 385 (2019)

Review of Anti-Bacterial Activities of Curcumin


against Pseudomonas aeruginosa
Zohreh Neyestani,a Seyed Ali Ebrahimi,b Anahita Ghazaghi,b Amin Jalili,c
Amirhossein Sahebkar,d,e,f,* & Hamid Reza Rahimid,*
a
Department of Microbiology, Faculty of Sciences, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran;
b
Student Research Committee, Islamic Azad University, Mashhad branch, Mashhad, Iran; cDepartment of Medical
Biotechnology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; dNeurogenic
Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; eBiotechnology Research
Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; fSchool of
Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
*Address all correspondence to: Amirhosein Sahebkar, Tel.: +985138002288, E-mail: sahebkara@mums.ac.ir; amir_saheb2000@yahoo.com; and
Hamid Reza Rahimi, Tel.: +985138002288, E-mail: rahimihr@mums.ac.ir

ABSTRACT: The prevention and pharmacotherapy of infectious diseases are of great importance. Among others, infec-
tions caused by Pseudomonas aeruginosa have a high mortality rate. This bacterium is the third most common cause of
nosocomial infections, and characteristics such as multiple virulence factors, ability to survive, environmental spread,
and resistance to antibiotics have made it a potential pathogen, especially for people with compromised immune systems.
Considering bacterial resistance to current medications, high cost, and side effects, the need to provide new and effective
therapies is highlighted. Curcumin is a dietary polyphenolic compound that has a wide range of therapeutic properties,
including antibacterial effects. It has been the subject of increasing research exploring its potential utility in infectious
diseases. In this review, the antibacterial effects of curcumin against Pseudomonas aeruginosa are discussed.

KEY WORDS: bacterial infections, Pseudomonas aeruginosa, curcumin, quorum sensing

I. INTRODUCTION attributed to curcumin, owing to its abundance in the


plant. Generally, using a combination of curcumi-
Curcumin is the main active ingredient of turmeric noids provides better effects compared to curcumin
(Curcuma longa L.), imparting the orange-yellow alone.5 Several studies have been conducted on the
color of this spice.1 Turmeric has a long repu- biological effects of curcumin and curcuminoids in
tation of therapeutic applications in several tra- recent years. More than 3,000 studies have shown
ditional systems of medicine, including Iranian their numerous pharmacological effects, including
traditional medicine, in which topical use is rec- antioxidant, antifungal, antiviral, anti-inflamma-
ommended for reducing joint pain and swelling tory, antiproliferative, proapoptotic, and especially
because of the anti-inflammatory effects of the antibacterial (Fig. 1).6–9 In addition, the therapeutic
plant.2 Modern phytopharmacological investiga- value of curcuminoids has been reported against
tions have revealed that many of the putative ther- numerous pathological conditions and human dis-
apeutic properties of turmeric are due to curcumin. eases, such as neurodegenerative diseases, arthritis,
Depending on the origin and quality of the plant rhi- diabetes, psoriasis, Alzheimer’s disease, depression,
zome, curcumin constitutes 2% to 8% of turmeric. AIDS, multiple sclerosis, allergies, inflammatory
Turmeric contains three different structural ana- bowel disease, nephrotoxicity, cardiovascular dis-
logs that are collectively known as curcuminoids, ease, and cancer.10
including curcumin, demethoxycurcumin, and Owing to its antimicrobial activity and safety
bisdemethoxycurcumin.3,4 of use even at high doses (12 g/day), curcumin has
Although it is not clear that all existing analogs been evaluated in some studies and has been used
have the same pharmacological potency, in most as a structural lead for designing new antimicrobial
cases the therapeutic properties of curcuminoids are agents with enhanced antimicrobial effects.6

1045-4403/19/$35.00 © 2019 by Begell House, Inc. www.begellhouse.com377


378 Neyestani et al.

FIG. 1: Antimicrobial and biological functions of curcumin

II. CURCUMIN at 183°C. Turmeric grows naturally throughout the


Indian subcontinent and in tropical countries, espe-
Curcumin was discovered about two centuries ago, cially in Southeast Asia and China, and has been
and for the first time, in 1815, Vogel and Pelletier iso- used in traditional medicine for thousands of years.16
lated it from the turmeric plant rhizome in the form Since the free form of curcumin has low bio-
of an impure colored compound.11 In 1842, Vogel availability, various strategies have been developed
purified curcumin; however, its formula remained to improve its bioavailability, leading to the produc-
unknown.12 In 1910, Milobedzka et al. identified the tion of curcumin nanomicelles and nanoparticles,17
structure and chemical formula of curcumin, and in which have been shown to significantly enhance
1913 he and Lampe synthesized it.13 In 1953, Sri- curcumin bioavailability.18 Research has shown that
nivasan identified and isolated components of cur- curcumin can protect the liver against alcoholic liver
cuminoids using chromatography.14 disease (ALD) by inhibiting the activation of sev-
The chemical formula of curcumin is eral NF-κB–reactive genes that are activated during
1,7-bis(4-hydroxy-3-methoxyphenyl)-1,6-heptadi- alcohol use.19 It has also been shown that this com-
ene-3,5-dione) (Fig. 2). It is insoluble in water and pound reduces HbA1c levels and improves glyce-
ether, but soluble in ethanol, dimethyl sulfoxide, mic control in type 2 diabetes.20
and acetone.15 Its molecular formula is C21H20O, In one study, oral administration of curcumin
and its molecular weight is 368.37 g/mol.12 It melts for 1 month (1 g/day) reduced the concentration of

FIG. 2: Chemical structure of curcumin

Critical ReviewsTM in Eukaryotic Gene Expression


Activities of Curcumin against Pseudomonas aeruginosa379

triglycerides in obese people.21 Other studies have against H. pylori.30 In vitro studies on the antibac-
shown that nano-curcumin can suppress the growth terial effects of three new curcumin compounds—
of esophageal squamous carcinoma cells, which indium curcumin, indium diacetylcurcumin, and
may be associated with decreased regulation of the diacetylcurcumin—against S. aureus, S. epider-
cyclin D1 gene.22 For reasons such as these, accord- midis, E. coli, and P. aeruginosa showed that indium
ing to its antimicrobial effects, its diverse therapeu- curcumin has better antibacterial effects than cur-
tic properties, and its low side effects,23 curcumin cumin itself against the tested bacteria while diace-
is used in the prevention and treatment of various tylcurcumin has none.31 These results indicated that
disorders. there might be a promising antimicrobial potential
in different curcumin derivatives.
III. CURCUMIN’S ANTIBACTERIAL FUNCTION Given the emergence of drug-resistant bacte-
ria, it is essential to study the synergistic effects of
For more than 50 years, extensive research has antibiotics in combination with herbal derivatives to
been carried out to obtain antibiotics from different develop antimicrobial strategies with a wide range
sources, as bacterial infections are one of the most of activities and lower side effects.32 Because com-
important infectious diseases. Although progress binational therapies increase the likelihood that the
has been made in developing antibacterial agents, infectious pathogen will be susceptible to at least one
the demand to find new ones still exists due to of the components of the drug, they can overcome
the emergence of multidrug-resistant bacteria.24 drug-resistant strains.33 Using two or more factors,
Increased drug resistance and multidrug resistant they also have synergistic effects that increase effi-
strains arising from the arbitrary use of antibiotics ciency or reduce the dosage while maintaining effi-
and, on the other hand, the intrinsic and acquired cacy.34 In this regard, curcumin showed synergistic
resistance of bacteria including P. aeruginosa, which effects in combination with some antibiotics, includ-
is resistant to three antibiotic families25—all of these ing ampicillin, oxacillin, and norfloxacin, against
factors have made controlling hospital infections methicillin-resistant S. aureus.35 Additionally, the
difficult.26 combination of curcumin with cobalt nanoparticles
In vitro studies have shown the antimicro- showed increased antimicrobial activity against E.
bial potential of curcumin against a wide range of coli.36 Curcumin-impregnated silver nanocompos-
microorganisms, including fungi,27 as well as sev- ite films also produced stronger antibacterial effects
eral Gram-positive and Gram-negative bacteria.25 In against E. coli.37 These results show that curcumin
this regard, antimicrobial evaluation of the aqueous is a natural antioxidant and anti-inflammatory com-
extract of the turmeric rhizome has shown that the pound with high efficacy and low cytotoxicity,
minimum inhibitory concentration is 4 to 16 g/L making it a potentially ideal candidate for use in
and the minimum bactericidal concentration is 16 combination therapy against bacterial infections.33
to 32 g/L against Staphylococcus epidermidis ATCC
12228, Staphylococcus aureus ATCC 25923, Kleb- IV. P. AERUGINOSA AND ITS
siella pneumoniae ATCC 10031, and Escherichia DRUG-RESISTANCE MECHANISMS
coli ATCC 25922. Furthermore, hexane and metha-
nol extracts of this plant showed antibacterial effects P. aeruginosa is known to be a Gram-negative,
against 13 bacteria, including a number of Vibrio, drug-resistant, opportunistic pathogen. Its multi-
Staphylococcus, and Streptococcus species.28 In drug resistance is the result of a set of factors, the
addition, it was shown that adding 0.3% w/v of entirety of which is beyond the scope of this review.
aqueous extract of turmeric to cheese reduces the Rather, we highlight only those associated with the
presence Salmonella typhimurium, P. aeruginosa, antimicrobial function of curcumin: low permeable
and Escherichia coli bacteria 0157:H7.29 outer membrane, expression of efflux pumps, and
Curcumin showed significant antibacterial a recently discovered phenomenon called quorum
activity, with MIC values between 5 and 50 μg/ml sensing, which is associated with biofilm formation.

Volume 29, Issue 5, 2019


380 Neyestani et al.

The outer membrane of Gram-negative bacteria hospitalization, long-term costs, and failure of
plays a crucial role as a permeability barrier that is treatment.29,46
dependent on solute size. The antimicrobial resis-
tance of P. aeruginosa has been linked to the low V. CURCUMIN’S ANTIBACTERIAL FUNCTION
permeability of its outer membrane. In two studies AGAINST P. AERUGINOSA
involving purified porins, Hancock et al. showed
that the P. aeruginosa porin produces a significantly A number of recent studies have confirmed the poten-
larger pore than the pores of some intestinal bacte- tial of curcumin against P. aeruginosa (Table 1).
ria such as S. typhimurium and E. coli.38,39 Increased One study carried out in 2014 reported that the bac-
sensitivity to antimicrobials was found after damage tericidal function of curcumin against P. aeruginosa
to or removal of the bacteria’s outer membrane.40 increased proportionally to its concentration and that
Yoshimura and Nikaido determined the permeabil- 100 μM of curcumin caused 100% death of the bac-
ity of P. aeruginosa’s outer membrane directly as terium in two hours.33 A similar in vivo animal study
about 100-fold lower than the outer membrane of showed that chitosan tri-polyphosphate nanoparti-
E. coli.41 These findings show that achieving high cles containing curcumin significantly suppressed
concentrations of antimicrobials in P. aeruginosa is the progression of P. aeruginosa infection in mice
difficult because its low permeability contributes to while chitosan tri-polyphosphate alone was unable
its antimicrobial resistance. to do so.34 Given these results, what is the under-
Four multidrug efflux pumps are characterized ling mechanism of the antimicrobial function of cur-
in P. aeruginosa: MexAB-OprM, MexEF-OprN, cumin from a microbiological point of view?
MexCD-OprJ, and MexXY-OprM. Efflux pumps A recent study demonstrated that the bacteri-
MexAB-OprM, MexEF-OprN, and MexCD-OprJ cidal activity of curcumin is associated with dam-
provide resistance to β-lactam antibiotics. Moreover, age to the bacterial membrane. The integrity of P.
overexpression of MexEF-OprN and MexCD-OprJ aeruginosa, E. coli, S. aureus, and E. faecalis mem-
enables the bacterium to resist fluoroquinolones, and branes was checked by two differential permeabi-
overexpression of MexXY-OprM enhances its resis- lization–indicating fluorescent probes: propidium
tance to aminoglycoside.42 Through these pumps, iodide and calcein. Membrane leakage in all four
the bacterium disposes of antimicrobial agents and bacteria exposed to curcumin was observed.33
other toxic substances, and causes difficult-to-treat, As aforementioned, efflux pumps are responsi-
life-threatening infections.43 If these efflux pumps ble for the discharge of antibiotics from P. aerugi-
are blocked or inhibited, increasing the drug con- nosa. It has been shown that curcumin inhibits their
centration in the bacteria can produce a sufficient expression and thus overcomes drug resistance. Negi
and lethal dosage. et al. used 30 multidrug-resistant strains of P. aeru-
Quorum sensing is a language that allows bac- ginosa derived from clinical isolates and derived
teria to manifest multicellular behaviors and engage MIC values for curcumin (50 mg/L) with and with-
in cell-to-cell communication. Using this system, out antibiotics. They observed significant reduc-
the bacterium determines its population density tions in MIC for curcumin combined with selected
and so regulates the expression of particular genes. antimicrobial agents in 30% of multidrug-resistant
Bacteria use quorum sensing to regulate a variety isolates of P. aeruginosa but no change in MIC for
of physiological functions, including virulence, curcumin alone, indicating curcumin’s efflux pump
conjugation, motility, and biofilm formation.44 Bio- inhibitory activity.47
films are an aggregate of microorganisms within a After determining the role of quorum sensing
self-produced matrix of extracellular polymeric sub- in various vital processes, including expression of
stances that adhere to each other or to a surface.45 pathogenicity factors and the development of resis-
They contribute to the chronicity and recurrence of tant bacterial infections, efforts were made to use it
infection by inhibiting the penetration of a drug into to directly target pathogenic factors as a solution to
the extracellular matrix. The end result is prolonged antibiotic resistance.48 In this regard, various studies

Critical ReviewsTM in Eukaryotic Gene Expression


Activities of Curcumin against Pseudomonas aeruginosa381

TABLE 1: Studies evaluating curcumin’s antimicrobial effects against P. aeruginosa


Study Year Study type Effect Ref
Tajbakhsh et al. 2008 In vitro Indium diacetylcurcumin activity against P. aeruginosa 31
Rudrappa and Bais 2008 In vivo/in vitro Reduced expression of genes involved in quorum sensing and 25
biofilm formation in P. aeruginosa
Niamsa and Sittiwet 2009 In vitro No significant inhibitory effect against P. aeruginosa 26
Hosny et al. 2011 In vitro Addition of 0.3% w/v of aqueous extract of turmeric to cheese 29
decreased P. aeruginosa
Mirnejad et al. 2014 In vivo Curcumin-loaded chitosan-TPP nanoparticles inhibited S. 34
aureus and P. aeruginosa growth
Packiavathy et al. 2014 In vitro Inhibition of P. aeruginosa biofilm formation possibly by 57
interfering with quorum sensing
Negi et al. 2014 In vitro Significant reduction in MIC after adding curcumin with 47
antimicrobials in 30% of multidrug-resistant isolates of P.
aeruginosa, indicating efflux pump inhibitor activity
Tyagi et al. 2015 In vitro 100% inhibition of P. aeruginosa membrane leakage of 33
bacteria
Bahari et al. 2017 In vitro Significant decrease in MIC of azithromycin and gentamicin, 56
indicating synergism against P. aeruginosa quorum sensing

have shown that curcumin inhibits quorum sensing differing reports are controversial. However, one
in P. aeruginosa by affecting pathogenicity factors,29 study reported the highest frequency for LasI (60%)
in this way preventing the formation of bacterial and LasR (48.3%). Aghamollaei et al. reported
biofilms. Many genes in P. aeruginosa that play a the frequency of the quorum-sensing LasI gene as
role in its pathogenicity are regulated and expressed 46.5%.46 In this regard, it has been shown that there
by quorum sensing—for instance, lasI, lasR, rhlI, is a correlation between defective virulence factor
and rhlR and the human lasB, apr, and rhlAB house- production and increased sensitivity to antibiotics.54
keeping genes.50 Therefore, the quorum-sensing process is a suitable
There are two complete quorum-sensing systems target for the development of anti-quorum-sensing
in P. aeruginosa: (1) las, which includes the lasR and therapies independent of antibiotics, which are con-
lasI transcription-activating genes that regulate elas- sidered an effective strategy to fight against bacte-
tase, exotoxin A, and alkaline protease production; rial infections.
and (2) rhl, which encompasses the rhlR and rhlI Since curcumin has shown synergic effects
genes that regulate the production of rhamnolipid, when combined with antibiotics against P. aerugi-
alkaline protease, elastase, cyanide, and pyocyanin.51 nosa,55 its inhibitory effect with azithromycin and
rhlR and rhlI produce transcription-regulating gentamicin against the bacterium’s quorum-sens-
proteins that activate virulence genes.52 The las ing signal transduction system was investigated.
system is responsible for the transcription and The results showed a significant reduction in the
expression of other virulence factors and pyocy- pathogenicity associated with quorum sensing, such
anin pigment, and for the production of biofilms as biofilm formation and motility with azithromy-
that contribute to bacterial invasion of host cells cin, gentamicin, and curcumin in 1.4 and 1.16 MIC
and resistance to host immunity.50,53 Because of alone and in combination. It was also reported that
the different virulence genes in P. aeruginosa and the combination of azithromycin and curcumin at a
because of the importance of quorum-sensing genes, concentration of 1.4 MIC had the highest inhibitory
the frequency of these genes is not well defined and effect on biofilm formation and motility.56

Volume 29, Issue 5, 2019


382 Neyestani et al.

Measurement of expression of quorum sensing– antimicrobial effects of curcumin against P. aerugi-


regulating genes, including lasI, laR, rhlI, and rhlR, nosa. The low bioavailability and variable pharma-
showed that azithromycin, gentamicin, and cur- cokinetics of different oral formulations of curcumin
cumin in 1.4 MIC alone and in combination inhibits may represent important limitations in the design
quorum sensing at the transcription level. RT-PCR and conduct of interventional studies. However, in
analysis showed that, compared to nonintervention recent years, new pharmacological techniques have
bacteria, the relative expression of lasR decreased largely contributed to solving the bioavailability
significantly in the presence of azithromycin alone problem. In this case, the drug Meriva is a good
and decreased by 80% with the addition of cur- example. Meriva is a proprietary curcumin-phos-
cumin. Azithromycin alone and in combination with phatidylcholine phytosome complex that has shown
curcumin reduced the expression of rhlI by 60% and 29-fold higher absorption than ordinary curcumin.58
67%, respectively.56 Another example is SinaCurcumin, in which cur-
It has been shown that the combination of cur- cumin is within the hydrophobic section of the
cumin and antibiotics such as azithromycin and nanomicelles. These nanomicelles increase the sol-
gentamicin exhibits synergistic effects against quo- ubility of curcumin in water and thus increase its
rum sensing in P. aeruginosa.56 Packiavathy et al. absorption.20 Given the promising data on the poten-
reported that curcumin consistently inhibited the for- tial antimicrobial function of curcumin, we suggest
mation of E. coli biofilms and P. aeruginosa PAO1, that researchers carry out randomized clinical trials
Serratia marcescens, and Proteus mirabilis by sup- addressing this issue.
pressing the signal cascade of the quorum-sensing
process (Fig. 3).57 VI. CONCLUSION
Finally, it should be noted that one of the cur-
rent issues surrounding the antimicrobial effects of Curcumin, the active ingredient in turmeric, has
curcumin is the lack of clinical studies. So far no been an object of study for scientists for many years
interventional studies have been carried out on the because of its unique biological and therapeutic
properties.59 As shown by the research discussed
in this review, curcumin has different antibacterial
properties against various types of Gram-negative
bacteria, in particular P. aeruginosa, which is the
focus of this paper.
The results of various studies show that the
expression of most genes producing P. aeruginosa
pathogenic factors is regulated by a gene system
called quorum sensing. Curcumin can inhibit quo-
rum sensing by influencing its pathogenicity fac-
tors.29 In doing so, it inhibits biofilm growth,29
reestablishing bacterial sensitivity to antibiotics.60
This has led to the use of curcumin as an effective
therapy in combination with antibiotics.6 Results
show that curcumin has the ability to overcome drug
resistance by inhibiting the expression of efflux
pumps in P. aeruginosa, bestowing promise for its
use in the treatment of multidrug-resistant bacterial
infections.6,53 Curcumin should therefore be consid-
ered a valuable recruit in the battle against bacterial
FIG. 3: Antibacterial function of curcumin against Pseu- infections caused by P. aeruginosa, though more
domonas aeruginosa comprehensive studies are needed to confirm this.

Critical ReviewsTM in Eukaryotic Gene Expression


Activities of Curcumin against Pseudomonas aeruginosa383

REFERENCES 18. Rahimi HR, Jaafari MR, Mohammadpour AH, Abnous


K, Ghayour Mobarhan M, Ramezanzadeh E, Mousavi F,
1. Jagetia GC, Aggarwal BB. “Spicing up” of the immune Kazemi Oskuee R. Curcumin: reintroduced therapeutic
system by curcumin. J Clin Immunol. 2007;27(1):19–35. agent from traditional medicine for alcoholic liver disease.
2. Rahimi HR, Kazemi Oskuee R. Curcumin from tradi- Asia Pacific J Med Toxicol. 2015;4(1):25–30.
tional Iranian medicine to molecular medicine. Razavi Int 19. Nanji AA, Jokelainen K, Tipoe GL, Rahemtulla A,
J Med. 2014;2(2):e19982. Thomas P, Dannenberg AJ. Curcumin prevents alcohol-
3. Mirzaei H, Shakeri A, Rashidi B, Jalili A, Banikazemi Z, induced liver disease in rats by inhibiting the expression of
Sahebkar A. Phytosomal curcumin: a review of pharma- NF-κB-dependent genes. Am J Physiolo-Gastrointestinal
cokinetic, experimental and clinical studies. Biomed Phar- Liver Physiol. 2003;284(2):G321–7.
macother. 2017;85:102–12. 20. Rahimi HR, Mohammadpour AH, Dastani M, Jaafari
4. Chainani-Wu N. Safety and anti-inflammatory activity of MR, Abnous K, Mobarhan MG, Oskuee RK. The effect
curcumin: a component of tumeric (Curcuma longa). J Alt of nano-curcumin on HbA1c, fasting blood glucose, and
Comp Med. 2003;9(1):161–8. lipid profile in diabetic subjects: a randomized clinical
5. Khurana A, Ho C-T. High performance liquid chro- trial. Avicenna J Phytomed. 2016;6(5):567.
matographic analysis of curcuminoids and their photo- 21. Mohammadi A, Sahebkar A, Iranshahi M, Amini M,
oxidative decomposition compounds in Curcuma longa L. Khojasteh R, Ghayour-Mobarhan M, Ferns GA. Effects
J Liquid Chromatography. 1988;11(11):2295–304. of supplementation with curcuminoids on dyslipidemia
6. Zorofchian Moghadamtousi S, Abdul Kadir H, Hassan- in obese patients: a randomized crossover trial. Phytother
darvish P, Tajik H, Abubakar S, Zandi K. A review on anti- Res. 2013;27(3):374–9.
bacterial, antiviral, and antifungal activity of curcumin. 22. Hosseini S, Chamani J, Rahimi H, Azmoodeh N, Ghasemi
BioMed Res Int. 2014;186864. F, Abadi PH. An in vitro study on curcumin delivery by
7. Momtazi AA, Shahabipour F, Khatibi S, Johnston TP, nano-micelles for esophageal squamous cell carcinoma
Pirro M, Sahebkar A. Curcumin as a microRNA regula- (KYSE-30). Rep Biochem Mol Biol. 2018;6(2):137.
tor in cancer: a review. Rev Physiol Biochem Pharmacol. 23. Singh S. From exotic spice to modern drug? Cell.
2016;171:1–38. 2007;130(5):765–8.
8. Karimian MS, Pirro M, Majeed M, Sahebkar A. Cur- 24. Wise R, Hart T, Cars O, Streulens M, Helmuth R,
cumin as a natural regulator of monocyte chemoattractant Huovinen P, Sprenger M. Antimicrobial resistance is
protein-1. Cytokine Growth Factor Rev. 2017;33:55–63. a major threat to public health. BMJ (Clin Res Ed).
9. Panahi Y, Kianpour P, Mohtashami R, Jafari R, Simental- 1998;317(7159):609–10.
Mendia LE, Sahebkar A. Curcumin lowers serum lipids 25. Rudrappa T, Bais HP. Curcumin, a known phenolic from
and uric acid in subjects with nonalcoholic fatty liver dis- Curcuma longa, attenuates the virulence of Pseudomo-
ease: a randomized controlled trial. J Cardiovasc Pharma- nas aeruginosa PAO1 in whole plant and animal patho-
col. 2016;68(3):223–9. genicity models. J Agricultural Food Chem. 2008;56(6):
10. Shishodia S, Sethi G, Aggarwal BB. Curcumin: getting back 1955–62.
to the roots. Ann New York Acad Sci. 2005;1056(1):206–17. 26. Niamsa N, Sittiwet C. Antimicrobial activity of Cur-
11. Vogel A, Pelletier J. Examen chimique de la racine de cur- cuma longa aqueous extract. J Pharmacol Toxicol.
cuma. J Pharm. 1815;1:289–300. 2009;4(4):173–7.
12. Vogel A. Mémoire sur la curcumine. J de Pharm et Chim. 27. Neelofar K, Shreaz S, Rimple B, Muralidhar S, Nikhat M,
1842;2:20–7. Khan LA. Curcumin as a promising anticandidal of clini-
13. Lampe V, Milobedzka J. Studien über curcumin. cal interest. Can J Microbiol. 2011;57(3):204–10.
Berichte der Deutschen Chemischen Gesellschaft. 28. Lawhavinit O-a, Kongkathip N, Kongkathip B. Antimi-
1913;46(2):2235–40. crobial activity of curcuminoids from Curcuma longa L.
14. Srinivasan K. A chromatographic study of the cur- on pathogenic bacteria of shrimp and chicken. Kasetsart
cuminoids in Curcuma longa L. J Pharm Pharmacol. J—Natural Sci. 2010;44(3):364–71.
1953;5(1):448–57. 29. Hosny I, El Kholy W, Murad H, El Dairouty R. Antimi-
15. Tønnesen HH, Karlsen J. Studies on curcumin and cur- crobial activity of curcumin upon pathogenic microor-
cuminoids. Zeitschrift für Lebensmitteluntersuchung ganisms during manufacture and storage of a novel style
und-Forschung A. 1985;180(5):402–4. cheese “Karishcum.” J Am Sci. 2011;7(5):611–8.
16. Maheshwari RK, Singh AK, Gaddipati J, Srimal RC. Mul- 30. De R, Kundu P, Swarnakar S, Ramamurthy T, Chowdhury
tiple biological activities of curcumin: a short review. Life A, Nair GB, Mukhopadhyay AK. Antimicrobial activity
Sci. 2006;78(18):2081–7. of curcumin against Helicobacter pylori isolates from
17. Hatcher H, Planalp R, Cho J, Torti F, Torti S. Curcumin: India and during infections in mice. Antimicrob Agents
from ancient medicine to current clinical trials. Cell Mol Chemother. 2009;53(4):1592–7.
Life Sci. 2008;65(11):1631–52. 31. Tajbakhsh S, Mohammadi K, Deilami I, Zandi K,

Volume 29, Issue 5, 2019


384 Neyestani et al.

Fouladvand M, Ramedani E, Asayesh G. Antibacterial 44. Miller MB, Bassler BL. Quorum sensing in bacteria. Ann
activity of indium curcumin and indium diacetylcurcumin. Rev Microbiol. 2001;55:165–99.
African J Biotechnol. 2008;7(21). 45. Vert M, Doi Y, Hellwich K-H, Hess M, Hodge P, Kubisa
32. Odds FC. Synergy, antagonism, and what the chequer- P, Rinaudo M, Schué F. Terminology for biorelated poly-
board puts between them. J Antimicrob Chemother. mers and applications (IUPAC Recommendations 2012).
2003;52(1):1. Pure Appl Chem. 2012. 2012;84:377–410.
33. Tyagi P, Singh M, Kumari H, Kumari A, Mukhopad- 46. Aghamollaei H, Azizi Barjini K, Moosazadeh Mogaddam
hyay K. Bactericidal activity of curcumin I is associ- M. Rapid detection of Pseudomonas aeruginosa by PCR
ated with damaging of bacterial membrane. PLoS One. method using specific primers of quorum sensing lasI
2015;10(3):e0121313. gene. Armaghane Danesh. 2013;18(9):723–35.
34. Mirnejad R, Jahromi M, Ali M, Al-Musawi S, Pirestani 47. Negi N, Prakash P, Gupta ML, Mohapatra TM. Possible
M, Fasihi Ramandi M, Ahmadi K, Rajayi H, Mohammad role of curcumin as an efflux pump inhibitor in multi drug
Hassan Z, Kamali M. Curcumin-loaded chitosan tripoly- resistant clinical isolates of Pseudomonas aeruginosa. J
phosphate nanoparticles as a safe, natural and effective Clin Diagn Res. 2014;8(10):DC04.
antibiotic inhibits the infection of Staphylococcus aureus 48. Miller MB, Bassler BL. Quorum sensing in bacteria. Ann
and Pseudomonas aeruginosa in vivo. Iranian J Biotech- Rev Microbiol. 2001;55(1):165–99.
nol. 2014;12(3):1–8. 49. Zhu H, Bandara R, Conibear TC, Thuruthyil SJ, Rice
35. Mun S-H, Joung D-K, Kim Y-S, Kang O-H, Kim S-B, SA, Kjelleberg S, Givskov M, Willcox MD. Pseudo-
Seo Y-S, Kim Y-C, Lee D-S, Shin D-W, Kweon K-T. monas aeruginosa with lasI quorum-sensing deficiency
Synergistic antibacterial effect of curcumin against meth- during corneal infection. Invest Ophthalmol Vis Sci.
icillin-resistant Staphylococcus aureus. Phytomedicine. 2004;45(6):1897–903.
2013;20(8-9):714–8. 50. Sharma G, Rao S, Bansal A, Dang S, Gupta S, Gabrani
36. Hatamie S, Nouri M, Karandikar S, Kulkarni A, Dhole S, R. Pseudomonas aeruginosa biofilm: potential therapeutic
Phase D, Kale S. Complexes of cobalt nanoparticles and targets. Biologicals. 2014;42(1):1–7.
polyfunctional curcumin as antimicrobial agents. Mater 51. Senturk S, Ulusoy S, Bosgelmez-Tinaz G, Yagci A. Quo-
Sci Eng: C. 2012;32(2):92–7. rum sensing and virulence of Pseudomonas aeruginosa
37. Varaprasad K, Mohan YM, Vimala K, Mohana Raju during urinary tract infections. J Infect Develop Coun-
K. Synthesis and characterization of hydrogel-silver tries. 2012;6(06):501–7.
nanoparticle-curcumin composites for wound dress- 52. Kumar R, Chhibber S, Gupta V, Harjai K. Screening
ing and antibacterial application. J Appl Polymer Sci. and profiling of quorum sensing signal molecules in
2011;121(2):784–96. Pseudomonas aeruginosa isolates from catheterized uri-
38. Hancock REW, Decad GM, Nikaido H. Identifica- nary tract infection patients. Indian J Med Res. 2011;
tion of the protein producing transmembrane diffusion 134(2):208.
pores in the outer membrane of Pseudomonas aerugi- 53. Chowdhury N, Bagchi A. Molecular insight into the activ-
nosa PA01. Biochim Biophys Acta—Biomembranes. ity of LasR protein from Pseudomonas aeruginosa in the
1979;554(2):323–31. regulation of virulence gene expression by this organism.
39. Hancock RE, Nikaido H. Outer membranes of Gram- Gene. 2016;580(1):80–7.
negative bacteria. XIX. Isolation from Pseudomonas 54. Marston HD, Dixon DM, Knisely JM, Palmore TN, Fauci
aeruginosa PAO1 and use in reconstitution and defini- AS. Antimicrobial resistance. JAMA. 2016;316(11):
tion of the permeability barrier. J Bacteriol. 1978;136(1): 1193–204.
381–90. 55. Betts JW, Wareham DW. In vitro activity of curcumin in
40. Sykes RB, Morris A. Resistance of Pseudomonas Aerugi- combination with epigallocatechin gallate (EGCG) ver-
nosa to Antimicrobial Drugs. In: Ellis GP, West GB, edi- sus multidrug-resistant Acinetobacter baumannii. BMC
tors. Progress in medicinal chemistry. vol. 12. New York: Microbiol. 2014;14(1):172.
Elsevier; 1975. p. 333–93. 56. Bahari S, Zeighami H, Mirshahabi H, Roudashti S, Haghi
41. Yoshimura F, Nikaido H. Permeability of Pseudomonas F. Inhibition of Pseudomonas aeruginosa quorum sensing
aeruginosa outer membrane to hydrophilic solutes. J Bac- by subinhibitory concentrations of curcumin with gen-
teriol. 1982;152(2):636–42. tamicin and azithromycin. J Global Antimicrob Resist.
42. Livermore DM. Multiple mechanisms of antimicrobial 2017;10:21–8.
resistance in Pseudomonas aeruginosa: our worst night- 57. Packiavathy IASV, Priya S, Pandian SK, Ravi AV. Inhi-
mare? Clin Infect Dis. 2002;34(5):634–40. bition of biofilm development of uropathogens by cur-
43. Aeschlimann JR. The role of multidrug efflux pumps in cumin—an anti-quorum sensing agent from Curcuma
the antibiotic resistance of Pseudomonas aeruginosa and longa. Food Chem. 2014;148:453–60.
other Gram-negative bacteria. Pharmacother: J Human 58. Cuomo J, Appendino G, Dern AS, Schneider E,
Pharmacol Drug Ther. 2012;23(7):916–24. McKinnon TP, Brown MJ, Togni S, Dixon BM.

Critical ReviewsTM in Eukaryotic Gene Expression


Activities of Curcumin against Pseudomonas aeruginosa385

Comparative absorption of a standardized curcuminoid ciated with infectious diarrhea. BioMed Res Int. 2014;
mixture and its lecithin formulation. J Natur Prod. 2011; 2014:8.
74(4):664–9. 60. Kali A, Devaraj Bhuvaneshwar P, Charles M, Seetha KS.
59. Sasidharan NK, Sreekala SR, Jacob J, Nambisan B. In Antibacterial synergy of curcumin with antibiotics against
vitro synergistic effect of curcumin in combination with biofilm producing clinical bacterial isolates. J Basic Clin
third generation cephalosporins against bacteria asso- Pharm. 2016;7(3):93.

Volume 29, Issue 5, 2019

You might also like