Skip to main content
Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
Integr Med Insights. 2017; 12: 1178633717702869.
Published online 2017 Apr 24. doi: 10.1177/1178633717702869
PMCID: PMC5406168
PMID: 28469409

Honey as a Complementary Medicine

Abstract

The beneficial effects of honey on human health have long been recognized. Today, many of those positive effects have been studied to elucidate its mode of action. This review briefly summarizes the best studied features of honey, highlighting it as an appealing alternative medicine. In these reports, the health benefits of honey range from antioxidant, immunomodulatory, and anti-inflammatory activity to anticancer action, metabolic and cardiovascular benefits, prebiotic properties, human pathogen control, and antiviral activity. These studies also support that the honey’s biological activity is mainly dependent on its floral or geographic origin. In addition, some promising synergies between honey and antibiotics have been found, as well as some antiviral properties that require further investigation. Altogether, these studies show that honey is effectively a nutraceutical foodstuff.

Keywords: Honey, pathogens, biofilm, prebiotic, metabolism, cancer, heart

Introduction

Honey is produced by bees from the genus Apis, which collect the nectar from plants or from secretions of aphids (plant-sucking insects belonging to the genus Rhynchota).1 It is a sweet and flavourful natural product, supersaturated in sugars, with high nutritive value. Besides the sugars, other minor components are present in honey, such as minerals, polyphenols, vitamins, carotenoids, amino acids, proteins, enzymes (glucose oxidase and catalase [CAT]), organic acids, and volatile compounds. In addition to the described prebiotic effects associated with oligosaccharides,2 most of the reported biological properties of honey (eg, antioxidant, antibacterial, antiviral, anti-inflammatory, antiulcerous, immunomodulating, vasodilative, hypotensive, antihypercholesterolemic, antibrowning, disinfectant, and antitumour), and many of its applications, may be attributed to those minor components.24 However, each honey’s particular composition is highly dependent on the floral source, geographical region, and season, as well as the processing conducted after the harvest.3,5

Chemical Composition

Honey is mainly constituted by sugars and in much lower quantities by amino acids, proteins, enzymes, organic acids, vitamins, minerals, volatile substances, and polyphenols.6

Fructose and glucose (monosaccharides) represent about 75% of the sugars found in honey. Disaccharides (sucrose, maltose, turanose, isomaltose, maltulose, trehalose, nigerose, and kojibiose) and trisaccharides (maltotriose and melezitose) among other oligosaccharides constitute the remaining 10% to 15% of the carbohydrates in honey.5,6

Usually, fructose is honey’s most predominant sugar, with acacia being one of the plants that yields the highest amount of this monosaccharide.7,8 Nevertheless, there are exceptions, eg, honey from rape (Brassica napus) and dandelion (Taraxacum officinale) has higher concentrations of glucose than fructose.7 These findings suggest that carbohydrates may play a role in the identification of botanical and/or geographical origins of monofloral honeys, along with other parameters, such as water activity and electrical conductivity.9 Beyond the concentrations of glucose and fructose, their ratio may also contribute to classifying monofloral honeys, as well as the concentrations of some minor oligosaccharides and/or their combinations.79

Proteins (0.5%) are present in honey mainly not only as enzymes (diastase or amylase, invertase or sucrase or α-glucosidase, CAT, and glucose oxidase) but also as individual amino acids (proline is the most important one, along with more than 20 amino acids).5,6

Organic acids (0.57%) include gluconic (the most abundant), aspartic, butyric, citric, acetic, formic, fumaric, galacturonic, gluconic, glutamic, glutaric, butyric, glyoxylic, 2-hydroxybutyric, α-hydroxyglutaric, isocitric, α-ketoglutaric, lactic, malic, malonic, methylmalonic, 2-oxopentanoic, propionic, pyruvic, quinic, shikimic, succinic, tartaric, oxalic, levulinic, and formic acids, among others.5

Honey also contains small amounts of vitamins: thiamine (B1), riboflavin (B2), nicotinic acid (B3), pantothenic acid (B5), pyridoxine (B6), biotin (B8 or H), folic acid (B9), and vitamin C.5

The mineral content in honey ranges from 0.04%, in light honeys, to 0.2%, in dark honeys. Potassium is the most abundant element. Other macroelements and microelements may also be present in honey: magnesium, calcium, iron, phosphorus, sodium, manganese, iodine, zinc, lithium, cobalt, nickel, cadmium, copper, barium, chromium, selenium, arsenic, and silver.5

The volatile compounds include diverse chemical groups, such as monoterpenes, C13-norisoprenoid, sesquiterpenes, benzene derivatives and, to a lower content of superior alcohols, esters, fatty acids, ketones, and aldehydes.5

Some volatile compounds can be used for determining floral sources and/or geographical origins of honeys. For example, methyl anthranilate and sinensal isomers were identified only in citrus honey samples,10,11 and cis-linalool oxide, nonanol, and decanal were identified in acacia honey.12 The volatile profile has been studied particularly in chestnut, heather, eucalyptus, and citrus from diverse regions, and the results have demonstrated some variability, even in the same region. However, the botanical and geographical origins are more easily achieved when the volatile compounds of hundreds of honey samples are analysed, along with their physicochemical properties, and then analysed by clustering, principal components analysis, and stepwise discriminant analysis. According to Oroian et al,13 this approach allowed the classification of honeys with more than 80% accuracy.

Polyphenols in honey may include phenolic acids (vanillic, caffeic, syringic, p-coumaric, ferulic, ellagic, 3-hydroxybenzoic, chlorogenic, 4-hydroxybenzoic, rosmarinic, gallic, and benzoic acids, among others) and flavonoids (quercetin, kaempferol, myricetin, pinobanksin, pinocembrin, chrysin, galangin, hesperetin, and others).5

The chemical diversity of phenols is also highly dependent on the botanical and geographical origins of honey. Several studies have proposed chemical markers for botanical origin taking into account the presence and abundance of one or more specific phenols.14 Table 1 presents the phenolic compounds which have been proposed as possible markers for inferring the honey’s botanical origin.

Table 1.

Potential biomarker compounds (particularly phenols) for pinpointing the botanical origins of unifloral honeys.

Floral originGeographical originCompoundsTechniqueReference
Erica sp (heather)Portugal (Coimbra)Ellagic acid, myricetin 3′-methyl etherHPLC-DADFerreres et al15
Erica sp (heather)Portugal (Serra da Lousã)Ellagic, p-hydroxybenzoic, syringic and o-coumaric acidsHPLC-DADAndrade et al16
Erica sp (heather)Not reportedEllagic acidCZE-DADAndrade et al17
Erica sp (heather)Germany, Denmark, Italy, Spain, France, The Netherlands, United Kingdom, PortugalDL-Phenyllactic acid, phenylacetic and benzoic acids, benzoic acidsHPLC-UVDimitrova et al18
Lavandula stoechas L. (lavender)Portugal (Serra da Lousã)Gallic acidHPLC-DADAndrade et al16
Lavandula stoechas L. (lavender)Not reportedNaringeninCZE-DADAndrade et al17
Lavandula sp (lavender)Germany, Denmark, Italy, Spain, France, The Netherlands, United Kingdom, PortugalGallic acid, caffeic acidHPLC-UVDimitrova et al18
Thymus capitatus Hoffgg. and LK. (thyme)Not reportedRosmarinic acidCZE-DADAndrade et al17
Eucalyptus spItaly, Portugal, Spain (all commercial)Myricetin, tricetin, luteolinHPLC-DADMartos et al19
Eucalyptus spSpain and ItalyMyricetin, tricetin, and luteolinLC-MS/MSTruchado et al20
Eucalyptus intermedia (bloodwood)AustraliaMyricetin, tricetin, luteolin, quercetin, 2 unknown flavonoidsHPLC-DADYao et al21
Eucalyptus ochrophloia (yapunyah) and Eucalyptus moluccana (gum top)AustraliaTricetin, luteolin, quercetinHPLC-DADYao et al21
Eucalyptus crebra (narrow-leaved ironbark)AustraliaLuteolin, tricetin, quercetin, unknownHPLC-DADYao et al21
Eucalyptus nubila (blue top ironbark)AustraliaLuteolin, tricetin, quercetin, myricetin, unknownHPLC_DADYao et al21
Eucalyptus globoidea (stringybark) and Eucalyptus largiflorens (black box)AustraliaTricetin, luteolin, isorhamnetinHPLC-DADYao et al21
Arbutus unedo (strawberry tree)Italy (Sardinia)Homogentisic acidHPLC-DADCabras et al22
Arbutus unedo (strawberry tree)Italy (Sardinia)Homogentisic acid, (±)-2-cis,4-trans-abscisic acid (c,t-ABA), (±)-2-trans,4-trans-abscisic acid (t,t-ABA), unedoneHPLC-DAD, LC-MS/MSTuberoso et al23
Kunzea ericoides (kanuka)North Island of New Zealand4-Methoxyphenyllactic acid, phenyllactic acid, methyl syringate, methoxylated benzoic acid, 2-methoxybenzoic acid, structural isomer of syringic acid, trimethoxybenzoic acidLC-MS/MS; GC-NPDStephens et al24
Castanea sativa Miller (chestnut)Germany, Denmark, Italy, Spain, France, The Netherlands, United Kingdom, and Portugal4-Hydroxybenzoic, DL-p-hydroxyphenyllactic, ferulic, phenylacetic acidsHPLC-UVDimitrova et al18
Helianthus annuus L. (sunflower)Germany, Denmark, Italy, Spain, France, The Netherlands, United Kingdom, and Portugalp-Coumaric, ferulic, caffeic acidsHPLC-UVDimitrova et al18
Tilia sp (Lime)Germany, Denmark, Italy, Spain, France, The Netherlands, United Kingdom, and Portugal3-Hydroxybenzoic acidHPLC-UVDimitrova et al18
Asphodelus microcarpus Salzm. et Viv. (asphodel)Italy (Sardinia)Methyl syringateLC-MS/MS; HPLC-DADTuberoso et al25

Abbreviations: CZE-DAD, capillary zone electrophoresis diode array detector; GC, gas chromatography; GC-NPD, gas chromatography with a nitrogen-phosphorus detector; HPLC-DAD, high-performance liquid chromatography-diode array detector; LC-MS/MS, liquid chromatography tandem mass spectrometry.

The certification of the floral origins for monofloral honeys based exclusively on phenols, including the flavonoids’ profile, is not enough. From Table 1, it is possible to ascertain that in some cases, for diverse floral origin of honeys, the same phenolic compounds can be found. According to Yao et al21 and Stephens et al,24 the differences lie on the percentages and/or proportions between such compounds, which hamper the floral origin authentication of honeys. In addition, although Ferreres and colleagues26,27 considered quercetin and kaempferol as biomarkers for sunflower and rosemary honeys,26,27 there were other studies that considered those not to be adequate because there are also relatively high levels of these 2 flavonoids in melon, pumpkin, rapeseed, and cherry blossom honeys.28 Some more examples have been reported: methyl syringate and lumichrome29 present in manuka honey, considered as biomarkers, are also present in cornflower, thistle, Satureja subspicata Vis., and sage honeys.3033 Therefore, other chemical parameters have been used for helping with the identification of the botanical and/or geographical origins of monofloral honeys: metal ions, carbohydrates, amino acids, or volatile compounds.9,3437

Beyond the identification of the honey’s floral origin, some authors have also reported that phenols may also be useful for determining its geographic origin. Gambacorta et al38 showed that the phenolic profile of sulla’s (Hedysarum sp) honey, produced in 8 different areas of Southern Italy, was influenced by their geographical origins, particularly the variations in the concentrations of gallic, chlorogenic, caffeic, p-coumaric, and ferulic acids, which were dependent on each honey’s production place.

This kind of work requires an adequate number of samples, and large data sets are oftentimes difficult to be properly analysed and correctly interpreted. For this reason, several researchers have recently followed a chemometric approach to maximize information from chemical systems. For example, Karabagias et al39 differentiated Greek thyme honeys according to their geographical origin, based on physicochemical parameters and phenolic profile, using multivariate analysis of variance and linear discriminant analysis. Other good example is that of Zhao et al40 that using principal components analysis and discriminant analysis selected common chromatography peak areas of phenolic acids, allowing them to correctly classify more than 85% of the honey samples from 18 different areas of China. After analysing several phenolic profiles and total phenolic contents, the composition of minerals, sugars, and antioxidant activity of 18 unifloral Salvia officinalis honey samples, by applying principal components analysis to the data, they could accurately certify those honeys.37

Most of the biological properties are attributed to the phenol components and, to a lesser extent, to some volatile compounds. However, other biophysical factors may also be involved: the glucose oxidase system (forms hydrogen peroxide), high osmotic pressure, acidity, low redox potential, high carbon to nitrogen ratio, bee defensin-1, and viscosity.34,41

Antioxidant Activity

In a recent review,42 it became evident that honey possesses antioxidant activity, both in vitro and in vivo, independently from its floral or geographical origin. The in vitro assays show that honey is able to scavenge free radicals (1,1-diphenyl-2-picrylhydrazyl, peroxyl radicals [oxygen radical absorbance capacity], 2,2′-azino-bis [3-ethylbenzothiazoline-6-sulphonic acid], nitric oxide [NO]), as well as reduce ferric cations, chelate metal ions, inhibit lipid peroxidation measured by thiobarbituric acid reactive substances, and inhibit β-carotene bleaching.4246

In addition, the in vivo studies reveal that honey is able to stimulate the antioxidant defence system in the tissues of mice and rats (namely, pancreas, serum, kidney, and liver), particularly enhancing the activities of cellular antioxidant enzymes, such as superoxide dismutase, CAT, glutathione peroxidase, and glutathione S-transferase, and increasing the levels of reduced glutathione.42

A strong correlation between the antioxidant activity of honeys and total phenolics (benzoic acids and their esters, cinnamic acids and their esters, and flavonoids and flavonoid aglycones) has been detected.42 However, the antioxidant activity must be considered as the result of a combined effect of several compounds present in honeys, which depend on floral and geographical origins, among other factors.4548

In the presence of oxidizing components (eg, hydrogen peroxide or transition metal ions), polyphenols may covalently bind to proteins after polyphenol auto-oxidation in the presence of O2. This bond causes a decrease in either antioxidant activity or pro-oxidant activity.49 According to Rice-Evans et al,50 this loss of activity can be attributed to the chemical structure of phenols, particularly flavonoids, which present a catechol group on the B-ring, the 2,3-double bond in conjugation with a acarbonyl group at position 4 in the C-ring, and, finally, the presence of hydroxyl groups at positions 3 and 5 of the A-ring (eg, quercetin, myricetin, and quercetagetin). These chemical features also explain the antioxidant activity of flavonoids.51 In essence, the antioxidant or pro-oxidant activities may be attributed to the same flavonoids. The presence of hydrogen peroxide, metal ions, or oxygen, which can react with flavonoids, may direct them towards a pro-oxidant role.49,50

The type of storage of the honey and/or its heat treatment increases the interaction between proteins and polyphenols, hence allowing the production of 2 types of complexes with distinct antioxidant power: the complexes with higher antioxidant activity are enriched for proteins with higher molecular weight, whereas the other complexes, enriched in polyphenols with lower molecular size, have lower antioxidant activity.52

When honey is stored over long periods of time, or when submitted to heating, high-molecular-weight sugar-derived protein adducts are formed, with appear brown, known as melanoidins. In honey, polyphenols can also be included into melanoidins.5355 The incorporation of polyphenols into these brown pigments may result in a decrease in its antioxidant activity54,56; although in some cases such inclusions were responsible for either an increase or a decrease in the antioxidant activity,55 depending on the colour of the honey. When heated, light and medium honeys gained antioxidant activity, in contrast to the dark ones, which show reduced antioxidant activity in response to heating.55

Antioxidant properties have also been attributed to gluconic, malic, and citric acids, which act by chelating metal ions and increasing the effect of the antioxidant activity of flavonoids. Moreover, glucose oxidase and CAT are also reported to display antioxidant activity,3 as well as other compounds commonly found in honey, such as some of its mineral components,45,57,58 proteins,57 and even some amino acids, including proline57,59; although some studies have shown no direct correlation between antioxidant activity and the levels of proline.45,46

The antioxidant activity of monofloral honeys from diverse geographical origins has hardly been studied, particularly regarding its capacity for scavenging free radicals (in vitro assays), which have recently been compiled.14

Due to its antioxidant activity, honey has been suggested as an alternative to the sulphites used to inhibit the action of polyphenol oxidases, which in the presence of oxygen give rise to undesirable brown pigments in fresh-cut fruits and vegetables in the fruit juice processing industry.3,60,61 The main advantage of using honey instead of sulphites is the prevention of potential asthma attacks or anaphylactic reactions induced by sulphites in susceptible individuals.3

Oxidative stress and inflammation are closely interrelated, and therefore, the antioxidant effect of honey may also contribute to its anti-inflammatory activity.42

Immunomodulatory Activity of Honey in Wound Healing

The process of wound healing is generally divided into 4 overlapping stages: haemostasis (seconds to minutes), inflammation (3-5 days), proliferation (4-14 days), and remodelling (8 days to 1 year). After haemostasis, and during inflammation, debris and bacteria are removed, followed by the proliferative stage, which is characterized by blood vessel invasion and regeneration of the connective tissue and epithelium, and finally, wound contraction. In the remodelling stage, collagen is rearranged, and excess tissue is removed by apoptosis.62 After the haemostasis stage, the tissue regeneration during wound healing starts with an immunostimulatory or inflammatory action as a defensive response to trauma and/or any pathogenic agents.4 However, in the presence of an unregulated inflammation, and particularly if this stage persists, a full recovery does not occur, giving rise to a chronic wound.4

Honey has been reported to display a dual role: (1) immunostimulatory or inflammatory action (stimulates the production of immunological mediators, such as tumour necrosis factor α [TNF-α], interleukin [IL]-1β, and IL-6, and induces the upregulation of prostaglandin E2 and cyclooxygenase-2 [COX-2]),6369 and (2) anti-inflammatory action (suppresses the production of certain molecules, such as matrix metalloproteinases [MMPs], reactive oxygen intermediates, and reactive oxygen species [ROS], inhibits the expression of TNF-α and COX-2, attenuates the activation of nuclear factor-κB [NF-κB], or reduces NO production).4,70 These properties have all been correlated with the floral origin of each honey.70,71

Over time, honey has frequently been used in the treatment of wounds and/or skin disorders (eg, eczema, dermatitis, burns, ulcers, and Fournier gangrene), not only for its antimicrobial abilities but also due to its other reported properties, such as the immunostimulatory or inflammatory activities, promotion of autolytic debridement, stimulation of wounded tissue growth to hasten healing and promote the healing process in dormant wounds, as well as its described anti-inflammatory features.62,63,7072

Table 2 presents some references describing the immunostimulatory properties of honeys of different botanical and/or geographical origins. Despite these reports, in some cases, an anti-inflammatory activity was also observed, inhibiting the release of some factors (cytokines, matrix metallopeptidase 9 [MMP-9], or ROS), either from immune or cutaneous cells, depending on wound conditions (acute or chronic inflammation), concentration, and type of honey.7376 It has been hypothesized that at low concentrations of inflammatory/stimulatory mediators, honey exerts a pro-inflammatory role via the stimulation of production of inflammatory cytokines and MMP-9. Nevertheless, when the inflammation is already in progress, and in the presence of an infection, honey might act as an anti-inflammatory compound, inhibiting the production of inflammatory cytokines and MMP-9.76

Table 2.

Examples of immunostimulatory properties shown in vitro for honeys of diverse botanical and/or geographical origins.

Honey (botanical/geographical origins)Type of cellsEffectsResponsible compounds (hypothesis)Reference
Not reported/JordanPeripheral blood B cells
Peripheral blood T cells
↑ (either in the presence or absence of mitogens LPS, PHA, and Con A)
↑ (either in the presence or absence of mitogens LPS, PHA, and Con A)
Unidentified lymphomitogensAbuharfeil et al77
Peroxide-generating honey from mixed floral source (pasture PS9)/not reportedThey are not used↑ production of hydroxyl radicals in the presence of Fe2+ and other radical speciesProduction of other radical species may be due to secondary reactions between hydroxyl radicals and organic compounds present in the honeyHenriques et al73
Manuka honey with high levels of methylglyoxal (MGO250)/New ZealandNeutrophils↑ TNF-α only at lower concentrations (100 mg/mL)Not referredChepulis and Francis74
Acacia/SlovakiaPrimary keratinocytesUpregulation of IL-1β, TGF-β, TNF-α, and MMP-9 mRNA expressionpH, hydrogen peroxide release and additional componentsMajtan et al67
Acacia, buckwheat, manuka (UMF 15+)/Japan
Buckwheat/Japan
Buckwheat, acacia/Japan
Manuka/Japan
HaCaT cells (human skin keratinocytes)
HaCaT cells
HaCaT cells
HaCaT cells
Stimulation of MMP-9 expression
Induction of syndecan-4
Upregulation of vimentin, HPRT-1, and STEAP-1
Upregulation of HPRT-1
Not referredRanzato et al75
Acacia, buckwheat/JapanHuman fibroblast cell line (46 BR.1N)↑ release of IL-4, IL-6, and IL-8Not referredRanzato et al78
Manuka honey (M61) and a mixed pasture honey (PS3)Human monocytic cell line (MM6)Induction of TNF-α releaseGlycosylated proteins may affect MM6 activationTonks et al64
Manuka honey (airborne f/d), pasture honey (Lorimers)/New Zealand; Jelly Bush Honey/Austr. bushHuman monocytic cell line MM6 and human monocytes isolated from peripheral blood↑ TNF-α, IL-1β, and IL-6 releaseComponent(s) that mediated the effects were unknownTonks et al65
Manuka honey/New ZealandMono Mac 6 cells (MM6) or murine bone marrow–derived macrophages from wild-type C57BL/6 mice or TLR2 or TLR4 knockout miceStimulation of the production of inflammatory cytokines TNF-α, IL-1β, or IL-6Active component(s) of 5 to 6 kDa present in honey signal through TLR4 but not TLR2Tonks et al66
Manuka honey UMF 16+/New Zealand), active 5+ Manuka honey/New Zealand), Danish honeyHuman monocytic cell line Mono Mac 6
Human promyelocytic leukaemia cells HL-60
Induction of interleukin-6 release
Release of reactive oxygen species
Heat stable substance with high molecular weight (>20 kDa)Timm et al79
Kanuka (Kunzea ericoides), manuka (Leptospermum scoparium), and clover (Trifolium spp) honeys/New Zealand
AGPs from kanuka honey
Human monocytic cell lines THP-1 and lymphoblast lung from human and U937All 3 honeys stimulated TNF-α release from THP-1 cells, with kanuka honey being the most active
AGPs purified from kanuka honey stimulated the release of TNF-α from THP-1 and U937 cells
Immunostimulatory properties may be attributed to their particular content of LPS, apalbumins, and AGPsGannabathula et al68
Thyme honey/FranceRaw 264.7 murine macrophagesIncrease in PGE2 production and overexpression of both COX-2 and TNF-α: overexpression and activation of the AP-1 and NF-κB transcription factor subunitsThe induction of these pathways may depend on some unidentified components of thyme honeyRaynaud et al69

Abbreviations: AGPs, arabinogalactans; AP-1, activator protein 1; Con A, concanavalin A; COX-2, cyclooxygenase 2; HPRT-1, hypoxanthine phosphoribosyl transferase 1; IL, interleukin; LPS, lipopolysaccharide; MMP-9, matrix metallopeptidase 9; mRNA, messenger RNA; NF-κB, nuclear factor κB; PHA, phytohaemagglutinin; STEAP-1, six-transmembrane epithelial antigen of the prostate 1; TNF-α, tumour necrosis factor; TLR4, toll-like receptor; UMF, Unique Manuka Factor; ↑, increase; ↓, reduce.

The assignment of immunostimulatory abilities to the respective compounds present in honey has been controversial. Although Timm et al attributed the induction of cytokines to the lipopolysaccharide (LPS) from contaminating bacteria present in honey,79 other authors consider that LPS has no role in honey’s immunostimulatory activity.66,67,69 Instead, other constituents are responsible for such feature, namely, type II arabinogalactans,68 methylglyoxal (MGO),74 and the major royal jelly protein 1 (MRJP1).67 Nevertheless, it is undisputed that the immunostimulatory activity of honey in wound healing is influenced by the pH and the release of hydrogen peroxide.72,80

This anti-inflammatory activity has been studied (1) in rabbits injected with LPS,81 (2) in the AGS cell line derived from a human gastric adenocarcinoma with Helicobacter pylori,82 (3) in rats with experimentally induced inflammatory bowel disease,83 (4) in alkali injury on rabbits’ eyes,84 and (5) in rats with ethanol-induced gastric ulcers,85 and several other examples previously compiled.4

More recently, considering only 2016, several authors reported in vitro and in vivo assays showing the anti-inflammatory activity of honey. For example, lavender honey shows better in vitro antihyaluronidase activity than citrus or strawberry tree honey, whereas the latter 2 perform better in inhibiting the lipoperoxidase activity.86 The ability for inhibiting hyaluronidase in vitro has also been previously reported,87 where oak, chestnut, and heather honeys displayed the best activity. The honey-induced inhibition of this enzyme was enhanced in combination with propolis.88 In vivo testing with laboratory animals has shown a gastroprotective effect of manuka honey against ethanol-induced gastric ulcers in rats by decreasing the concentration of plasma inflammatory cytokines such as TNF-α, IL-1β, and IL-6.85 Moreover, multifloral honey was shown to downregulate the oxidative and inflammatory markers (decreased levels of tissue TNF-α and inducible nitric oxide synthase) in rats with colitis induced through dextran sodium sulphate administration.89 However, other studies revealed no effect of honey when given to rats with colitis induced by acetic acid.90

Several factors have been reported to contribute to the healing of wounds promoted by the usage of honey: hydrogen peroxide, glucose oxidase, gluconic acid, MGO, and polyphenols, along with its physical properties such as hygroscopicity, hypertonicity, and lower pH.4,76,91,92 However, the anti-inflammatory activity of honey on wounds starts with microbial elimination. Because they are the main source of inflammation, honey can act as a direct anti-inflammatory compound through several alternative mechanisms.

Owing to its antimicrobial, antioxidant, and anti-inflammatory activities, honey has long been used in wound healing (burns, ulcers, and others).93 Several studies have reported the great potential of this bee product for wound healing: either applied alone94,95 on rat wounds or in association with other natural products (Aloe vera and milk or ascorbic acid), tested on burns in guinea pig and second-degree burns in rats,96,97 as chitosan nanofiber wound dressing enriched with Allium sativum and Cleome droserifolia applied on mice’s wounds.98

The benefits of honey in healing different types of human wounds have also been reported and reviewed in a recent publication,93 although other contradictory reports show no additional benefit in using honey for some wound types.99

The effectiveness of honey on wound healing has undergone some criticisms, particularly for the following reasons: poor study design, heterogeneous group of patients included in the studies, results not directly comparable between studies due to the usage of different methods and/or non-validated measurement methods, lack of appropriate blinding in the experiments, not enough study replication efforts, and usage of not so relevant comparisons which would help in making a clinical decision.93,100,101 Altogether, these factors hamper a definitive conclusion regarding the effectiveness and benefits of honey for healing wounds, independent of its floral or geographic origin.101

Anticancer Activity

The potential effects of honey on both the prevention of cancer and of tumour development and progression have been studied. Most of these studies have been performed in vitro. When tested on several types of human cancer cell lines (breast, prostate, endometrial, cervical, lung, skin, kidney, bladder, liver, oral squamous cell carcinoma, and osteosarcoma), honeys from diverse floral sources (Abies cephalonica, Acacia, Citrus, Erica arborea, forest, multifloral, Pinus sp, rosemary, thyme, and tualang tree) have demonstrated potential anticancer activity.102112 However, some in vivo studies have been conducted in rats/mice where cancer was either induced (breast, hepatic, fibrosarcoma, adenocarcinoma)112115 or transplanted (bladder, melanoma, mammary carcinoma).102,107,113

Honey potentially inhibits the development of cancer by blocking the 3 main stages of carcinogenesis: initiation, proliferation, and progression. The reported mechanisms of its antiproliferative, antimetastatic, and anticancer effects include induction of apoptosis, modulation of oxidative stress, cell cycle arrest, activation of mitochondrial pathways, induction of mitochondrial outer membrane permeabilization (MOMP), amelioration of inflammation, modulation of insulin signalling, modulation of oestrogen receptor activity, and inhibition of angiogenesis.116

Honey has been referred to as an apoptotic inducer through the upregulation and modulation of proapoptotic proteins (p53, Bax, caspase 3, and caspase 9) and downregulation of antiapoptotic proteins (B-cell lymphoma 2 protein [Bcl-2]). In addition, honey also produces ROS which will activate p53 that, in turn, modulates the expression of Bax and Bcl-2 and induces DNA fragmentation and activation of poly(adenosine diphosphate ribose) polymerase.116

Some honey proteins (royal jelly [RJ] proteins apalbumin 1 and apalbumin 2) can stimulate macrophages to release cytokines, such as TNF-α, IL-1β, and IL-6. The production of TNF-α may be a key factor in this process. This cytokine is well known for regulating many cellular processes, including apoptosis, as it binds to the TNF receptor (TNFR), the TNFR-associated death domain protein, the TNFR-associated factor, and the receptor-interacting protein, hence stimulating both caspases 3 and 8.117,118

Honey has antiproliferative activity by affecting cell cycle arrest and blocking the cell cycle of cancer cell lines (colon, glioma, and melanoma) in G0/G1, G1, and G2/M phases. The inhibition of cell proliferation induced by honey is due to the downregulation of several cellular pathways, via tyrosine cyclooxygenase, ornithine descarboxilase, and kinase.116

The antiproliferative activity of honey may also include the activation of the mitochondrial pathways and the release of proteins, such as cytochrome C, the induction of MOMP with the leakage of intermembrane space proteins moving into the cytosol causing cell death, and anti-inflammatory effect through the attenuation of pro-inflammatory mediators and inhibition of both NF-κB and mitogen-activated protein kinase signalling pathways, which contribute considerably to the reduction in the second stage of cancer development (promotion).84,116,117,119

These reported effects were dependent on the floral origin of the honey, as well as on the type of cancer cell. Along with the aforementioned RJ proteins, which are able to stimulate the release of cytokines, the anticancer/antiproliferative properties of honey may also be attributed to phenols (flavonoids and phenol acids) because it has established a significant correlation between these activities and the honey’s phenol content.103,105,120,121 In addition, a positive correlation has been found between the anticancer activity and the amount of flavonoids (chrysin, apigenin, quercetin, acacetin, and pinocembrin) and phenolic acids (p-coumaric, vanillic, protocatechuic, caffeic, and p-hydroxybenzoic acids)103,105,116,117,119122 present in honey, leading these authors to speculate about the anticancer potential of these secondary metabolites.

Metabolic and Cardiovascular Effects

Some metabolic alterations may increase the risk of cardiovascular diseases, namely, abdominal obesity, atherogenic dyslipidaemia, hypertension, insulin resistance, and glucose intolerance.122 Cardiovascular diseases are associated with chronic low-grade inflammation. Accordingly, if honey possesses anti-inflammatory activity, as discussed above, it may also play a role in the prevention of cardiovascular diseases, particularly if associated with other specific foods and dietary elements also beneficial to health, such as whole-grain foods and fresh fruit and vegetables.123 Some studies report that when children consume honey and jam at breakfast, particularly older girls, the risk of cardiovascular diseases decreases.122 Moreover, González-Gil et al123 report that moderate honey intake in European children decreases the probability of having higher concentrations of high-sensitivity C-reactive protein (CRP). Therefore, inflammation seems to be somehow associated with the type of diet.123

In a 2008 study,124 it was reported that natural honey ameliorates cardiovascular risk factors, both in healthy subjects and in patients with high risk factors. In both cases, natural honey reduced total cholesterol, low-density lipoprotein cholesterol (LDL-C), triacylglycerols, fasting blood glucose, and CRP and increased high-density lipoprotein cholesterol (HDL-C), with the advantage of not increasing body weight, particularly in overweight or obese individuals.

However, the proclaimed effects of honey on metabolic and cardiovascular systems are still controversial. For example, in healthy humans, Al-Waili125 observed that honey reduced cholesterol, LDL-C, triglycerides (TG), homocysteine, and CRP and slightly elevated HDL-C. In addition, they observed that in patients with hypertriglyceridaemia, honey decreased TG; in patients with hyperlipidaemia, honey decreased LDL-C, and in patients with diabetes, honey decreased the levels of glucose in the plasma. Such observations support the positive role of honey in the prevention of cardiovascular diseases. Conversely, other publications show that honey consumption increases the levels of glycosylated haemoglobin in patients with diabetes, suggesting that it does not help with the glycaemic control in these patients.126 These findings were further investigated by a different group of authors127 who argue that those results pertain to the administration of high doses of honey. In fact, this publication shows that low doses of honey, irrespective of its geographical origin,128,129 improve hyperglycaemia and dyslipidaemia in alloxan-induced diabetic rats. These properties are most likely derived from the fructose and other oligosaccharides present in honey.129,130 In fact, glucose and fructose have a synergistic effect in the gastrointestinal tract by enhancing intestinal fructose absorption and/or stimulation of secretion of insulin. At the same time, fructose improves the hepatic glucose uptake and glycogen synthesis and storage, activating the hepatic glucokinase and glycogen synthase.129

It has been proposed that honey mimics some characteristic effects of diverse enzyme inhibitors, namely, α-glucosidase inhibitors, insulin secretagogues, thiazolidinediones and biguanides, dipeptidyl peptidase-4 inhibitors, and anti-obesity drugs.131 The putative antidiabetic benefits of honey have been demonstrated in vivo using laboratory model organisms. However, in humans, the studies demonstrating the beneficial effects of this natural bee product, particularly regarding the lowering of glucose in the blood, are still insufficient to draw reliable conclusions, mainly because the assays lasted only a few weeks (8-12 weeks). Because diabetes is a chronic disorder, such a limited amount of time is not enough for categorical answers regarding the antidiabetic potential of honey.132 Additional constraints that hamper the objective study of honey’s antidiabetic effect are the small size of the samples, absence of randomized clinical trials where patients with diabetes are grouped by disease severity (mild, moderate, and severe), treatment with different dosages of honey, as well as the absence of continued clinical studies concerning the short-term and long-term effects of honey consumption in patients with diabetes.131

Hypertension is another important public health disease, not only because of its high prevalence but also because of its another critical risk factor for the development of cardiovascular diseases.133 Some authors demonstrated that honey was able to reduce elevated systolic blood pressure in spontaneously hypertensive rats. At the same time, the honey supplement was also able to improve the oxidative stress observed in the kidneys of those experimental animals through the decrease in malondialdehyde levels.134

The metabolic and cardiovascular effects of honey are highly dependent on its chemical composition, akin to its other biological effects. Quercetin and kaempferol (flavonoids present in honey, whose concentration depends on its floral/geographical origin) and their metabolites may have a protective effect on cardiovascular disease via several reported mechanisms: (1) activation of Ca2+-activated K+ channels and increasing the activity of endothelial NO synthase with stimulation of arterial relaxation, (2) protection of vessels against hypochlorous acid–induced endothelial dysfunction in isolated arteries by the activation of adenosine monophosphate protein kinase and subsequent increase in NO production, and (3) suppression of TNF-α production as well as activation of NF-κB, with the consequent inhibitory activity on cell adhesion and migration to endothelial cells.59,135139 These properties attributed to flavonoids and their derivatives have mostly been demonstrated in cell cultures or in laboratory animals. However, other compounds alone or, most likely, in association may also play a decisive role in the achievement of the beneficial properties attributed to honey.

Prebiotic Properties

Prebiotics are non-digestible food ingredients with functional effects on the gastrointestinal tract, which are usually short-chain carbohydrate compounds, primarily oligosaccharides. They stimulate the growth of bifidobacteria and lactobacilli populations in the colon, being selectively fermented by them. These events increase the production of short-chain fatty acids, decrease the pH, and reduce the fat absorption and ammonia production, overall enhancing the host’s health.140,141

Fructooligosaccharides, galactooligosaccharides, and inulin, which are present in some natural foods, are examples of prebiotics,142,143 whose degree of polymerization and type of oligomer, which differ from food to food, are responsible for much of their effectiveness.140 Honey, having circa 0.75% of fructooligosaccharides in its constitution,144,145 may act as a prebiotic. Moreover, the detrimental effects of bile salts on Bifidobacterium spp, a ubiquitous inhabitant of the human gastrointestinal tract, can be overcome by the action of fructooligosaccharides and their monomeric derivatives.146 The prebiotic effect of honey has been observed in several monofloral honeys: sourwood, alfalfa and sage honey,147 honeydew honey,148 chestnut and acacia honey,149 clover,150 and eucalyptus,151 each with strengths highly dependent on its floral origin.147,149 In addition, some authors observed that the prebiotic activity of honey was not comparable with the one demonstrated by inulin or fructooligosaccharides, being substantially inferior,141,148 acting preferentially on Bifidobacterium,152 or otherwise.148,153

Control of Human Pathogens With Honey

Since ancient times, honey has been used to control infections. Nowadays, this practice has seen a renewed interest in virtue of the emergence of antibiotic-resistant bacterial pathogens, which increase the risk of simple medical interventions. The antibacterial features of honey have been linked to different factors, including the amount of hydrogen peroxide (H2O2), low pH, high osmolarity, phenolic and aromatic acid content, honey glycoproteins, as well as products of the Maillard reaction (this non-enzymatic browning reaction involves the interaction between reducing carbohydrates and amino acids or proteins).154158 A recent publication demonstrated the antibacterial activity of the MRJP1 honey glycoprotein against bacterial clinical isolates.157 However, these findings were not reproduced by the subsequent study conducted by Bucekova and Majtan159 that tested the action of the purified MRJP1 honey glycoprotein isolated from 3 honey types, which are used in wound healing (manuka honey, Revamil source honey, and honeydew honey). The discordant results between the 2 studies may, however, be related to the differences in the methodology used to isolate and purify the honey’s glycoproteins.

The antibacterial activity of the different honeys has been shown to be linked to its geographical origin, botanical origin, season of harvest, and conditions of processing and storage.160,161 Despite the recognition of the valuable action of honey as an antibacterial agent, its mechanism of action against pathogens is still poorly understood. However, there is an increase in the number of studies aiming at investigating the interactions between honey and bacterial cells, particularly with manuka honey, which is one of the most effective honeys.161166 Those studies show that, depending of the bacterial species, the honey’s action influences the bacterial size (shorter or longer cells), morphology, cell division (incomplete division), surface (irregular), motility, injury to nucleic acids, and lysis.

Bacterial pathogens in nature typically live in biofilms, ie, adherent to biotic or abiotic surfaces. Of particular concern for the human health is the fact that bacteria adhere to medical devices and colonize the mucosal membranes, as well as the skin and open wounds. The ability to adhere and maintain a cohesive biofilm structure is mainly regulated by a cellular communication process denominated quorum sensing (QS). Several studies have explored the potential of honey for the impairment of the natural ability of bacteria to adhere and form a biofilm, disrupt an already established one, and even inhibit the QS process.166171

Disruption of QS and biofilm

Quorum sensing is a cell-cell communication system that involves the production of signalling molecules, such as N-acyl-l-homoserine lactones (AHLs), which control cellular processes linked to population density, including biofilm formation and virulence.172,173

In the study of Truchado et al,167 chestnut honey (Castanea sativa) and its aqueous extract inhibited the QS of 2 common biosensor bacteria, namely, Chromobacterium violaceum CV026 and Agrobacterium tumefaciens NTJ, and its QS inhibitory ability was additionally confirmed in 3 relevant pathogens: Erwinia carotovora, Yersinia enterocolitica, and Aeromonas hydrophila. The QS inhibitory features of chestnut honey were associated with both inhibition of AHL production and its degradation. Simultaneously, pathogen growth was not affected, but biofilm formation, which is monitored by QS, was impaired.

The QS inhibitory potential of manuka honey was identified either via the testing of the QS system in Pseudomonas aeruginosa65 or by evaluating the biosensor of C violaceum.171 Both QS systems are crucial for these pathogens to overcome environmental stresses and adequately adjust their virulence state. Both manuka and Italian honeys interfere with the 2 QS systems of P aeruginosa. Interestingly, the authors show that the QS inhibition was associated with the honey’s sugar content because the expression of the pqsA gene was nearly 50% reduced at a concentration of 1% of honey (both manuka and Italian). Moreover, the bacterial growth was not inhibited for up to 6% concentrations of either manuka or Italian honey.169 The effect of honey’s sugar content on the inhibition of QS has also been reported by Lee et al,168 who tested the effect of diluted honey (a monofloral [acacia] and a polyfloral Korean honey) on the QS system of the enterohemorrhagic Escherichia coli 0157:H7. The authors suggested that certain sugar concentrations might block E coli’s AHLs from entering the cell and for that reason prevent AHL ligation to its receptor.

Slovakian honey (hawthorn [Crataegus laevigata], honeydew [Abies alba Mill], and acacia [Robinia pseudoacacia]) and also manuka honey could prevent biofilm formation by the wound-associated bacteria Proteus mirabilis and Enterobacter cloacae. At 50%, these honeys display the potential to induce the detachment of bacterial cells from aggregates.174 However, none of the tested honeys has shown the ability to break up E cloacae’s biofilm.174 Another interesting study, using a modified Lubbock chronic wound biofilm system, examined the effect of honey and a recombinant form of the honey defensin 1 (Def-1) against a multispecies biofilm (Staphylococcus aureus, Streptococcus agalactiae, P aeruginosa, and Enterococcus faecalis).175,176 These authors found that the pathogens S aureus, S agalactiae, and P aeruginosa were indeed perturbed by the tested honeys (manuka and honeydew). Defensin 1 affected S aureus and P aeruginosa but not S agalactiae. Interestingly, E faecalis sessile cells were resistant to both honey and Def-1.

The anti-biofilm activity of honey has been associated with different substances, either alone or in combination, namely, with sugars, phenols, hydrogen peroxide, dicarbonyl methylglyoxal, and even its low pH and high osmolarity.169,175

The effect of honey on biofilm inhibition can be improved if honey is associated with antibiotics. Campeau and Patel177 tested manuka honey together with vancomycin, showing a synergistic activity of this association against the biofilms of S aureus (the minimum biofilm eradication concentration [MBEC] of manuka honey used alone was 3% and with 1 µg/mL vancomycin was 1%), whereas the association of manuka with gentamycin showed an additive activity against P aeruginosa biofilm (the MBEC value of manuka honey used alone was 30%, with 75 µg/mL of gentamycin was 20%, and when associated with 100 µg/mL of gentamycin dropped to 17.5%).

Antivirulence action

Antivirulence agents (AVAs) impair virulence factors (VFs) by negatively affecting their production/activity, constituting a supplementary or alternative approach to combat the emerging resistant pathogens. This activity is associated with the fact that AVAs do not disturb the bacterial growth, hence not exerting the environmental pressure that classical antimicrobials do, which lead to a positive selection of the genetic mutations that allow the resistant cells to subsist. Honey offers the possibility of acting as a natural anti-VF. Inside the host, some pathogens experience iron deficiency, and to overcome this, they depend on siderophores to acquire iron. As such, inhibition of siderophore production constitutes an alternative means of controlling pathogens. In the study by Kronda et al,178 it was observed that manuka honey impairs P aeruginosa’s virulence by interfering with siderophore production.

Using a combined proteomics and transcriptomics approach, Jenkins et al170 observed that manuka honey affected the expression of VFs of methicillin-resistant Staphylococcus aureus (MRSA), namely, the expression of the virulence genes sec3, fnb, hlgA, lip, and hla. However, its real impact in vivo was not demonstrated.

The number of case reports about the usage of honey on wounds and ulcers resilient to healing is very limited.179,180 However, the published data pointed out to a considerable enhancement of infection clearing after unsuccessful antibiotic treatment.179,180

Using animal models to study infection and wound healing is not consensual. Nevertheless, the impact of honey on the virulence potential should be clearly evaluated using, eg, insect models, such as the greater wax moth (Galleria mellonella), whose innate immune response shows significant similarity with the vertebrate immune response.181 In a recent study, da Silva et al86 have shown that Portuguese honeys were able to impair the virulence potential of S aureus and MRSA strains, using the G mellonella insect model. The potential action of honey against S aureus is timely and pertinent as this pathogen is one of the main sources of infection associated with health care institutions.182

The usage of honey in combination with other antimicrobial agents

The usage of honey in combination with other antimicrobial agents has been the subject of some investigations.183185 Combining honey with other antimicrobial agents diminishes the chances of pathogen survival after exposure, consequently reducing the likelihood of emergence of resistant bacteria. The usage of manuka honey, in combination with the antibiotics tobramycin and colistin, to combat P aeruginosa and Burkholderia cepacia infections resulted in synergistic or additive effects against the 2 strains of each bacterial species.184

Furthermore, the combination of manuka honey with rifampicin, oxacillin, gentamycin, and clindamycin was tested against different MRSA strains and methicillin-susceptible Staphylococcus aureus clinical isolates, finding that, depending on the strain, it yielded synergistic or additive effects or even just increased the susceptibility of the bacterial isolates to the antibiotic.185

As mentioned above, the combination of honey with antibiotics has been shown to improve the inhibition of biofilm formation.178

An interesting report demonstrated that the action of 2 honeys collected from the stingless bees Scaptotrigona bipunctata (Lepeletier, 1836) and Scaptotrigona postica (Latreille, 1807), when in combination, shows a synergistic type of action against several bacterial pathogens, including multiresistant bacteria.186 It is possible that such synergistic action arises from the combination of the constituents of the 2 honeys. Other studies have also reported synergistic effects between bee products, particularly the combination of honey with propolis, against individual or polybacterial resistant pathogens.187 Another intriguing synergistic effect has been found with the combination of honey with potato starch.188 This combination was shown to be very effective against the bacterium Klebsiella pneumoniae and was correlated with the content of fructose, glucose, and diastase in the mixture.

Antiviral Activity of Honey

Studies examining the antiviral activity of honey are limited. The manuka honey, one of the best studied honey types, revealed to be effective against influenza virus in vitro, using Madin-Darby canine kidney cells.189 Manuka and clover honey were also effective in vitro against varicella zoster virus using human malignant melanoma cells (MeWo).190 Furthermore, commercial and manuka honey seem to act against herpes simplex virus (HSV-1) isolates in vitro (using Vero cells),191,192 and honey solutions act against rubella virus also in vitro (using monkey kidney cell cultures).193 Al-Waili194 showed that topical honey application was effective in healing labial and genital recurrent herpes lesions. The report discusses that the amelioration of symptoms and signs of herpetic lesions by honey might be attributed to the inhibition of prostaglandins at the lesion site.

In these studies, the antiviral activity has not been attributed to any particular compound, or group of compounds, present in honey. Viuda-Martos et al,3 however, consider that flavonoids may have an important role for this activity because galangin has been proven to be effective against HSV and coxsackie B virus, whereas quercetin and rutin show antiviral activity against HSV, syncytial virus, poliovirus, and Sindbis virus. Although these flavonoids may be found in honey, an unequivocal connection between the antiviral properties of honey and its chemical composition has not yet been formally established.

Disadvantages

Despite the limited evidence performed in the clinical environment to thoroughly compare the risk/benefits of using honey versus the conventional treatments, it is undisputed that honey is more and more used, not only as a food source but also for alleviating particular ailments.

Regardless of its usage as food or remedy, honey may be contaminated by pesticides, antibiotics, heavy metals, and other toxic compounds. The presence of such compounds may be attributed not only to accidental exposure and environmental hazards but also to compounds added by beekeepers to control honeybee diseases.195 However, these contaminants may be harmful for human health, producing unexpected consequences.

In addition to those chemical compounds, honey may also be contaminated with pathogens, particularly Clostridium botulinum and its spores. As such, the consumption of honey or its derivatives is considered to be dangerous for infants, the elderly, and immunocompromised individuals.196 For this reason, honey used for therapeutic purposes should be sterilized using gamma irradiation.195

For external usage, honey may also present some disadvantages or adverse effects: preparation of impregnated dressings may be difficult; it becomes more fluid at high temperatures, potentially liquefying at wound temperature; transient stinging sensation may occur when honey is applied topically; topical application in large wounded areas of patients with diabetes may increase the concentration of glucose in the blood up to dangerous levels; excessive application of honey may lead to tissue dehydration; and allergic reaction to pollen or to bee proteins in the honey may occur.197199

Conclusions

Despite the abundant potentially bioactive properties reported above, a comprehensive overview of the mechanisms underlying honey’s health benefits is still under active scrutiny. The studies conducted so far support that each honey’s activity is strongly dependent on its floral or geographic origin. The most investigated properties of honey for its potential usage as an alternative medicine include (1) its antioxidant activity, (2) its ability to induce anti-inflammatory and immunomodulatory responses, (3) delaying cancer development and cardiovascular diseases, (4) inhibiting bacterial pathogens by interfering with their pathogenesis potential, (5) its ability to control viral infection, and, finally, (6) improving the health of the gut by acting as a prebiotic agent.

The observed synergistic actions between honey and some antibiotics highlight the need for the adjustment of antibiotic dosage for those patients who are using honey as a natural supplement. Exploring the synergistic effects of honey with other bee products, or vegetable products, seems also to be another promising venue of investigation. In addition, pinpointing the honey’s chemical constituents responsible for its reported antiviral properties is also a remarkable feature that requires further research. In addition, there is a pressing need to chemically characterize the different types of honey, particularly regarding their geographical and floral origin, to create a catalogue of beneficial compounds that are present in each honey type, allowing the end users to choose the type of honey that best fits their needs.

Altogether, there are various pieces of evidence reported in numerous comprehensive studies, clearly demonstrating that honey constitutes an efficient nutraceutical agent, potentially operating on several vital health systems.

Acknowledgments

The authors are particularly grateful to Isabel Duarte from CBMR for the helpful comments, suggestions, and English language revision.

Footnotes

Peer review:Five peer reviewers contributed to the peer review report. Reviewers’ reports totalled 897 words, excluding any confidential comments to the academic editor.

Funding:The author(s) received no financial support for the research, authorship, and/or publication of this article.

Declaration of conflicting interests:The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Contributed by

Author Contributions: Conceived the concept: MGM, MDA, MLF. Wrote the first draft of the manuscript: MGM, MDA, MLF. Contributed to the writing of the manuscript: MGM, MDA, MLF. Agreed with manuscript results and conclusions: MGM, MDA, MLF. Made critical revisions and approved the final version: MGM, MDA, MLF. All authors reviewed and approved the final manuscript.

References

1. Alvarez-Suarez JM, Gasparrini M, Forbes-Hernández TY, Mazzoni L, Giampieri F. The composition and biological activity of honey: a focus on Manuka honey. Foods. 2014;3:420–432. [PMC free article] [PubMed] [Google Scholar]
2. Bogdanov S. Functional and biological properties of the bee products: a review. www.bee-hexagon.net. Bee Product Science; Published February 1, 2011. [Google Scholar]
3. Viuda-Martos M, Navajas Y, Fernández-López J, Pérez-Álvarez JA. Functional properties of honey, propolis, and royal jelly. J Food Sci. 2008;73:R117–R124. [PubMed] [Google Scholar]
4. Hadagali MD, Chua LS. The anti-inflammatory and wound healing properties of honey. Eur Food Res Technol. 2014;239:1003–1014. [Google Scholar]
5. da Silva PM, Gauche C, Gonzaga LV, Costa ACO, Fett R. Honey: chemical composition, stability and authenticity. Food Chem. 2016;196:309–323. [PubMed] [Google Scholar]
6. Bogdanov S, Jurendic T, Sieber R, Gallmann P. Honey for nutrition and health: a review. Am J Col Nutr. 2008;27:677–689. [PubMed] [Google Scholar]
7. Persano Oddo L, Piro R. Main European unifloral honeys: descriptive sheets. Apidologie. 2004;35:S38–S81. [Google Scholar]
8. Escuredo O, Dobre I, Fernández-González M, Seijo MC. Contribution of botanical origin and sugar composition of honeys on the crystallization phenomenon. Food Chem. 2014;149:84–90. [PubMed] [Google Scholar]
9. Kaškonienė V, Venskutonis PR. Floral markers in honey of various botanical and geographic origins: a review. Compr Rev Food Sci Food Saf. 2010;9:620–634. doi: 10.1111/j.1541-4337.2010.00130.x. [PubMed] [CrossRef] [Google Scholar]
10. Alissandrakis E, Tarantilis PA, Harizanis PC, Polissiou M. Aroma investigation of unifloral Greek citrus honey using solid-phase microextraction coupled to gas chromatographic-mass spectrometric analysis. Food Chem. 2007;100:396–404. [Google Scholar]
11. Castro-Vázquez L, Díaz-Maroto MC, Pérez-Coello MS. Aroma composition and new chemical markers of Spanish citrus honeys. Food Chem. 2007;103:601–606. [Google Scholar]
12. Petretto GL, Tuberoso CI, Vlahopoulou G, et al. Volatiles, color characteristics and other physico-chemical parameters of commercial Moroccan honeys. Nat Prod Res. 2016;30:286–292. [PubMed] [Google Scholar]
13. Oroian M, Amariri S, Rosu A, Gutt G. Classification of unifloral honey using multivariate analysis. J Essent Oil Res. 2015;27:533–544. [Google Scholar]
14. Ciulu M, Spano N, Pilo MI, Sanna G. Recent advances in the analysis of phenolic compounds in unifloral honeys. Molecules. 2016;24:451. doi: 10.3390/molecules21040451. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
15. Ferreres F, Andrade P, Gil MI, Tomás-Barberán FA. Floral nectar phenolics as biochemical markers for the botanical origin of heather honey. Z Lebensm Unters Forsch. 1996;202:40–44. [Google Scholar]
16. Andrade P, Ferreres F, Amaral MT. Analysis of honey phenolic acids by HPLC, its application to honey botanical characterization. J Liq Chromatogr Relat Technol. 1997;20:2281–2288. [Google Scholar]
17. Andrade P, Ferreres F, Gil MI, Tomas-Barberan FA. Determination of phenolic compounds in honeys with different floral origin by capillary zone electrophoresis. Food Chem. 1997;60:79–84. [Google Scholar]
18. Dimitrova B, Gevrenova R, Anklam E. Analysis of phenolic acids in honeys of different floral origin by solid-phase extraction and high-performance liquid chromatography. Phytochem Anal. 2007;18:24–32. [PubMed] [Google Scholar]
19. Martos I, Ferreres F, Tomás-Barberán FA. Identification of flavonoid markers for the botanical origin of Eucalyptus honey. J Agric Food Chem. 2000;48:1498–1502. [PubMed] [Google Scholar]
20. Truchado P, Ferreres F, Tomas-Barberan FA. Liquid chromatography-tandem mass spectrometry reveals the widespread occurrence of flavonoid glycosides in honey, and their potential as floral origin markers. J Chromatogr A. 2009;1216:7241–7248. [PubMed] [Google Scholar]
21. Yao L, Jiang Y, D’Arcy B, et al. Quantitative high-performance liquid chromatography analyses of flavonoids in Australian Eucalyptus honeys. J Agric Food Chem. 2004;52:210–214. [PubMed] [Google Scholar]
22. Cabras P, Angioni A, Tuberoso C, et al. Homogentisic acid: a phenolic acid as a marker of strawberry-tree (Arbutus unedo) honey. J Agric Food Chem. 1999;47:4064–4067. [PubMed] [Google Scholar]
23. Tuberoso CIG, Bifulco E, Caboni P, Cottiglia F, Cabras P, Floris I. Floral markers of strawberry tree (Arbutus unedo L.) honey. J Agric Food Chem. 2010;58:384–389. [PubMed] [Google Scholar]
24. Stephens JM, Schlothauer RC, Morris BD, et al. Phenolic compounds and methylglyoxal in some New Zealand manuka and kanuka honeys. Food Chem. 2010;120:78–86. [Google Scholar]
25. Tuberoso CIG, Bifulco E, Jerković I, Caboni P, Cabras P, Floris I. Methyl syringate: a chemical marker of asphodel (Asphodelus microcarpus Salzm. et Viv.) monofloral honey. J Agric Food Chem. 2009;57:3895–3900. [PubMed] [Google Scholar]
26. Tomás-Barberán FA, Martos I, Ferreres F, Radovic BS, Auklam E. HPLC flavonoid profiles as markers for the botanical origin of European unifloral honeys. J Sci Food Agric. 2001;81:485–496. [Google Scholar]
27. Martos I, Cossentini M, Ferreres F, Tomás-Barberán FA. Flavonoid composition of Tunisian honeys and propolis. J Agric Food Chem. 1997;45:2824–2829. [Google Scholar]
28. Petrus K, Schwartz H, Sontag G. Analysis of flavonoids in honey by HPLC coupled with coulometric electrode array detection and electrospray ionization mass spectrometry. Anal Bioanal Chem. 2011;400:2555–2563. [PubMed] [Google Scholar]
29. Oelschlaegel S, Gruner M, Wang P-N, Boettcher A, Koelling-Speer I, Speer K. Classification and characterization of manuka honeys based on phenolic compounds and methylglyoxal. J Agric Food Chem. 2012;60:7229–7237. [PubMed] [Google Scholar]
30. Tuberoso CI, Jerković I, Bifulco E, Marijanović Z, Congiu F, Bubalo D. Riboflavin and lumichrome in Dalmatian sage honey and other unifloral honeys determined by LC-DAD technique. Food Chem. 2012;135:1985–1990. [PubMed] [Google Scholar]
31. Tuberoso CI, Jerković I, Bifulco E, Marijanović Z. Biodiversity of Salix spp. honeydew and nectar honeys determined by RP-HPLC and evaluation of their antioxidant capacity. Chem Biodivers. 2011;8:872–879. [PubMed] [Google Scholar]
32. Kús PM, Jerković I, Tuberoso CIG, Marijanović Z, Congiu F. Cornflower (Centaurea cyanus L.) honey quality parameters: chromatographic fingerprints, chemical biomarkers, antioxidant capacity and others. Food Chem. 2014;142:12–18. [PubMed] [Google Scholar]
33. Jerković I, Kranjac M, Marijanović Z, et al. Screening of Satureja subspicata Vis. honey by HPLC-DAD, GC-FID/MS and UV/Vis: prephenate derivatives as biomarkers. Molecules. 2016;21:377. doi: 10.3390/molecules21030377. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
34. Manyi-Loh CE, Ndip RN, Clarke AM. Volatile compounds in honey: a review on their involvement in aroma botanical origin determination and potential biomedical activities. Int J Mol Sci. 2011;12:9514–9532. [PMC free article] [PubMed] [Google Scholar]
35. Cuevas-Glory LF, Pino JA, Santiago LS, Sauri-Duch E. A review of volatile analytical methods for determining the botanical origin of honey. Food Chem. 2007;103:1032–1043. [Google Scholar]
36. Pohl P. Determination of metal content in honey by atomic absorption and emission spectrometries. Trends Anal Chem. 2009;28:117–128. [Google Scholar]
37. Gašić UM, Natić MM, Mišić DM, et al. Chemical markers for the authentication of unifloral Salvia officinalis L. honey. J Food Compos Anal. 2015;44:128–138. [Google Scholar]
38. Gambacorta E, Simonetti A, Garrisi N, Intaglietta I, Perna A. Antioxidant properties and phenolic content of sulla (Hedysarum sp.)honeys from Southern Italy. Int J Food Sci Technol. 2014;49:2260–2268. [Google Scholar]
39. Karabagias IK, Vavoura MV, Badeka A, Kontakos S, Kontominas MG. Differentiation of Greek thyme honeys according to geographical origin based on the combination of phenolic compounds and conventional quality parameters using chemometrics. Food Anal Methods. 2014;7:2113–2121. [Google Scholar]
40. Zhao J, Du X, Cheng N, et al. Identification of monofloral honeys using HPLC-ECD, and chemometrics. Food Chem. 2016;194:167–174. [PubMed] [Google Scholar]
41. Arvanitoyannis IS, Chalhoub C, Gotsiou P, Lydarkis-Simantiris N, Kefalas P. Novel quality control methods in conjunction with chemometrics (multivariate analysis) for detecting honey authenticity. Crit Rev Food Sci Nutr. 2005;45:193–203. [PubMed] [Google Scholar]
42. Erejuwa OO, Sulaiman SA, Ab Wahab MS. Honey: a novel antioxidant. Molecules. 2012;17:4400–4423. [PMC free article] [PubMed] [Google Scholar]
43. Ferreira ICFR, Aires E, Barreira JCM, Estevinho LM. Antioxidant activity of Portuguese honey samples: different contributions of the entire honey and phenolic extract. Food Chem. 2009;114:1438–1443. [Google Scholar]
44. Alvarez-Suarez JM, Tulipani S, Romandini S, Vidal A, Battino M. Methodological aspects about determination of phenolic compounds and in vitro evaluation of antioxidant capacity in the honey: a review. Curr Anal Chem. 2009;5:293–302. [Google Scholar]
45. Aazza S, Lyoussi B, Antunes D, Miguel MG. Physico-chemical characterization and antioxidant activity of commercial Portuguese honeys. J Food Sci. 2013;78:C1159–C1165. [PubMed] [Google Scholar]
46. Aazza S, Lyoussi B, Antunes D, Miguel MG. Physico-chemical characterization and antioxidant activity of 17 commercial Moroccan honeys. Int J Food Nutr. 2014;65:449–457. [PubMed] [Google Scholar]
47. Gheldof N, Wang X-H, Engeseth NJ. Identification and quantification of antioxidant components of honeys from various floral sources. J Agric Food Chem. 2002;50:5870–5877. [PubMed] [Google Scholar]
48. Karabagias IK, Dimitriou E, Kontakos S, Kontominas MG. Phenolic profile, colour intensity, and radical scavenging activity of Greek unifloral honeys. Eur Food Res Technol. 2016;242:1201–1210. [Google Scholar]
49. Brudzynski K, Maldonado-Alvarez L. Polyphenol-protein complexes and their consequences for the redox activity, structure and function of honey. A current view and new hypothesis – a review. Pol J Food Nutr Sci. 2015;65:71–80. [Google Scholar]
50. Rice-Evans CA, Miller NJ, Paganga G. Antioxidant properties of phenolic compounds. Trends Plant Sci. 1997;2:152–159. [Google Scholar]
51. Sghaier MB, Skandrani I, Nasr N, Franca MD, Chekir-Ghedira L, Ghedira K. Flavonoids and sesquiterpenes from Tecurium ramosissimum promote antiproliferation of human cancer cells and enhance antioxidant activity: a structure-activity relationship study. Environ Toxicol Pharmacol. 2011;32:336–348. [PubMed] [Google Scholar]
52. Brudzynski K, Sjaarda C, Maldonado-Alvarez L. A new look on protein-polyphenol complexation during honey storage: is this a random or organized event with the help of dirigent-like proteins? PLoS ONE. 2013;8:e72897. [PMC free article] [PubMed] [Google Scholar]
53. Turkmen N, Sari F, Poyrazoglu ES, Velioglu YS. Effects of prolonged heating on antioxidant activity and color of honey. Food Chem. 2006;95:653–657. [Google Scholar]
54. Brudzynski K, Miotto D. Honey melanoidins. Analysis of a composition of the high molecular weight melanoidin fractions exhibiting radical scavenging capacity. Food Chem. 2011;127:1023–1030. [PubMed] [Google Scholar]
55. Brudzynski K, Miotto D. The recognition of high molecular weight melanoidins as the main components responsible for radical-scavenging capacity of unheated and heat-treated Canadian honeys. Food Chem. 2011;125:570–575. [Google Scholar]
56. Chaikham P, Kemsawasd V, Apichartsrangkoon A. Effects of conventional on physicochemical properties and antioxidant capacity of floral honeys from Northern Thailand. Food Biosci. 2016;15:19–26. [Google Scholar]
57. Escuredo O, Míguez M, Fernández-González M, Seijo MC. Nutritional value and antioxidant activity of honeys produced in a European Atlantic area. Food Chem. 2013;138:851–856. [PubMed] [Google Scholar]
58. Rodríguez-Flores S, Escuredo O, Seijo MC. Characterization and antioxidant capacity of sweet chestnut honey produced in North-West Spain. J Apic Sci. 2016;60:19–30. [Google Scholar]
59. Alvarez-Suarez JM, Giampier F, Battino M. Honey as a source of dietary antioxidants: structures, bioavailability and evidence of protective effects against human chronic diseases. Curr Med Chem. 2013;20:621–638. [PubMed] [Google Scholar]
60. Jeon M, Zhao Y. Honey in combination with vacuum impregnation to prevent enzymatic browning of fresh-cut apples. Int J Food Sci Nutr. 2005;56:165–176. [PubMed] [Google Scholar]
61. Chang X, Wang J, Yang S, Chen S, Song Y. Antioxidative, antibrowning and antibacterial activities of sixteen floral honeys. Food Funct. 2011;2:541–546. [PubMed] [Google Scholar]
62. Song JJ, Salcido R. Use of honey in wound care: an update. Adv Skin Wound Care. 2011;24:40–44. [PubMed] [Google Scholar]
63. Barrientos S, Stojadinovic O, Golinko MS, Brem H, Tomic-Canic M. Growth factors and cytokines in wound healing. Wound Rep Reg. 2008;16:585–601. [PubMed] [Google Scholar]
64. Tonks A, Cooper R, Price AJ, Molan PC, Jones KP. Stimulation of TNF-alpha release in monocytes by honey. Cytokine. 2001;14:240–242. [PubMed] [Google Scholar]
65. Tonks AJ, Cooper RA, Jones KP, Blair S, Parton J, Tonks A. Honey stimulates inflammatory cytokine production from monocytes. Cytokine. 2003;21:242–247. [PubMed] [Google Scholar]
66. Tonks AJ, Dudley E, Porter NG, et al. A 5.8 kDa component of manuka honey stimulates immune cells via TLR4. J Leukocyte Biol. 2007;82:1147–1155. [PubMed] [Google Scholar]
67. Majtan J, Kumar P, Majtan T, Walls AF, Klaudiny J. Effect of honey and its major royal jelly protein 1 on cytokine and MMP-9 mRNA transcripts in human keratinocytes. Exp Dermatol. 2010;19:e73–e79. [PubMed] [Google Scholar]
68. Gannabathula S, Skinner MA, Rosendale D, et al. Arabinogalactan proteins contribute to the immunostimulatory properties of New Zealand honeys. Immunopharmacol Immunotoxicol. 2012;34:598–607. [PubMed] [Google Scholar]
69. Raynaud A, Ghezali L, Gloaguen V, Liagre B, Quero F, Petit JM. Honey-induced macrophage stimulation: AP-1 and NF-κB activation and cytokine production are unrelated to LPS content of honey. Int Immunopharmacol. 2013;17:874–879. [PubMed] [Google Scholar]
70. McLoone P, Warnock M, Fyfe L. Honey: an immunomodulatory agent for disorders of the skin. Food Agric Immunol. 2016;27:338–349. [Google Scholar]
71. Oryan A, Alemzadeh E, Moshiri A. Biological properties and therapeutic activities of honey in wound healing: a narrative review and meta-analysis. J Tissue Viability. 2016;25:98–118. [PubMed] [Google Scholar]
72. Pereira RF, Bártolo PJ. Traditional therapies for skin wound healing. Adv Wound Care. 2016;5:208–229. [PMC free article] [PubMed] [Google Scholar]
73. Henriques A, Jackson S, Cooper R, Burton N. Free radical production and quenching in honeys with wound healing potential. J Antimicrob Chemother. 2006;58:773–777. [PubMed] [Google Scholar]
74. Chepulis LM, Francis E. An initial investigation into the anti-inflammatory activity and antioxidant capacity of alpha-cyclodextrin-complexed manuka honey. J Complement Integr Med. 2012;9:25. doi: 10.1515/1553-3840.1646. [PubMed] [CrossRef] [Google Scholar]
75. Ranzato E, Martinotti S, Burlando B. Epithelial mesenchymal transition traits in honey-driven keratinocyte wound healing: comparison among different honeys. Wound Repair Regen. 2012;20:778–785. [PubMed] [Google Scholar]
76. Majtan J. Honey: an immunomodulator in wound healing. Wound Repair Regen. 2014;22:187–192. [PubMed] [Google Scholar]
77. Abuharfeil N, Aloran R, Aboshehada M. The effect of bee honey on the proliferative activity of human B and T-lymphocytes and the activity of phagocytes. Food Agric Immunol. 1999;11:169–177. [Google Scholar]
78. Ranzato E, Martinotti S, Burlando B. Honey exposure stimulates wound repair of human dermal fibroblasts. Burns Trauma. 2013;1:32–38. [PMC free article] [PubMed] [Google Scholar]
79. Timm M, Bartfelt S, Hansen EW. Immunomodulatory effects of honey cannot be distinguished from endotoxin. Cytokine. 2008;42:113–120. [PubMed] [Google Scholar]
80. Bang LM, Buntting C, Molan PC. The effect of dilution on the rate of hydrogen peroxide production in honey and its implications for wound healing. J Alt Compl Med. 2003;9:267–273. [PubMed] [Google Scholar]
81. Kassim M, Mansor M, Al-Abd N, Yusoff KM. Gelam honey has a protective effect against lipopolysaccharide (LPS)-induced organ failure. Int J Mol Sci. 2012;13:6370–6381. [PMC free article] [PubMed] [Google Scholar]
82. Keenan JI, Salm N, Wallace AJ, Hampton MB. Using food to reduce H. pylori-associated inflammation. Phytother Res. 2012;26:1620–1625. [PubMed] [Google Scholar]
83. Prakash A, Medhi B, Avti PK, Saikia UN. Effect of different doses of manuka honey in experimentally induced inflammatory bowel disease in rats. Phytother Res. 2008;22:1511–1519. [PubMed] [Google Scholar]
84. Bashkaran K, Zunaina E, Bakiah S, Sulaiman SA, Sirajudeen KN, Naik V. Anti-inflammatory and antioxidant effects of Tualang honey in alkali injury on the eyes of rabbits: experimental animal study. BMC Complement Altern Med. 2011;11:90. doi: 10.1186/1472-6882-11-90. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
85. Almasandi SB, El-Shitani NA, Abbas AT, et al. Antioxidant, anti-inflammatory, and antiulcer potential of manuka honey against gastric ulcer in rats. Oxid Med Cell Longev. 2016;2016:3643824. doi: 10.1155/2016/3643824. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
86. da Silva CI, Aazza S, Faleiro ML, Miguel MG, Neto L. The antibacterial, anti-biofilm, anti-inflammatory and virulence inhibition properties of Portuguese honeys. J Apic Res. 2016;55:292–304. [Google Scholar]
87. Kolayli S, Sahin H, Can Z, Yildiz O, Sahin K. Honey shows potent inhibitory activity against the bovine testes hyaluronidase. J Enzyme Inhib Med Chem. 2016;31:599–602. [PubMed] [Google Scholar]
88. Osés SM, Pascual-Maté A, Fernández-Muiño MA, Lopez-Díaz TM, Sancho MT. Bioactive properties of honey with propolis. Food Chem. 2016;196:1215–1223. [PubMed] [Google Scholar]
89. Nooh HZ, Nour-Eldien NM. The dual anti-inflammatory and antioxidant activities of natural honey promote cell proliferation and neural regeneration in a rat model of colitis. Acta Histochem. 2016;118:588–595. [PubMed] [Google Scholar]
90. de Assis POA, Guerra GCB, Araújo DFS, et al. Intestinal anti-inflammatory activity of goat milk and goat yoghurt in the acetic acid model of rat colitis. Int Dairy J. 2016;56:45–54. [Google Scholar]
91. Devasvaran K, Yong Y-K. Anti-inflammatory and wound healing properties of Malaysia Tualang Honey. Curr Sci. 2016;110:47–51. [Google Scholar]
92. Al-Waili NS, Salom K, Al-Ghamdi A. Honey for wound healing, ulcers, and burns; data supporting its use in chemical practice. Sci World J. 2011;11:766–787. [PMC free article] [PubMed] [Google Scholar]
93. Vandamme L, Heyneman A, Hoeksema H, Verbelen J, Moustrey S. Honey in modern wound care: a systematic review. Burns. 2013;39:1514–1525. [PubMed] [Google Scholar]
94. Khoo Y-T, Halim AS, Singh K-KB, Mohamad N-A. Wound contraction effects and antibacterial properties of Tualang honey on full-thickness burn wounds in rats in comparison to hydrofibre. BMC Complement Altern Med. 2010;10:48. [PMC free article] [PubMed] [Google Scholar]
95. Osuagwu FC, Oladejo OW, Imosemi IO, et al. Enhanced wound contraction in fresh wounds dressed with honey in wistar rats (Rattus novergicus). West Afr J Med. 2004;23:114–118. [PubMed] [Google Scholar]
96. Farzadinia P, Jofreh N, Khatamsaz S, et al. Anti-inflammatory and wound healing activities of Aloe vera, honey and milk ointment on second-degree burns in rats. Int J Low Extrem Wounds. 2016;15:241–247. [PubMed] [Google Scholar]
97. Schencke C, Vásquez B, Sandoval C, Del Sol M. El rol de la miel en los procesos morfofisiologicos de reparación de heridas. Int J Morphol. 2016;34:385–395. [Google Scholar]
98. Sarhan WA, Azzazy HM, El-Sherbiny IM. Honey/chitosan nanofiber wound dressing enriched with Allium sativum and Cleome droserifolia: enhanced antimicrobial and wound healing activity. ACS Appl Mater Interfaces. 2016;8:6379–6390. [PubMed] [Google Scholar]
99. Jull A, Walker N, Parag V, Molan P, Rodgers A. Randomized clinical trial of honey-impregnated dressings for venous leg ulcers. Br J Surg. 2008;95:175–182. [PubMed] [Google Scholar]
100. Kateel R, Adhikari P, Augustine AJ, Ullal S. Topical honey for the treatment of diabetic foot ulcer: a systematic review. Complement Ther Clin Pract. 2016;24:130–133. [PubMed] [Google Scholar]
101. Jull AB, Cullum N, Dumville JC, Westby MJ, Deshpande S, Walker N. Honey as a topical treatment for wounds. Cochrane Database Syst Rev. 2015;3:CD005083. [PMC free article] [PubMed] [Google Scholar]
102. Fernandez-Cabezudo MJ, El-Kharrag R, Torab F, et al. Intravenous administration of manuka honey inhibits tumor growth and improves host survival when used in combination with chemotherapy in a melanoma mouse model. PLoS ONE. 2013;8:e55993. [PMC free article] [PubMed] [Google Scholar]
103. Spilioti E, Jaakkola M, Tolonen T, et al. Phenolic acid composition, antiatherogenic and anticancer potential of honeys derived from various regions in Greece. PLoS ONE. 2014;9:e94860. [PMC free article] [PubMed] [Google Scholar]
104. Ghashm AA, Othman NH, Khattak MN, Ismail NM, Saini R. Antiproliferative effect of tualang honey on oral squamous cell carcinoma and osteosarcoma cell lines. BMC Complement Altern Med. 2010;10:49. [PMC free article] [PubMed] [Google Scholar]
105. Tsiapara AV, Jaakkola M, Chinou I, et al. Bioactivity of Greek honey extracts on breast cancer (MCF-7), prostate cancer (PC-3) and endometrial cancer (Ishikawa) cells: profile analysis of extracts. Food Chem. 2009;116:702–708. [Google Scholar]
106. Yaacob NS, Nengsih A, Norazmi MN. Tualang honey promotes apoptotic cell death induced by tamoxifen in breast cancer cell lines. Evid Based Complement Alternat Med. 2013;2013:989841. doi: 10.1155/2013/989841. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
107. Swellam T, Miyanaga N, Onozawa M, et al. Antineoplastic activity of honey in an experimental bladder cancer implantation model: in vivo and in vitro studies. Int J Urol. 2003;10:213–219. [PubMed] [Google Scholar]
108. Samarghandian S, Afshari JT, Davoodi S. Honey induces apoptosis in renal cell carcinoma. Pharmacogn Mag. 2011;7:46–52. [PMC free article] [PubMed] [Google Scholar]
109. Pichichero E, Cicconi R, Mattei M, Muzi MG, Canini A. Acacia honey and chrysin reduce proliferation of melanoma cells through alterations in cell cycle progression. Int J Oncol. 2010;37:973–981. [PubMed] [Google Scholar]
110. Aliyu M, Odunola OA, Farooq AD, et al. Molecular mechanism of antiproliferation potential of Acacia honey on NCI-H460 cell line. Nutr Cancer. 2013;65:296–304. [PubMed] [Google Scholar]
111. Morales P, Haza AI. Antiproliferative and apoptotic effects of Spanish honeys. Pharmacogn Mag. 2013;9:231–237. [PMC free article] [PubMed] [Google Scholar]
112. Kadir EA, Sulaiman SA, Yahya NK, Othman NH. Inhibitory effects of tualang honey on experimental breast cancer in rats: a preliminary study. Asian Pac J Cancer Prev. 2013;14:2249–2254. [PubMed] [Google Scholar]
113. Oršolić N, Terzić S, Šver L, Bašić I. Honey-bee products in prevention and/or therapy of murine transplantable tumours. J Sci Food Agric. 2005;85:363–370. [Google Scholar]
114. El-Kott AF, Kandeel AA, El-Aziz SFA, Ribea HM. Anti-tumor effects of bee honey on PCNA and P53 expression in the rat hepatocarcinogenesis. Int J Cancer Res. 2012;8:130–139. [Google Scholar]
115. Mabrouk GM, Moselhy SS, Zohny SF, et al. Inhibition of methylnitrosourea (MNU) induced oxidative stress and carcinogenesis by orally administered bee honey and Nigella grains in Sprague Dawely rats. J Exp Clin Cancer Res. 2002;21:341–346. [PubMed] [Google Scholar]
116. Erejuwa OO, Sulaiman SA, Wahab MS. Effects of honey and its mechanisms of action on the development and progression of cancer. Molecules. 2014;19:2497–2522. [PMC free article] [PubMed] [Google Scholar]
117. Ahmed S, Othman NH. Honey as a potential natural anticancer agent: a review of its mechanisms. Evid Based Complement Alternat Med. 2013;2013:829070. doi: 10.1155/2013/829070. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
118. MacEwan DJ. TNF ligands and receptors – a matter of life and death. Brit J Pharmacol. 2002;135:855–875. [PMC free article] [PubMed] [Google Scholar]
119. Jaganathan SK, Arunpandian B, Vellayappan MV, et al. A review on antiproliferative and apoptotic activities of natural honey. Anticancer Agent Med Chem. 2015;15:48–56. [PubMed] [Google Scholar]
120. Portokalakis I, Yusof H, Ghanotakis D, Nigam P, Owusu-Apenten R. Manuka honey-induced cytotoxicity against MCF7 breast cancer cells is correlated to total phenol content and antioxidant power. J Adv Biol Biotechnol. 2016;8:1–10. [Google Scholar]
121. Fauzi AN, Norazmi MN, Yaacob NS. Tualang honey induces apoptosis and disrupts the mitochondrial membrane potential of human breast and cervical cancer cell lines. Food Chem Toxicol. 2011;49:871–878. [PubMed] [Google Scholar]
122. Bel-Serrat S, Mouratidou T, Börnhorst C, et al. Food consumption and cardiovascular risk factors in European children: the IDEFICS study. Pediatr Obes. 2012;8:225–236. [PubMed] [Google Scholar]
123. González-Gil EM, Santabárbara J, Russo P, et al. Food intake and inflammation in European children: the IDEFICS study. Eur J Nutr. 2016;55:2459–2468. [PubMed] [Google Scholar]
124. Yaghoobi N, Al-Waili N, Ghayour-Mobarhan M, et al. Natural honey and cardiovascular risk factors; effects on blood glucose, cholesterol, triacylglycerole, CRP, and body weight compared with sucrose. Sci World J. 2008;8:463–469. [PMC free article] [PubMed] [Google Scholar]
125. Al-Waili NS. Natural honey lowers plasma glucose, C-reactive protein, homocysteine, and blood lipids in healthy, diabetic, and hyperlipidemic subjects: comparison with dextrose and sucrose. J Med Food. 2004;7:100–107. [PubMed] [Google Scholar]
126. Bahrami M, Ataie-Jafari A, Hosseini S, Foruzanfar MH, Rahmani M, Pajouhi M. Effects of natural honey consumption in diabetic patients: an 8-week randomized clinical trial. Int J Food Sci Nutr. 2009;60:618–626. [PubMed] [Google Scholar]
127. Erejuwa OO, Nwobodo NN, Akpan JL, et al. Nigerian honey ameliorates hyperglycemia and dyslipidemia in alloxan-induced diabetic rats. Nutrients. 2016;8:95. doi: 10.3390/nu8030095. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
128. Erejuwa OO, Sulaiman SA, Ab Wahab MS, Sirajudeen KNS, Salzihan MS. Effects of Malaysian tualang honey supplementation on glycemia, free radical scavenging enzymes and markers of oxidative stress in kidneys of normal and streptozotocin-induced diabetic rats. Int J Cardiol. 2009;137:S45. [Google Scholar]
129. Erejuwa OO, Sulaiman SA, Wahab MS. Fructose might contribute to the hypoglycemic effect of honey. Molecules. 2012;17:1900–1915. [PMC free article] [PubMed] [Google Scholar]
130. Erejuwa OO, Sulaiman SA, Wahab MS. Oligosaccharides might contribute to the antidiabetic effect of honey: a review of the literature. Molecules. 2011;17:248–266. [PMC free article] [PubMed] [Google Scholar]
131. Erejuwa OO, Sulaiman SA, Wahab MS. Honey-a novel antidiabetic agent. Int J Biol Sci. 2012;8:913–934. [PMC free article] [PubMed] [Google Scholar]
132. Erejuwa OO. Effect of honey in diabetes mellitus: matters arising. J Diabetes Metab Disord. 2014;13:23 http://www.jdmdonline.com/content/13/1/23. [PMC free article] [PubMed] [Google Scholar]
133. Kearney PM, Whelton M, Reynolds K, Muntner P, Whelton PK, He J. Global burden of hypertension: analysis of worldwide data. Lancet. 2005;365:217–223. [PubMed] [Google Scholar]
134. Erejuwa OO, Sulaiman SA, Ab Wahab MS, Sirajudeen KN, Salleh S, Gurtu S. Honey supplementation in spontaneously hypertensive rats elicits antihypertensive effect via amelioration of renal oxidative stress. Oxid Med Cell Longev. 2012;2012:374037. [PMC free article] [PubMed] [Google Scholar]
135. Sanchez M, Galisteo M, Vera R, et al. Quercetin downregulates NADPH oxidase, increases eNOS activity and prevents endothelial dysfunction in spontaneously hypertensive rats. J Hypertens. 2006;24:75–84. [PubMed] [Google Scholar]
136. Cogolludo A, Frazziano G, Briones AM, et al. The dietary flavonoid quercetin activates BKCa currents in coronary arteries via production of H2O2. Role in vasodilatation. Cardiovasc Res. 2007;73:424–431. [PubMed] [Google Scholar]
137. Shen Y, Croft KD, Hodgson JM, et al. Quercetin and its metabolites improve vessel function by inducing eNOS activity via phosphorylation of AMPK. Biochem Pharmacol. 2012;15:1036–1044. [PubMed] [Google Scholar]
138. Panchal SK, Poudyal H, Brown L. Quercetin ameliorates cardiovascular, hepatic, and metabolic changes in diet-induced metabolic syndrome in rats. J Nutr. 2012;42:1026–1032. [PubMed] [Google Scholar]
139. Kim TH, Ku SK, Lee IC, Bae JS. Anti-inflammatory effects of kaempferol-3-O-sophoroside in human endothelial cells. Inflamm Res. 2012;61:217–224. [PubMed] [Google Scholar]
140. Roberfroid MB. Prebiotics and probiotics: are they functional foods? Am J Clin Nutr. 2000;71:1682S–1687S. [PubMed] [Google Scholar]
141. Nagpal R, Kaur A. Symbiotic effect of various prebiotics on in vitro activities of prebiotic lactobacilli. Ecol Food Nutr. 2011;50:63–68. [PubMed] [Google Scholar]
142. Reddy BS, Hamid R, Rao CV. Effect of dietary oligofructose and inulin on colonic preneoplastic aberrant crypt foci inhibition. Carcinogenesis. 1997;18:1371–1374. [PubMed] [Google Scholar]
143. Slavian J. Fiber and prebiotics: mechanisms and health benefits. Nutrients. 2013;5:1417–1435. [PMC free article] [PubMed] [Google Scholar]
144. Flamm G, Glinsmann W, Kritchevsky D, Prosky L, Roberfroid M. Inulin and oligofructose as dietary fiber: a review of the evidence. Crit Rev Food Sci Nutr. 2001;41:353–362. [PubMed] [Google Scholar]
145. Macfarlane GT, Steed H, Macfarlane S. Bacterial metabolism and health-related effects of galacto-oligosaccharides and other prebiotics. J Appl Microbiol. 2008;104:305–344. [PubMed] [Google Scholar]
146. Perrin S, Warchol M, Grill JP, Schneider F. Fermentations of fructooligosaccharides and their components by Bifidobacterium infantis ATCC 15697 on batch culture in semi-synthetic medium. J Appl Microbiol. 2001;90:859–865. [PubMed] [Google Scholar]
147. Shin H-S, Ustunol Z. Carbohydrate composition of honey from different floral sources and their influence on growth of selected intestinal bacteria: an in vitro comparison. Food Res Int. 2005;38:721–728. [Google Scholar]
148. Sanz ML, Polenis N, Morales V, et al. In vitro investigation into the potential prebiotic activity of honey oligosaccharides. J Agric Food Chem. 2005;53:2914–2921. [PubMed] [Google Scholar]
149. Lucan M, Slacanac V, Hardi J, et al. Inhibitory effect of honey-sweetened goat and cow milk fermented with Bifidobacterium lactis Bb-12 on the growth of Listeria monocytogenes. Mljekarstvo. 2009;59:96–106. [Google Scholar]
150. Chick H, Shin HS, Ustunol Z. Growth and acid production by lactic acid bacteria and bifidobacteria grown in skim milk containing honey. J Food Sci. 2001;66:478–481. [Google Scholar]
151. Cardarelli HR, Saad SM, Gibson GR, Vulevic J. Functional petit-suisse cheese: measure of the prebiotic effect. Anareobe. 2007;13:200–207. [PubMed] [Google Scholar]
152. Macedo LN, Luchese RH, Guerra AF, Barbosa CG. Prebiotic effect of honey on growth and viability of Bifidobacterium spp. and Lactobacillus spp. in milk. Cienc Tecnol Aliment. 2008;28:935–942. [Google Scholar]
153. Kajiwara S, Gandhi H, Ustunol Z. Effect of honey on the growth of and acid production by human intestinal Bifidobacterium spp.: an in vitro comparison with commercial oligosaccharides and inulin. J Food Prot. 2002;65:214–218. [PubMed] [Google Scholar]
154. Brudzynski K, Miotto D. The relationship between the content of Maillard reaction-like products and bioactivity of Canadian honeys. Food Chem. 2010;124:869–874. [Google Scholar]
155. Wang Y, Juliani R, Simon JE, Ho C. Amino acid-dependent formation pathways of 2-acetylfuran and 2,5-dimethyl-4-hydroxy-3[2H]-furanone in the Maillard reaction. Food Chem. 2009;115:233–237. [Google Scholar]
156. Wang J, Li QX. Chemical composition, characterization and differentiation of honey botanical and geographical origins. Adv Food Nutr Res. 2011;62:89–137. [PubMed] [Google Scholar]
157. Brudzynski K, Sjaarda C, Lannigan R. MRJP1-containing glycoproteins isolated from honey, a novel antibacterial drug candidate with broad spectrum activity against multi-drug resistant clinical isolates. Front Microbiol. 2015;6:711. doi: 10.3389/fmicb.2015.00711. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
158. Sousa JM, Souza EL, Marques G, et al. Polyphenolic profile and antioxidant and antibacterial activities of monofloral honeys produced by Meliponini in the Brazilian semiarid region. Food Res Int. 2016;84:61–68. [Google Scholar]
159. Bucekova M, Majtan J. The MRJP1 honey glycoprotein does not contribute to the overall antibacterial activity of natural honey. Eur Food Res Technol. 2016;242:625–629. [Google Scholar]
160. Brudzynski K, Kim L. Storage-induced chemical changes in active components of honey de-regulate its antibacterial activity. Food Chem. 2011;126:1155–1163. [Google Scholar]
161. Lu J, Carter DA, Turnbull L, et al. The effect of New Zealand kanuka, manuka and clover honeys on bacterial growth dynamics and cellular morphology varies according to the species. PLoS ONE. 2013;8:e55898. [PMC free article] [PubMed] [Google Scholar]
162. Henriques AF, Jenkins RE, Burton NF, Cooper RA. The effect of manuka honey on the structures of Pseudomonas aeruginosa. Eur J Clin Microbiol Infect Dis. 2011;30:167–171. [PubMed] [Google Scholar]
163. Roberts AE, Maddocks SE, Cooper RA. Manuka honey is bactericidal against Pseudomonas aeruginosa and results in differential expression of oprF and algD. Microbiol. 2012;158:3005–3013. [PubMed] [Google Scholar]
164. Roberts AE, Maddocks SE, Cooper RA. Manuka honey reduces the motility of Pseudomonas aeruginosa by suppression of flagella-associated genes. J Antimicrob Chemother. 2015;70:716–725. [PubMed] [Google Scholar]
165. Roberts AE, Brown HL, Jenkins RE. On the antibacterial effects of manuka honey: mechanistic insights. Res Rep Biol. 2015;6:215–224. [Google Scholar]
166. Wasfi R, Elkhatib WF, Khairalla AS. Effects of selected Egyptian honeys on the cellular ultrastructure and the gene expression profile of Escherichia coli. PLoS ONE. 2016;11:e0150984. [PMC free article] [PubMed] [Google Scholar]
167. Truchado P, López-Gálvez F, Gil MI, Tomás-Barberán FA, Allende A. Quorum sensing inhibitory and antimicrobial activities of honeys and the relationship with individual phenolics. Food Chem. 2009;115:1337–1344. [Google Scholar]
168. Lee JH, Park JH, Kim JA, et al. Low concentrations of honey reduce biofilm formation, quorum sensing, and virulence in Escherichia coli O157:H7. Biofouling. 2011;27:1095–1104. [PubMed] [Google Scholar]
169. Wang R, Starkey M, Hazan R, Rahme LG. Honey’s ability to counter bacterial infections arises from both bactericidal compounds and QS inhibition. Frontr Microbiol. 2012;3:144. doi: 10.3389/fmicb.2012.00144. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
170. Jenkins R, Burton N, Cooper R. Proteomic and genomic analysis of methicillin-resistant Staphylococcus aureus (MRSA) exposed to manuka honey in vitro demonstrated down-regulation of virulence markers. J Antimicrobial Chemother. 2014;69:603–615. [PMC free article] [PubMed] [Google Scholar]
171. Gemiarto AT, Ninyio NN, Lee SW, et al. Isoprenyl caffeate, a major compound in manuka propolis, is a quorum-sensing inhibitor in Chromobacterium violaceum. Antonie Van Leeuwenhoek. 2015;108:491–504. [PubMed] [Google Scholar]
172. Ni N, Li M, Wang J, Wang B. Inhibitors and antagonists of bacterial quorum sensing. Med Res Rev. 2009;29:65–124. [PubMed] [Google Scholar]
173. Høiby N, Ciofu O, Johansen HK, et al. The clinical impact of bacterial biofilms. Int J Oral Sci. 2011;3:55–65. [PMC free article] [PubMed] [Google Scholar]
174. Halstead FD, Webber MA, Rauf M, Burt R, Dryden M, Oppenheim BA. In vitro activity of an engineered honey, medical-grade honeys, and antimicrobial wound dressings against biofilm-producing clinical bacterial isolates. J Wound Care. 2016;25:93–94. [PubMed] [Google Scholar]
175. Sojka M, Valachova I, Bucekova M, Majtan J. Antibiofilm efficacy of honey and bee-derived defensin-1 on multispecies wound biofilm. J Med Microbiol. 2016;65:337–344. [PubMed] [Google Scholar]
176. Lu J, Turnbull L, Burke CM, et al. Manuka-type honeys can eradicate biofilms produced by Staphylococcus aureus strains with different biofilm-forming abilities. Peer J. 2014;e326. doi: 10.7717/peerj.326. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
177. Campeau ME, Patel R. Antibiofilm activity of Manuka honey in combination with antibiotics. Int J Bacteriol. 2014;2014:795281. [PMC free article] [PubMed] [Google Scholar]
178. Kondra JM, Cooper RA, Maddocks SE. Manuka honey inhibits siderophore production in Pseudomonas aeruginosa. J Appl Microbiol. 2013;115:86–90. [PubMed] [Google Scholar]
179. Regulski M. A novel wound care dressing for chronic leg ulcerations. Pediatry Manag. 2008;27:235–246. [Google Scholar]
180. Smith T, Hanft JR, Legel K. Topical Leptospermum honey in recalcitrant venous leg wounds: a preliminary case series. Adv Skin Wound Care. 2009;22:68–71. [PubMed] [Google Scholar]
181. Tsai CJ, Loh JM, Proft T. Galleria mellonella infection models for the study of bacterial diseases and for antimicrobial drug testing. Virulence. 2016;7:214–229. [PMC free article] [PubMed] [Google Scholar]
182. Sydnor ERM, Perl TM. Hospital epidemiology and infection control in acute-care settings. Clin Microbiol Rev. 2011;24:141–173. [PMC free article] [PubMed] [Google Scholar]
183. Ewnetu Y, Lemma W, Birhane N. Synergetic antimicrobial effects of mixtures of Ethiopian honeys and ginger powder extracts on standard and resistant clinical bacteria isolates. Evid Based Complement Alternat Med. 2014;2004;562804. doi: 10.1155/2014/562804. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
184. Jenkins R, Wooton M, Howe R, Cooper R. A demonstration of the susceptibility of clinical isolates obtained from cystic fibrosis patients to manuka honey. Arch Microbiol. 2015;197:597–601. [PMC free article] [PubMed] [Google Scholar]
185. Liu M, Lu J, Müller P, et al. Antibiotic specific differences in the response of Staphylococcus aureus to treatment with antimicrobials combined with manuka honey. Front Microbiol. 2015;5:779. doi: 10.3389/fmicb.2014.00779. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
186. Nishio EK, Ribeiro JM, Oliveira AG, et al. Antibacterial synergic effect of honey from two stingless bees: Scaptotrigona bipunctata Lepeletier, 1836, and S. postica Latreille, 1807. Sci Rep. 2016;6:21641. [PMC free article] [PubMed] [Google Scholar]
187. Al-Waili N, Al-Ghamdi A, Ansari MJ, Al-Attal Y, Salom K. Synergistic effects of honey and propolis toward drug multi-resistant Staphylococcus aureus, Escherichia coli and Candida albicans isolates in single and polymicrobial cultures. Int J Med Sci. 2012;9:793–800. [PMC free article] [PubMed] [Google Scholar]
188. Ahmed M, Djebli N, Aissat S, Bacha S, Meslem A, Khiati B. Synergistic inhibition of natural honey and potato starch and their correlation with diastase number and sugar content against Klebsiella pneumoniae ATCC 27736. Nat Prod Chem Res. 2012;1:102. [Google Scholar]
189. Watanabe K, Rahmasari R, Matsunaga A, Haruyama T, Kobayashi N. Anti-influenza viral effects of honey in vitro: potent high activity of manuka honey. Arch Med Res. 2014;45:359–365. [PubMed] [Google Scholar]
190. Shahzad A, Cohrs RJ. In vitro antiviral activity of honey against varicella zoster virus (VZV): a translational medicine study for potential remedy for shingles. Transl Biomed. 2012;3:2. doi: 10.3823/434. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
191. Ghapanchi J, Moattari A, Tadbir AA, Talatof Z, Shahidi SP, Ebrahimi H. The in vitro anti-viral activity of honey on type 1 herpes simplex virus. Aust J Basic Appl Sci. 2011;5:849–852. [Google Scholar]
192. Hashemipour MA, Tavakolineghad Z, Arabzadeh SA, Iranmanesh Z, Nassab SA. Antiviral activities of honey, royal jelly, and acyclovir against HSV-1. Wounds. 2014;26:47–54. [PubMed] [Google Scholar]
193. Zeina B, Othman O, Al-Assad S. Effect of honey versus thyme on Rubella virus survival in vitro. J Altern Complement Med. 1996;2:345–348. [PubMed] [Google Scholar]
194. Al-Waili NS. Topical honey application vs. acyclovir for the treatment of recurrent herpes simplex lesions. Med Sci Monit. 2004;10:MT94–MT98. [PubMed] [Google Scholar]
195. Ajibola A, Chamunorwa JP, Erlwanger KH. Nutraceutical values of natural honey and its contribution to human health and wealth. Nutr Metabol. 2012;9:61. [PMC free article] [PubMed] [Google Scholar]
196. Miguel MG, Antunes MD, Aazza S, Duarte J, Faleiro ML. Honey-based ‘água-mel’ chemical characterization and microbiological quality. Ital J Food Sci. 2013;25:275–282. [Google Scholar]
197. Lusby PE, Coombes A, Wilkinson JM. Honey: a potent agent for wound healing? J Wound Ostomy Continence Nurs. 2002;29:295–300. [PubMed] [Google Scholar]
198. Eteraf-Oskouei T, Najafi M. Traditional and modern uses of natural honey in human diseases: a review. Iran J Basic Med Sci. 2013;16:731–742. [PMC free article] [PubMed] [Google Scholar]
199. Schencke C, Vasconcellos A, Sandoval C, Torres P, Acevedo F, del Sol M. Morphometric evaluation of wound healing in burns treated with Ulmo (Eucryphia cordifolia) honey alone and supplemented with ascorbic acid in guinea pig (Cavia porcellus). Burns Trauma. 2016;4:25. [PMC free article] [PubMed] [Google Scholar]

Articles from Integrative Medicine Insights are provided here courtesy of SAGE Publications

-