Next Article in Journal
The Effect of Storage and Pasteurization (Thermal and High-Pressure) Conditions on the Stability of Phycocyanobilin and Phycobiliproteins
Next Article in Special Issue
Chemical Components and Antioxidant Activity of Geotrigona sp. and Tetragonisca fiebrigi Stingless Bee Cerumen Reduce Juglone-Induced Oxidative Stress in Caenorhabditis elegans
Previous Article in Journal
Stimulation of Hepatic Ferritinophagy Mitigates Irp2 Depletion-Induced Anemia
Previous Article in Special Issue
Bee Pollen as Functional Food: Insights into Its Composition and Therapeutic Properties
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Honey: A Promising Therapeutic Supplement for the Prevention and Management of Osteoporosis and Breast Cancer

1
Department of Zoology and Anthropology, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, 949 01 Nitra, Slovakia
2
Department of Botany and Genetics, Faculty of Natural Sciences and Informatics, Constantine the Philosopher University in Nitra, 949 01 Nitra, Slovakia
3
Department of Genetics, Cancer Research Institute, Biomedical Research Center of Slovak Academy of Sciences, 845 05 Bratislava, Slovakia
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Antioxidants 2023, 12(3), 567; https://doi.org/10.3390/antiox12030567
Submission received: 7 February 2023 / Revised: 19 February 2023 / Accepted: 21 February 2023 / Published: 24 February 2023
(This article belongs to the Special Issue Antioxidant Activity of Honey Bee Products)

Abstract

:
Osteoporosis and breast cancer are serious diseases that have become a significant socioeconomic burden. There are biochemical associations between the two disorders in terms of the amended function of estrogen, receptor activator of nuclear factor kappa beta ligand, oxidative stress, inflammation, and lipid accumulation. Honey as a functional food with high antioxidant and anti-inflammatory properties can contribute to the prevention of various diseases. Its health benefits are mainly related to the content of polyphenols. This review aims to summarize the current knowledge from in vitro, animal, and human studies on the use of honey as a potential therapeutic agent for osteoporosis and breast cancer. Preclinical studies have revealed a beneficial impact of honey on both bone health (microstructure, strength, oxidative stress) and breast tissue health (breast cancer cell proliferation and apoptosis, tumor growth rate, and volume). The limited number of clinical trials, especially in osteoporosis, indicates the need for further research to evaluate the potential benefits of honey in the treatment. Clinical studies related to breast cancer have revealed that honey is effective in increasing blood cell counts, interleukin-3 levels, and quality of life. In summary, honey may serve as a prospective therapeutic supplement for bone and breast tissue health.

1. Introduction

Menopause is a biological process characterized by dysfunction of ovarian follicles and estrogen deficiency, oxidative stress, and inflammation, that together lead to different chronic disorders [1,2]. When the organism is exposed to high levels of oxidative stress following estrogen depletion, lipid accumulation also occurs [3]. Osteoporosis and breast cancer are considered serious diseases in which the aforementioned factors are involved and are currently becoming a significant socioeconomic burden worldwide.
Generally, postmenopausal osteoporosis is characterized by reduced bone mineral density (BMD) and increased risk of fragility fractures that are associated with significant pain, suffering, and disability [4]. Moreover, hip and vertebral fractures are consistent with significantly increased mortality [5]. It has been reported that a decrease in estrogen production represents a major cause of reduced bone mass [6,7,8]. During menopause, the osteoprotective effect of estrogen is weakened, leading to elevated expression of pro-inflammatory cytokines that promote osteoclastogenesis [9,10,11,12]. In general, estrogen regulates bone metabolism through two receptors: estrogen receptor alpha (ERα) and estrogen receptor beta (ERβ), with ERα being more dominant. Loss of estrogen also influences osteoblast progenitor cells via reduced ERα expression and lower responsiveness to mechanical stimulation [13]. Thus, estrogen deficiency not only directly affects the differentiation of precursor cells more toward active osteoclasts and less toward osteoblasts but can also influence their cellular energetics. Increased adiposity and inflammation after menopause can indirectly lead to bone loss as well [14].
Similar to bones, breast tissue is also dependent on estrogen [15]. In breast carcinogenesis, elevated exposure to estrogen is linked with early menarche, late menopause, obesity, and estrogen replacement therapy. High blood estrogen levels are able to increase the risk, incidence, and severity of breast malignancy in pre- and postmenopausal women [16]. In general, breast cancer is the second leading cause of cancer death in women, with a higher prevalence in postmenopausal women [1,17]. Consequently, postmenopausal women are at risk of morbidity and mortality, which are a combination of both diseases mentioned above.
Current pharmacological treatment for osteoporosis and breast cancer is often associated with adverse side effects; therefore, various natural therapeutic substances have been intensively studied to find an alternative and effective treatment method with less harmful impacts [17,18]. Honey and other bee products (e.g., royal jelly, propolis, bee bread, drone brood homogenate) are widely used as a functional food due to their high antioxidant and anti-inflammatory properties, which contribute to the prevention of various diseases, including diabetes, osteoporosis, cancer, reproductive disorders [19,20,21,22,23].
In general, honey is a sweet viscous liquid stored in combs after bees collect it from plants. It is produced by regurgitation, enzymatic activity, and evaporation of water in the hives. In addition to the source of nectar, bees also collect insect secretions (belonging to the genus Rhynchota) to produce honeydew honey [2,24,25]. Honey consists of at least 181 substances, mainly carbohydrates such as fructose (38%) and glucose (31%). It also contains enzymes, proteins, amino acids, polyphenols, vitamins, and minerals in lower quantities [26]. The content of polyphenols, which cover a wide spectrum of phytochemicals and are found in almost all types of natural honey, contributes to its health-promoting potential. Such polyphenols include flavonoids (e.g., quercetin, kaempferol, luteolin, hesperetin, chrysin, apigenin, galangin), phenolic acids (e.g., ellagic, caffeic, gallic, ferulic, benzoic, ascorbic), antioxidant enzymes (e.g., catalase, glucose oxidase, peroxidase) and carotenoids [27,28]. Most of these compounds interact with each other to create a range of synergistic antioxidant properties. Many studies revealed antioxidant, antibacterial, antiviral, immunomodulatory, anti-inflammatory, hypocholesterolemic, hypotensive, and antitumor impacts of honey [2,29]. The composition of a particular honey sample depends to a large extent on the nectar composition, the method of nectar collecting, environmental and seasonal factors, geographical origin, as well as storage conditions [30].
This review aims to summarize the current knowledge from preclinical and clinical studies regarding the use of honey as a potential therapeutic agent for osteoporosis and breast cancer due to their elevated incidence in postmenopausal women. Biochemical connections between the two disorders are also provided.

2. Biochemical Associations between Osteoporosis and Breast Cancer

Biochemical connections between osteoporosis and breast cancer include the amended function of receptor activator of nuclear factor kappa beta ligand (RANKL), estrogen, reactive oxygen species (ROS)-induced oxidative stress, chronic low-grade inflammation, and lipid accumulation [1,2]. A clearer understanding of the associations between these diseases can lead to the development of a therapeutic target for postmenopausal breast cancer patients. Figure 1 illustrates the influence of RANKL, estrogen, ROS, and inflammation on the development of osteoporosis and breast cancer.
RANKL is an important cytokine that is a member of the tumor necrosis factor (TNF) family and is encoded by the tumor necrosis factor ligand super family 11 (TNFSF11) gene [31]. It plays an important role in human physiology by controlling the differentiation and activation of osteoclasts [32]. Generally, RANKL binds to the receptor activator of nuclear factor kappa beta (RANK) on osteoclast precursor cells. RANKL/RANK interaction subsequently activates nuclear factor kΒ (NF-kB) and supports the expression of other osteoclastogenic factors. Conversely, a soluble decoy receptor for RANKL-osteoprotegerin (OPG) prevents RANKL from binding to RANK. Therefore, RANKL/RANK/OPG system is considered a key mediator of osteoclastogenesis [18,33]. Moreover, RANKL/RANK pathway has been implicated in breast development as well as breast carcinogenesis. According to Fata et al. [34], lactating mammary gland did not develop cancer in RANK and RANKL receptor-deficient mice. In the study by Gonzalez-Suarez et al. [35], the development of mammary carcinogenesis was related to a higher expression of RANKL in 7,12-dimethylbenzeneanthracene (DMBA)-induced mice, with accelerated breast carcinogenesis identified in RANK-transgenic mice. RANKL also initiates the formation of pre-cancerous lesions and the metastatic process. Additionally, RANKL up-regulates the angiogenic process by stimulating the proliferation and survival of endothelial cells (Figure 1).
Both bone and breast tissues are dependent on estrogen. Moreover, high BMD can be associated with the risk of breast cancer [36]. The hormone estrogen is a key regulator of BMD [37], maintaining the balance between bone formation and bone resorption [38]. Specifically, estrogens stimulate osteoblast differentiation and activate Wnt signaling. They also have an indirect effect through suppression of RANKL and up-regulation of OPG, which ultimately inhibits osteoclastogenesis. Another mechanism for preventing bone resorption is the induction of apoptosis in osteoclasts. Furthermore, estrogens act at the osteocyte level since estrogen decreases sclerostin level and osteocyte apoptosis (Figure 1). Epidemiological and clinical evidence has shown that factors consistent with raised estrogen levels during a woman’s lifetime (e.g., early menarche, late menopause, late first full-term pregnancy, obesity) are related to increased risk of breast cancer [39]. Estrogens are generally believed to induce breast cancer cell proliferation via the ER and serve as a transcription factor to regulate the expression of target genes encoding proteins with important biological functions [40]. The impact of estrogen on both aforementioned diseases documents the fact that women who develop ER-positive breast cancer at a relatively younger age and are treated with anti-estrogen drugs such as tamoxifen have an elevated risk of postmenopausal osteoporosis [41]. Due to the role of estrogen in breast cancer, aromatase inhibitors (inhibitors of the estrogen-metabolizing enzyme aromatase) are used in the treatment of postmenopausal individuals with ER-positive breast cancer, despite problems with bone fractures. Recently, researchers are examining the potential of denosumab, an anti-RANKL antibody, in preventing aromatase inhibitor-associated bone loss [42], which could provide major benefits for postmenopausal breast cancer patients.
Oxidative stress is a contributing factor in many chronic diseases, including osteoporosis and breast cancer [43,44]. ROS directly promote osteoclast formation in a process mediated by RANKL-RANK interaction and enhance bone resorption [45,46]. This signaling pathway includes redox-sensitive components such as tumor necrosis factor receptor-associated factor 6 (TRAF6), Rac1 (a member of the Rho-GTPase subfamily), and nicotinamide adenine dinucleotide phosphate oxidases (NOX) [47]. Moreover, ROS induce apoptosis of osteoblasts and osteocytes by activating numerous signaling pathways. Mitogen-activated protein kinases such as ERK and JNK are involved in this process (Figure 1). ROS also reduce osteoblast activity and differentiation, thus mineralization and osteogenesis [48]. Postmenopausal women are not only exposed to high levels of oxidative stress, but also to elevated levels of nitric oxide (NO) in erythrocytes [49]. NO can increase the ability of cytokines to stimulate osteoclast activity and enhances their inhibitory impacts on osteoblast growth [50,51]. Ultimately, bone formation prevails over bone resorption. In breast cancer, oxidative stress has been implicated in the initiation, promotion, and progression grades of breast carcinogenesis [52]. Mammary tissue is a complex combination of different cell types, including stromal and neoplastic cells [53]. In cancerous breast tissue, stromal fibroblasts acquire a phenotype characterized by raised levels of cytokines, growth factors, and metalloproteinases [54]. In the tumor microenvironment, an altered redox state in favor of pro-oxidants induces the formation of activated fibroblasts, leading to modifications of epithelial cells that support tumorigenesis [55]. Oxidative stress in the tumor microenvironment is also characterized by activated stromal cells that generate tumor-enhancing signals, thereby promoting tumor growth and vascularization [56]. Elevated ROS induce oncogenes and DNA damage, inhibit tumor suppressor genes, and can interfere with different signaling pathways (Figure 1).
Chronic age-related inflammation also plays an important role in the pathogenesis of osteoporosis by affecting bone remodeling [57]. In the presence of RANKL, pro-inflammatory cytokines such as tumor necrosis factor alpha (TNF-α), interleukins (IL)-1, and IL-6 cause the excessive formation of osteoclasts and simultaneously inhibit the activities of osteoblasts [58]. The aforementioned cytokines also stimulate osteoclast development and elevate the production of macrophage colony-stimulating factor (M-CSF) by bone marrow stromal cells (BMSC) [59,60]. They also suppress osteoblasts in releasing OPG [58]. According to several studies, raised levels of cytokine-mediated acute phase C-reactive protein [61], and pro-inflammatory cytokines, including IL-6 [62], IL-1β [63], and TNF- α [64] are found in breast cancer patients, documenting that breast cancer is associated with inflammation. Elevation of these cytokines has been linked with breast cancer invasiveness and has also been used as a prognostic factor in breast cancer patients [65]. Inflammatory cells such as macrophages play a role during tumor progression by stimulating angiogenesis via the production of pro-inflammatory cytokines and VEGF (Figure 1).
Obesity, one of the abnormalities of lipid metabolism, has been hypothesized to protect the skeleton by increasing BMD [1] through mechanical loading, which stimulates bone formation by reducing apoptosis and increasing the proliferation and differentiation of osteoblasts and osteocytes [66,67]. This mechanism is supposed to be controlled by the Wnt/β-catenin signaling pathway [68,69]. For this reason, bone mass increases as a compensatory mechanism to adapt to a greater load [70]. However, several researchers reported conflicting findings. According to Hsu et al. [71] and Pollock et al. [72], excess fat mass was associated with reduced total BMD and total bone mineral content. The link between obesity and 13 cancer types, including ER-positive postmenopausal breast cancer, was established by International Agency for Cancer Research [73]. Moreover, obesity was consistent with poor response outcomes in patients with ER-positive breast cancer [74]. Therefore, obesity presents a challenge in treating individuals with postmenopausal breast cancer who suffer from osteoporosis [75]. Targeting the metabolic pathways linked to estrogen production and immune surveillance modulation might represent an effective trend in breast cancer prevention and treatment [76]. Studies on estradiol depletion by aromatase inhibitors in subjects with postmenopausal breast cancer indicate that higher levels of aromatase activity associated with elevated adipose tissue mass, reduce the efficacy of aromatase inhibitor therapy [77]. Nowadays, bisphosphonates are used to prevent aromatase inhibitor-induced bone loss and improve survival in postmenopausal patients with ER-positive breast cancer [78,79].

3. Honey and Osteoporosis

Honey is able to protect the bone mainly due to antioxidant and anti-inflammatory properties, primarily through its content of polyphenols, which act on several signaling pathways, resulting in anabolic and antiresorptive effects [2]. From the group of polyphenols, the anti-osteoporotic impact of quercetin, kaempferol, and luteolin was recorded [4]. In addition, vitamin D3 and its hydroxyderivatives with antioxidant properties were also detected in honey [80,81]. Vitamin D3 supplementation was found to have protective effects on the inhibition of bone loss and BMD in both experimental animals and postmenopausal women [82,83,84].
According to Zaid et al. [85], the thickness of trabecular bone was elevated in ovariectomized (OVX) rats receiving Tualang honey (a type of Malaysian honey that is especially produced by the rock bee) at the dose of 0.2 g/kg/day for 2 weeks compared to OVX rats fed calcium [86]. Additionally, identical Tualang honey administration (0.2 g/kg/day for 2 weeks) significantly increased BMD in OVX rats [87]. The study of Husniati Y et al. [88] showed that daily consumption of Tualang honey (20 mg/day for 4 months) in postmenopausal women resulted in similar bone densitometry findings as in individuals with hormone replacement therapy. Moreover, Shafin et al. [89] revealed that postmenopausal women consuming Tualang honey (20 g for 16 weeks) had comparable blood oxidative stress (e.g., glutathione peroxidase, catalase, superoxide dismutase) levels to those receiving estrogen-progestin therapy. The aforementioned beneficial effects of Tualang honey can be attributed to the highest content of kaempferol, quercetin, ellagic acid, gallic acid, hesperetin, and catechin among different types of honey, indicating its highest antioxidant potential [90,91,92].
According to Kamaruzzaman et al. [93], the administration of Kelulut honey (a type of Malaysian honey that is mainly produced by stingless bumblebees) at doses of 200 mg/kg/day and 400 mg/kg/day for 2 months alleviated glucocorticoid-induced osteoporosis through its antioxidant activity in rats. Significantly elevated bone volume/tissue volume, trabecular number, osteoblast surface, superoxide dismutase level and decreased osteoclast surface and malondialdehyde levels were determined in osteoporotic rats fed this type of honey. The impact of Kelulut honey supplementation (1 g/kg for 8 weeks) on the bone health of rats with metabolic syndrome was investigated by Ekeuku et al. [94]. Oxidative stress and chronic low-grade inflammation present in metabolic syndrome are known to play a major role in osteoporosis induction or bone loss [19]. Rats receiving Kelulut honey showed a significant reduction in osteoclast surface compared to the control group, other bone parameters did not differ between the two groups [94]. However, Ramli et al. [24] report that honey has a strong potential to be used in the management of metabolic syndrome and related osteoporosis by exerting anti-obesity, hypolipidemic, antidiabetic, and hypotensive activities.
Yudaniayanti et al. [30] examined the impact of honey supplements on bone strength in OVX rats. These authors determined significantly increased bone strength in OVX rats receiving honey (2 g/kg and 4 g/kg for 12 weeks) in comparison with the untreated group. According to Hasib et al. [95], honey administration (1 g/kg, 2 g/kg, and 4 g/kg for 2 weeks) had a favorable effect on osteoporotic fracture healing in rat femur by promoting osteoblastogenesis. The pro-osteoblastic influence of honey was documented by an enhanced level of alkaline phosphatase (ALP) in the serum.
Abu-Serie et al. [96] revealed the ameliorative impact of a combined extract of Greek thyme (Thymus vulgaris) and honey on hydrocortisone-induced osteoporosis in rat bone cells through modulation of bone turnover, oxidative stress, and inflammation. Moreover, a stronger anti-osteoporotic effect of the combined extract was recorded compared to a commonly used bisphosphonate drug (alendronate).
Interestingly, Manuka honey (a type of New Zealand honey with antimicrobial and antioxidant capacities) was used as an antibacterial agent incorporated into a biopolymer coating based on corn protein zein to evaluate the combined effects of bioactive glass and Manuka honey in a new type of scaffold. According to the results of Arango-Ospina et al. [97], Manuka honey and zein coatings imparted antibacterial properties and excellent mechanical properties to bioactive glass bone tissue scaffolds.
From the information mentioned above it is clear that honey may serve as a promising therapeutic supplement for the prevention and management of osteoporosis. Anyway, more scientific or epidemiological evidence is needed for the use of any type of honey in the treatment of postmenopausal osteoporosis in women due to the limited number of clinical trials. Summary data from the aforementioned research is presented in Table 1.

4. Honey and Breast Cancer

Honey as a potential preventive and therapeutic supplement is currently gaining attention in cancer research. Various studies have been reported to investigate the anticancer benefits of different types of honey from different origins. The anticancer activity of honey has been demonstrated against various cancer cell lines and tissues, such as breast, prostate, colorectal, endometrial, and renal [98,99,100,101,102,103,104,105,106]. In general, the chemo-preventive properties of honey are consistent with its bioactive compounds, mostly quercetin, luteolin, chrysin, and esters of caffeic [107]. Although the exact mechanism is still unclear, some studies revealed the interference of bioactive compounds with anti-proliferative [108], antioxidant [109], and pro-apoptotic cell-signaling pathways [110]. Choi et al. [111] documented the anti-proliferative effects of quercetin in the human breast cancer cell line MCF-7 by inhibiting cell cycle progression via transient accumulation in the M phase followed by G2 arrest. Moreover, quercetin treatment activated apoptosis in MCF-7 cells via the p38MAPK signaling pathway [112]. Kim et al. [113] detected melatonin and its metabolites in honey, which possess strong free radical scavenging properties [114]. However, high concentrations of melatonin can induce the production of ROS, leading to apoptosis in a variety of cancers [115,116,117]. In general, favorable impacts of honey against breast cancer have been proven in both preclinical and clinical studies.
Tualang honey has been found to induce apoptosis of MDA-MB-231 and MCF-7 breast cancer cells through activation of the mitochondrial apoptotic pathway by elevating caspase-3/7 and caspase-9 and reducing mitochondrial membrane potential [102]. Moreover, Tualang honey combined with tamoxifen enhanced the anticancer activity of tamoxifen, activated multiple caspase enzymes, and increased mitochondrial membrane depolarization, leading to a breast cancer cell (MCF-7 and MDA-MB-231) apoptosis [118]. Tualang honey with tamoxifen can therefore be used as an alternative for the treatment of breast cancer, thereby reducing the required dose of tamoxifen and subsequently eliminating the side effects of tamoxifen. According to Kadir et al. [119], the growth of DMBA-induced mammary tumors was inhibited by Tualang honey administration (0.2–2 g/kg for 150 days) in rats. Additionally, vascular endothelial growth factor (VEGF), a pro-angiogenic factor, was reduced in honey-supplemented rats. In the study of Zakaria et al. [120], elevated levels of alanine aminotransferase were determined in postmenopausal women with breast cancer compared to those consuming Tualang honey (20 g/day for 12 weeks). Moreover, an increase in creatinine levels, leukocyte, and platelet counts was observed in the honey-treated group. In a clinical trial by Hizan et al. [121], the combination of Tualang honey with the aromatase inhibitor anastrozole lowered background parenchyma enhancement (a correlate of cancer relapse) more efficiently than anastrozole treatment alone (42% vs. 10% reduction) in patients with ER-positive breast cancer.
The anti-proliferative impact of Manuka honey was determined in MDA-MB-231 and MCF-7 breast cancer cells and was time- and dose-dependent. Moreover, the IL-6/STAT3 signaling pathway was highlighted as one of the first potential targets for Manuka honey-induced breast cancer cell suppression [122]. In another study, Aryappalli et al. [123] found that inhibition of tyrosine-phosphorylated STAT3 in breast cancer cells by Manuka honey is mediated by selective antagonism of the IL-6 receptor. Ahmed et al. [124] revealed that supplementation with both Manuka and Tualang kinds of honey (1.0 g/kg for 120 days) was able to reduce tumor volume, numbers, weight, and growth rate in the 1-methyl-1-nitrosourea (MNU)-induced breast cancer in rats. In addition, a higher expression of pro-apoptotic proteins and lowered expression of anti-apoptotic proteins were recorded. These types of honey administered orally exhibit anticancer effects by modulating the immune system and activating the intrinsic apoptotic pathway.
Greek honey extract (pine, thyme, and fir) reduced the viability of MCF-7 breast cancer cells [100,125], while thyme honey inhibited the progression of MCF-7 cells by suppressing estrogenic impacts [100]. Anatolian honey with different botanical origins (pine, chestnut, and cedar) produced stronger inhibitory effects on MDA-MB-231, MCF-7, and SKBR3 breast cancer cells in a time- and dose-dependent manner [126]. In MCF-7 and MDA-MB-231 cancer cells, the aforementioned types of honey suppressed breast cancer through the IL-6/STAT3 signaling pathway.
Kurniawan et al. [127] examined the impact of apis Dorsata honey (two tablespoons orally, 3 times/day for 15 days) on IL-3 (multi-potential hematopoietic growth factor) levels in breast cancer patients undergoing chemotherapy. These authors determined increased levels of IL-3 in the honey-treated group compared to the control group. The effect of Dorsata honey on IL-6 (breast cancer metastases factor) levels and T lymphocytes in post-chemotherapy breast cancer individuals was investigated by Syam et al. [107]. It has been found that there is a significant increase in the levels of T lymphocytes, which can indirectly enhance the immune system and inhibit tumor cell growth in honey-treated patients with breast cancer. The results also showed that Dorsata honey consumption did not affect IL-6 levels in contrast to the Manuka honey, where differences were noted.
The ability of honey to mitigate the chemo- and radiotherapy-induced oral mucositis (OM) was documented in numerous studies that mainly involved patients with head and neck cancers [128]. The studies on honey-treatment toxicity associations are limited in breast cancer but a pilot randomized trial comprising breast cancer patients receiving doxorubicin and cyclophosphamide reported the clinical efficacy of propolis plus bicarbonate in OM prevention [129]. According to Aghamohammadi et al. [130], a mixture of honey (30 g) and cinnamon (4 g) powder administered to breast cancer patients three times a day for 1 week led to a significant improvement in overall health and quality of life after the treatment.
Although honey supplementation has been associated with breast cancer modification in most of the experimental studies mentioned above, further experiments (especially animal studies and prospective randomized clinical trials) are still needed to evaluate the potential usefulness of honey as a therapeutic supplement in prevention and management of breast cancer. Table 2 provides summary data from the aforementioned studies.

5. Conclusions

Nowadays, the administration of dietary supplements and functional food intake in standard care of osteoporotic and oncological patients is gaining more attention. Honey is one of the oldest organic natural substances used for medical purposes. Many studies have pointed to the antioxidant, antibacterial, antiviral, immunomodulatory, anti-inflammatory, hypocholesterolemic, hypotensive, and antitumor impacts of honey, making it beneficial for human health.
In this review, the current knowledge from in vitro, animal, and human studies concerning the use of honey as a potential therapeutic supplement for osteoporosis and breast cancer is presented, due to their increasing incidence in postmenopausal women. Preclinical studies related to osteoporosis have reported favorable effects of honey on cortical and trabecular bone microstructure, bone strength, and oxidative stress. The limited number of clinical trials suggests the need for further research to evaluate the potential benefits of honey in the treatment of postmenopausal osteoporosis. In relation to breast cancer, in vitro experiments revealed the anti-proliferative and pro-apoptotic impact of honey on breast cancer cells, as well as their increased apoptosis. Animal studies have shown that honey reduces the number, growth rate, volume, and tumor weight. Findings from clinical trials reported its immunomodulatory properties showing that honey is effective in increasing leukocyte and platelet counts, IL-3 levels, and quality of life. In this context, the potential role of honey and its oligosaccharides as prebiotics for specific beneficial bacteria might be examined in future clinical studies.
In conclusion, we can state that honey represents a prospective therapeutic supplement for bone and breast tissue health. However, several issues need to be addressed before administration, including the presence of allergens or pesticides, antibiotics, and contaminants. Since the existing differences among honey types, precise identification and quantification of bioactive compound content should be provided in detail. In addition, clinical studies published so far are limited by a small sample size without the involvement of all different ethnicities, a single dose of honey and often a short duration of experiments and different parameters analyzed. Therefore, further clinical trials should also be aimed at eliminating these shortcomings. Importantly, large-scale placebo-controlled clinical studies concerning nutrigenomics are highly warranted to evaluate the effects of honey with its bioactive components on global gene and protein expression.

Author Contributions

Conceptualization, M.M. and R.O.; methodology, M.M., V.K. and R.O.; formal analysis, V.M., N.Z., M.B. and R.B; writing-original draft preparation, M.M., V.K., S.C. and R.O.; writing-review and editing, M.M., S.C. and R.O.; visualization, V.M., N.Z., M.B. and R.B.; supervision, M.M. and R.O.; funding acquisition, M.M. and R.O. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Ministry of Education, Science, Research and Sport of the Slovak Republic, grant number VEGA 1/0444/20 and KEGA 012UKF-4/2023.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Muhammad, A.; Mada, S.B.; Malami, I.; Forcados, G.E.; Erukainure, O.L.; Sani, H.; Abubakar, I.B. Postmenopausal osteoporosis and breast cancer: The biochemical links and beneficial effects of functional foods. Biomed. Pharmacother. 2018, 107, 571–582. [Google Scholar] [CrossRef] [PubMed]
  2. Kamaruzzaman, M.A.; Chin, K.-Y.; Mohd Ramli, E.S. A Review of Potential Beneficial Effects of Honey on Bone Health. Evid. Based Complement. Altern. Med. 2019, 2019, e8543618. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Badeau, M.; Adlercreutz, H.; Kaihovaara, P.; Tikkanen, M.J. Estrogen A-ring structure and antioxidative effect on lipoproteins. J. Steroid Biochem. Mol. Biol. 2005, 96, 271–278. [Google Scholar] [CrossRef] [PubMed]
  4. Martiniakova, M.; Babikova, M.; Mondockova, V.; Blahova, J.; Kovacova, V.; Omelka, R. The Role of Macronutrients, Micronutrients and Flavonoid Polyphenols in the Prevention and Treatment of Osteoporosis. Nutrients 2022, 14, 523. [Google Scholar] [CrossRef] [PubMed]
  5. Teng, G.G.; Curtis, J.R.; Saag, K.G. Mortality and osteoporotic fractures: Is the link causal, and is it modifiable? Clin. Exp. Rheumatol. 2008, 26, S125–S137. [Google Scholar] [PubMed]
  6. 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. [Google Scholar] [PubMed]
  7. Ahmed, S.; Sulaiman, S.A.; Baig, A.A.; Ibrahim, M.; Liaqat, S.; Fatima, S.; Jabeen, S.; Shamim, N.; Othman, N.H. Honey as a Potential Natural Antioxidant Medicine: An Insight into Its Molecular Mechanisms of Action. Oxidative Med. Cell. Longev. 2018, 2018, e8367846. [Google Scholar] [CrossRef] [Green Version]
  8. Kassim, M.; Achoui, M.; Mustafa, M.R.; Mohd, M.A.; Yusoff, K.M. Ellagic acid, phenolic acids, and flavonoids in Malaysian honey extracts demonstrate in vitro anti-inflammatory activity. Nutr. Res. 2010, 30, 650–659. [Google Scholar] [CrossRef]
  9. Hussein, S.Z.; Mohd Yusoff, K.; Makpol, S.; Mohd Yusof, Y.A. Gelam Honey Inhibits the Production of Proinflammatory, Mediators NO, PGE(2), TNF-α, and IL-6 in Carrageenan-Induced Acute Paw Edema in Rats. Evid. Based Complement. Altern. Med. 2012, 2012, 109636. [Google Scholar] [CrossRef] [Green Version]
  10. Owoyele, B.V.; Adenekan, O.T.; Soladoye, A.O. Effects of honey on inflammation and nitric oxide production in Wistar rats. Zhong Xi Yi Jie He Xue Bao J. Chin. Integr. Med. 2011, 9, 447–452. [Google Scholar] [CrossRef]
  11. Afroz, R.; Tanvir, E.M.; Paul, S.; Bhoumik, N.C.; Gan, S.H.; Khalil, M.I. DNA Damage Inhibition Properties of Sundarban Honey and its Phenolic Composition. J. Food Biochem. 2016, 40, 436–445. [Google Scholar] [CrossRef]
  12. Hussein, S.Z.; Mohd Yusoff, K.; Makpol, S.; Mohd Yusof, Y.A. Gelam honey attenuates carrageenan-induced rat paw inflammation via NF-κB pathway. PLoS ONE 2013, 8, e72365. [Google Scholar] [CrossRef] [Green Version]
  13. Desroches, S.; Lamarche, B. The evolving definitions and increasing prevalence of the metabolic syndrome. Appl. Physiol. Nutr. Metab. 2007, 32, 23–32. [Google Scholar] [CrossRef] [PubMed]
  14. Thapa, S.; Nandy, A.; Rendina-Ruedy, E. Endocrinal metabolic regulation on the skeletal system in post-menopausal women. Front. Physiol. 2022, 13, 1052429. [Google Scholar] [CrossRef]
  15. Nestel, P.; Lyu, R.; Low, L.P.; Sheu, W.H.-H.; Nitiyanant, W.; Saito, I.; Tan, C.E. Metabolic syndrome: Recent prevalence in East and Southeast Asian populations. Asia Pac. J. Clin. Nutr. 2007, 16, 362–367. [Google Scholar]
  16. Lear, S.A.; Gasevic, D. Ethnicity and Metabolic Syndrome: Implications for Assessment, Management and Prevention. Nutrients 2019, 12, 15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  17. Qadirifard, M.S.; Fathabadi, A.; Hajishah, H.; Gholami, K.; Abbasi, M.; Sami, N.; Zo, M.M.; Kadkhodaei, F.; Sina, M.; Ansari, A.; et al. Anti-breast cancer potential of honey: A narrative review. OncoReview 2022, 12, 5–15. [Google Scholar] [CrossRef]
  18. Martiniakova, M.; Babikova, M.; Omelka, R. Pharmacological agents and natural compounds: Available treatments for osteoporosis. J. Physiol. Pharmacol. 2020, 71, 307–320. [Google Scholar] [CrossRef]
  19. Chin, K.-Y.; Wong, S.K.; Ekeuku, S.O.; Pang, K.-L. Relationship Between Metabolic Syndrome and Bone Health—An Evaluation of Epidemiological Studies and Mechanisms Involved. Diabetes Metab. Syndr. Obes. 2020, 13, 3667–3690. [Google Scholar] [CrossRef] [PubMed]
  20. Luchese, R.H.; Prudêncio, E.R.; Guerra, A.F.; Luchese, R.H.; Prudêncio, E.R.; Guerra, A.F. Honey as a Functional Food; IntechOpen: Rijeka, Croatia, 2017; ISBN 978-953-51-2880-9. [Google Scholar]
  21. Blahova, J.; Martiniakova, M.; Babikova, M.; Kovacova, V.; Mondockova, V.; Omelka, R. Pharmaceutical Drugs and Natural Therapeutic Products for the Treatment of Type 2 Diabetes Mellitus. Pharmaceuticals 2021, 14, 806. [Google Scholar] [CrossRef]
  22. Sidor, E.; Dżugan, M. Drone Brood Homogenate as Natural Remedy for Treating Health Care Problem: A Scientific and Practical Approach. Molecules 2020, 25, 5699. [Google Scholar] [CrossRef] [PubMed]
  23. Martiniakova, M.; Blahova, J.; Kovacova, V.; Babikova, M.; Mondockova, V.; Kalafova, A.; Capcarova, M.; Omelka, R. Bee Bread Can Alleviate Lipid Abnormalities and Impaired Bone Morphology in Obese Zucker Diabetic Rats. Molecules 2021, 26, 2616. [Google Scholar] [CrossRef] [PubMed]
  24. Mohd Ramli, E.S.; Sukalingam, K.; Kamaruzzaman, M.A.; Soelaiman, I.N.; Pang, K.-L.; Chin, K.-Y. Direct and Indirect Effect of Honey as a Functional Food Against Metabolic Syndrome and Its Skeletal Complications. Diabetes Metab. Syndr. Obes. 2021, 14, 241–256. [Google Scholar] [CrossRef] [PubMed]
  25. Tomczyk, M.; Bocian, A.; Sidor, E.; Miłek, M.; Zaguła, G.; Dżugan, M. The Use of HPTLC and SDS-PAGE Methods for Coniferous Honeydew Honey Fingerprinting Compiled with Mineral Content and Antioxidant Activity. Molecules 2022, 27, 720. [Google Scholar] [CrossRef]
  26. Alvarez-Suarez, J.M.; Tulipani, S.; Romandini, S.; Bertoli, E.; Battino, M. Contribution of honey in nutrition and human health: A review. Mediterr. J. Nutr. Metab. 2010, 3, 15–23. [Google Scholar] [CrossRef]
  27. da Silva, P.M.; Gauche, C.; Gonzaga, L.V.; Costa, A.C.O.; Fett, R. Honey: Chemical composition, stability and authenticity. Food Chem. 2016, 196, 309–323. [Google Scholar] [CrossRef]
  28. Cianciosi, D.; Forbes-Hernández, T.Y.; Afrin, S.; Gasparrini, M.; Reboredo-Rodriguez, P.; Manna, P.P.; Zhang, J.; Bravo Lamas, L.; Martínez Flórez, S.; Agudo Toyos, P.; et al. Phenolic Compounds in Honey and Their Associated Health Benefits: A Review. Molecules 2018, 23, 2322. [Google Scholar] [CrossRef] [Green Version]
  29. Miguel, M.; Antunes, M.; Faleiro, M. Honey as a Complementary Medicine. Integr. Med. Insights 2017, 12, 1178633717702869. [Google Scholar] [CrossRef] [Green Version]
  30. Yudaniayanti, I.S.; Primarizky, H.; Nangoi, L. The effects of honey (Apis dorsata) supplements on increased bone strength in ovariectomized rat as animal model of osteoporosis. AIP Conf. Proc. 2018, 1945, 020004. [Google Scholar] [CrossRef]
  31. Boman, A.; Kokkonen, H.; Ärlestig, L.; Berglin, E.; Rantapää-Dahlqvist, S. Receptor activator of nuclear factor kappa-B ligand (RANKL) but not sclerostin or gene polymorphisms is related to joint destruction in early rheumatoid arthritis. Clin. Rheumatol. 2017, 36, 1005–1012. [Google Scholar] [CrossRef] [Green Version]
  32. Hofbauer, L.C.; Rachner, T.D.; Hamann, C. From bone to breast and back—The bone cytokine RANKL and breast cancer. Breast Cancer Res. 2011, 13, 107. [Google Scholar] [CrossRef] [Green Version]
  33. Park, J.H.; Lee, N.K.; Lee, S.Y. Current Understanding of RANK Signaling in Osteoclast Differentiation and Maturation. Mol. Cells 2017, 40, 706–713. [Google Scholar] [CrossRef] [Green Version]
  34. Fata, J.E.; Kong, Y.Y.; Li, J.; Sasaki, T.; Irie-Sasaki, J.; Moorehead, R.A.; Elliott, R.; Scully, S.; Voura, E.B.; Lacey, D.L.; et al. The osteoclast differentiation factor osteoprotegerin-ligand is essential for mammary gland development. Cell 2000, 103, 41–50. [Google Scholar] [CrossRef] [Green Version]
  35. Gonzalez-Suarez, E.; Jacob, A.P.; Jones, J.; Miller, R.; Roudier-Meyer, M.P.; Erwert, R.; Pinkas, J.; Branstetter, D.; Dougall, W.C. RANK ligand mediates progestin-induced mammary epithelial proliferation and carcinogenesis. Nature 2010, 468, 103–107. [Google Scholar] [CrossRef]
  36. Trémollieres, F.A. Screening for osteoporosis after breast cancer: For whom, why and when. Maturitas 2014, 79, 343–348. [Google Scholar] [CrossRef] [PubMed]
  37. Cauley, J.A. Estrogen and bone health in men and women. Steroids 2015, 99, 11–15. [Google Scholar] [CrossRef] [PubMed]
  38. Manolagas, S.C. From estrogen-centric to aging and oxidative stress: A revised perspective of the pathogenesis of osteoporosis. Endocr. Rev. 2010, 31, 266–300. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Yue, W.; Wang, J.-P.; Li, Y.; Fan, P.; Liu, G.; Zhang, N.; Conaway, M.; Wang, H.; Korach, K.S.; Bocchinfuso, W.; et al. Effects of estrogen on breast cancer development: Role of estrogen receptor independent mechanisms. Int. J. Cancer 2010, 127, 1748–1757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Wen, C.; Wu, L.; Fu, L.; Wang, B.; Zhou, H. Unifying mechanism in the initiation of breast cancer by metabolism of estrogen (Review). Mol. Med. Rep. 2017, 16, 1001–1006. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Taxel, P.; Choksi, P.; Poznak, C.V. The Management of Osteoporosis in Breast Cancer Survivors. Maturitas 2012, 73, 275–279. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Trémollieres, F.A.; Ceausu, I.; Depypere, H.; Lambrinoudaki, I.; Mueck, A.; Pérez-López, F.R.; van der Schouw, Y.T.; Senturk, L.M.; Simoncini, T.; Stevenson, J.C.; et al. Osteoporosis management in patients with breast cancer: EMAS position statement. Maturitas 2017, 95, 65–71. [Google Scholar] [CrossRef] [Green Version]
  43. Mirończuk-Chodakowska, I.; Witkowska, A.M.; Zujko, M.E. Endogenous non-enzymatic antioxidants in the human body. Adv. Med. Sci. 2018, 63, 68–78. [Google Scholar] [CrossRef]
  44. Rao, L.G.; Rao, A.V.; Rao, L.G.; Rao, A.V. Oxidative Stress and Antioxidants in the Risk of Osteoporosis—Role of the Antioxidants Lycopene and Polyphenols; IntechOpen: Rijeka, Croatia, 2013; ISBN 978-953-51-1066-8. [Google Scholar]
  45. Callaway, D.A.; Jiang, J.X. Reactive oxygen species and oxidative stress in osteoclastogenesis, skeletal aging and bone diseases. J. Bone Miner. Metab. 2015, 33, 359–370. [Google Scholar] [CrossRef] [PubMed]
  46. Li, D.-Z.; Zhang, Q.-X.; Dong, X.-X.; Li, H.-D.; Ma, X. Treatment with hydrogen molecules prevents RANKL-induced osteoclast differentiation associated with inhibition of ROS formation and inactivation of MAPK, AKT and NF-kappa B pathways in murine RAW264.7 cells. J. Bone Miner. Metab. 2014, 32, 494–504. [Google Scholar] [CrossRef] [PubMed]
  47. Kanzaki, H.; Shinohara, F.; Kanako, I.; Yamaguchi, Y.; Fukaya, S.; Miyamoto, Y.; Wada, S.; Nakamura, Y. Molecular regulatory mechanisms of osteoclastogenesis through cytoprotective enzymes. Redox Biol. 2016, 8, 186–191. [Google Scholar] [CrossRef] [Green Version]
  48. Zhang, B.; Xie, Q.; Quan, Y.; Pan, X.; Liao, D. Reactive oxygen species induce cell death via Akt signaling in rat osteoblast-like cell line ROS 17/2.8. Toxicol. Ind. Health 2015, 31, 1236–1242. [Google Scholar] [CrossRef] [PubMed]
  49. Ozgocmen, S.; Kaya, H.; Fadillioglu, E.; Aydogan, R.; Yilmaz, Z. Role of antioxidant systems, lipid peroxidation, and nitric oxide in postmenopausal osteoporosis. Mol. Cell. Biochem. 2007, 295, 45–52. [Google Scholar] [CrossRef]
  50. van’t Hof, R.J.; Ralston, S.H. Cytokine-induced nitric oxide inhibits bone resorption by inducing apoptosis of osteoclast progenitors and suppressing osteoclast activity. J. Bone Miner. Res. 1997, 12, 1797–1804. [Google Scholar] [CrossRef]
  51. MacPherson, H.; Noble, B.S.; Ralston, S.H. Expression and functional role of nitric oxide synthase isoforms in human osteoblast-like cells. Bone 1999, 24, 179–185. [Google Scholar] [CrossRef] [PubMed]
  52. Hecht, F.; Pessoa, C.F.; Gentile, L.B.; Rosenthal, D.; Carvalho, D.P.; Fortunato, R.S. The role of oxidative stress on breast cancer development and therapy. Tumour Biol. 2016, 37, 4281–4291. [Google Scholar] [CrossRef] [PubMed]
  53. Yu, T.; Di, G. Role of tumor microenvironment in triple-negative breast cancer and its prognostic significance. Chin. J. Cancer Res. 2017, 29, 237–252. [Google Scholar] [CrossRef]
  54. Chen, Y.; Zeng, C.; Zhan, Y.; Wang, H.; Jiang, X.; Li, W. Aberrant low expression of p85α in stromal fibroblasts promotes breast cancer cell metastasis through exosome-mediated paracrine Wnt10b. Oncogene 2017, 36, 4692–4705. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Bussard, K.M.; Mutkus, L.; Stumpf, K.; Gomez-Manzano, C.; Marini, F.C. Tumor-associated stromal cells as key contributors to the tumor microenvironment. Breast Cancer Res. 2016, 18, 84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Jezierska-Drutel, A.; Rosenzweig, S.A.; Neumann, C.A. Role of oxidative stress and the microenvironment in breast cancer development and progression. Adv. Cancer Res. 2013, 119, 107–125. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Flegar, D.; Šućur, A.; Markotić, A.; Kovačić, N.; Grčević, D. Osteoporosis in the view of osteoimmunology: Common feature underlined by different pathogenic mechanisms. Period. Biol. 2015, 117, 35–43. [Google Scholar]
  58. Pietschmann, P.; Mechtcheriakova, D.; Meshcheryakova, A.; Föger-Samwald, U.; Ellinger, I. Immunology of Osteoporosis: A Mini-Review. Gerontology 2016, 62, 128–137. [Google Scholar] [CrossRef] [Green Version]
  59. Tencerova, M.; Kassem, M. The Bone Marrow-Derived Stromal Cells: Commitment and Regulation of Adipogenesis. Front. Endocrinol 2016, 7, 127. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Souza, P.P.C.; Lerner, U.H. The role of cytokines in inflammatory bone loss. Immunol. Investig. 2013, 42, 555–622. [Google Scholar] [CrossRef]
  61. Kaur, R.P.; Rubal; Banipal, R.P.S.; Vashistha, R.; Dhiman, M.; Munshi, A. Association of elevated levels of C-reactive protein with breast cancer, breast cancer subtypes, and poor outcome. Curr. Probl. Cancer 2019, 43, 123–129. [Google Scholar] [CrossRef] [PubMed]
  62. Kozłowski, L.; Zakrzewska, I.; Tokajuk, P.; Wojtukiewicz, M.Z. Concentration of interleukin-6 (IL-6), interleukin-8 (IL-8) and interleukin-10 (IL-10) in blood serum of breast cancer patients. Rocz. Akad. Med. Bialymst. 2003, 48, 82–84. [Google Scholar]
  63. Nicolini, A.; Carpi, A.; Rossi, G. Cytokines in breast cancer. Cytokine Growth Factor Rev. 2006, 17, 325–337. [Google Scholar] [CrossRef] [PubMed]
  64. García-Tuñón, I.; Ricote, M.; Ruiz, A.; Fraile, B.; Paniagua, R.; Royuela, M. Role of tumor necrosis factor-alpha and its receptors in human benign breast lesions and tumors (in situ and infiltrative). Cancer Sci. 2006, 97, 1044–1049. [Google Scholar] [CrossRef] [PubMed]
  65. Zakaria, Z.; Hamid, W.Z.W.A.; Mohamed, M. Tualang honey supplementation improves inflammatory and bone markers among postmenopausal breast cancer patients : A randomised controlled trial. Sains Malays. 2021, 50, 1971–1985. [Google Scholar] [CrossRef]
  66. Kopelman, P.G. Obesity as a medical problem. Nature 2000, 404, 635–643. [Google Scholar] [CrossRef]
  67. Villareal, D.T.; Apovian, C.M.; Kushner, R.F.; Klein, S.; American Society for Nutrition; NAASO, The Obesity Society. Obesity in older adults: Technical review and position statement of the American Society for Nutrition and NAASO, The Obesity Society. Am. J. Clin. Nutr. 2005, 82, 923–934. [Google Scholar] [CrossRef] [Green Version]
  68. Bonewald, L.F.; Johnson, M.L. Osteocytes, mechanosensing and Wnt signaling. Bone 2008, 42, 606–615. [Google Scholar] [CrossRef] [Green Version]
  69. Sawakami, K.; Robling, A.G.; Ai, M.; Pitner, N.D.; Liu, D.; Warden, S.J.; Li, J.; Maye, P.; Rowe, D.W.; Duncan, R.L.; et al. The Wnt co-receptor LRP5 is essential for skeletal mechanotransduction but not for the anabolic bone response to parathyroid hormone treatment. J. Biol. Chem. 2006, 281, 23698–23711. [Google Scholar] [CrossRef] [Green Version]
  70. Ehrlich, P.J.; Lanyon, L.E. Mechanical strain and bone cell function: A review. Osteoporos. Int. 2002, 13, 688–700. [Google Scholar] [CrossRef]
  71. Hsu, Y.-H.; Venners, S.A.; Terwedow, H.A.; Feng, Y.; Niu, T.; Li, Z.; Laird, N.; Brain, J.D.; Cummings, S.R.; Bouxsein, M.L.; et al. Relation of body composition, fat mass, and serum lipids to osteoporotic fractures and bone mineral density in Chinese men and women. Am. J. Clin. Nutr. 2006, 83, 146–154. [Google Scholar] [CrossRef] [Green Version]
  72. Pollock, N.K.; Laing, E.M.; Baile, C.A.; Hamrick, M.W.; Hall, D.B.; Lewis, R.D. Is adiposity advantageous for bone strength? A peripheral quantitative computed tomography study in late adolescent females. Am. J. Clin. Nutr. 2007, 86, 1530–1538. [Google Scholar] [CrossRef] [Green Version]
  73. Lauby-Secretan, B.; Scoccianti, C.; Loomis, D.; Grosse, Y.; Bianchini, F.; Straif, K.; International Agency for Research on Cancer Handbook Working Group. Body Fatness and Cancer--Viewpoint of the IARC Working Group. N. Engl. J. Med. 2016, 375, 794–798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Ewertz, M.; Jensen, M.-B.; Gunnarsdóttir, K.Á.; Højris, I.; Jakobsen, E.H.; Nielsen, D.; Stenbygaard, L.E.; Tange, U.B.; Cold, S. Effect of obesity on prognosis after early-stage breast cancer. J. Clin. Oncol. 2011, 29, 25–31. [Google Scholar] [CrossRef] [PubMed]
  75. Cheung, A.M.; Heisey, R.; Srighanthan, J. Breast cancer and osteoporosis. Curr. Opin. Endocrinol. Diabetes Obes. 2013, 20, 532–538. [Google Scholar] [CrossRef] [PubMed]
  76. Brown, K.A. Metabolic pathways in obesity-related breast cancer. Nat. Rev. Endocrinol. 2021, 17, 350–363. [Google Scholar] [CrossRef]
  77. Pfeiler, G.; Königsberg, R.; Hadji, P.; Fitzal, F.; Maroske, M.; Dressel-Ban, G.; Zellinger, J.; Exner, R.; Seifert, M.; Singer, C.; et al. Impact of body mass index on estradiol depletion by aromatase inhibitors in postmenopausal women with early breast cancer. Br. J. Cancer 2013, 109, 1522–1527. [Google Scholar] [CrossRef] [Green Version]
  78. Fournier, A.; Mesrine, S.; Gelot, A.; Fagherazzi, G.; Baglietto, L.; Clavel-Chapelon, F.; Boutron-Ruault, M.-C.; Chabbert-Buffet, N. Use of Bisphosphonates and Risk of Breast Cancer in a French Cohort of Postmenopausal Women. J. Clin. Oncol. 2017, 35, 3230–3239. [Google Scholar] [CrossRef]
  79. Rennert, G.; Pinchev, M.; Gronich, N.; Saliba, W.; Flugelman, A.; Lavi, I.; Goldberg, H.; Fried, G.; Steiner, M.; Bitterman, A.; et al. Oral Bisphosphonates and Improved Survival of Breast Cancer. Clin. Cancer Res. 2017, 23, 1684–1689. [Google Scholar] [CrossRef]
  80. Kim, T.-K.; Atigadda, V.; Brzeminski, P.; Fabisiak, A.; Tang, E.K.Y.; Tuckey, R.C.; Slominski, A.T. Detection of 7-Dehydrocholesterol and Vitamin D3 Derivatives in Honey. Molecules 2020, 25, 2583. [Google Scholar] [CrossRef]
  81. Slominski, A.T.; Chaiprasongsuk, A.; Janjetovic, Z.; Kim, T.-K.; Stefan, J.; Slominski, R.M.; Hanumanthu, V.S.; Raman, C.; Qayyum, S.; Song, Y.; et al. Photoprotective Properties of Vitamin D and Lumisterol Hydroxyderivatives. Cell Biochem. Biophys. 2020, 78, 165–180. [Google Scholar] [CrossRef]
  82. Omelka, R.; Martiniakova, M.; Svik, K.; Slovak, L.; Payer, J.; Oppenbergerova, I.; Kovacova, V.; Babikova, M.; Soltesova-Prnova, M. The effects of eggshell calcium (Biomin H®) and its combinations with alfacalcidol (1α-hydroxyvitamin D3) and menaquinone-7 (vitamin K2) on ovariectomy-induced bone loss in a rat model of osteoporosis. J. Anim. Physiol. Anim. Nutr. 2021, 105, 336–344. [Google Scholar] [CrossRef]
  83. Kazemian, E.; Pourali, A.; Sedaghat, F.; Karimi, M.; Basirat, V.; Sajadi Hezaveh, Z.; Davoodi, S.H.; Holick, M.F. Effect of supplemental vitamin D3 on bone mineral density: A systematic review and meta-analysis. Nutr. Rev. 2022, nuac068. [Google Scholar] [CrossRef]
  84. Iwamoto, J.; Takeda, T.; Ichimura, S. Effect of combined administration of vitamin D3 and vitamin K2 on bone mineral density of the lumbar spine in postmenopausal women with osteoporosis. J. Orthop. Sci. 2000, 5, 546–551. [Google Scholar] [CrossRef]
  85. Zaid, S.S.M.; Sulaiman, S.A.; Othman, N.H.; Soelaiman, I.-N.; Shuid, A.N.; Mohamad, N.; Muhamad, N. Protective effects of Tualang honey on bone structure in experimental postmenopausal rats. Clinics 2012, 67, 779–784. [Google Scholar] [CrossRef]
  86. Ahmed, S.; Othman, N.H. Review of the Medicinal Effects of Tualang Honey and a Comparison with Manuka Honey. Malays. J. Med. Sci. 2013, 20, 6–13. [Google Scholar] [PubMed]
  87. Zaid, S.S.; Sulaiman, S.A.; Sirajudeen, K.N.; Othman, N.H. The effects of tualang honey on female reproductive organs, tibia bone and hormonal profile in ovariectomised rats—Animal model for menopause. BMC Complement. Altern. Med. 2010, 10, 82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Husniati, Y.L.; Hazlina, N.H.N.; Amrah, S.S.; Idiana, H.I.; Azidah, A.K.; Norhayati, M.N.; Bahari, I.S.; Rahimah, Z.; Shaniza, S.N.; Juhara, H.; et al. The effects of Tualang Honey on postmenopausal women. In Proceedings of the Journal of ApiProduct and ApiMedical Science, Kota Bharu, Malaysia, 13–15 January 2010; Volume 2, p. 33. [Google Scholar]
  89. Shafin, N.; Othman, Z.; Zakaria, R.; Nik Hussain, N.H. Tualang Honey Supplementation Reduces Blood Oxidative Stress Levels/Activities in Postmenopausal Women. Int. Sch. Res. Not. 2014, 2014, e364836. [Google Scholar] [CrossRef] [Green Version]
  90. Hussein, S.Z.; Yusoff, K.M.; Makpol, S.; Yusof, Y.A.M. Antioxidant Capacities and Total Phenolic Contents Increase with Gamma Irradiation in Two Types of Malaysian Honey. Molecules 2011, 16, 6378–6395. [Google Scholar] [CrossRef] [PubMed]
  91. Mohd Effendy, N.; Mohamed, N.; Muhammad, N.; Mohamad, I.N.; Shuid, A.N. The Effects of Tualang Honey on Bone Metabolism of Postmenopausal Women. Evid. Based Complement. Altern. Med. 2012, 2012, e938574. [Google Scholar] [CrossRef]
  92. Moniruzzaman, M.; Khalil, M.I.; Sulaiman, S.A.; Gan, S.H. Physicochemical and antioxidant properties of Malaysian honeys produced by Apis cerana, Apis dorsata and Apis mellifera. BMC Complement. Altern. Med. 2013, 13, 43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Kamaruzzaman, M.A.; Thanu, A.; Yusof, M.R.; Soelaiman, I.N.; Ramli, E.S. Kelulut honey ameliorates glucocorticoid induced osteoporosis via its antioxidant activity in rats. Asian Pac. J. Trop. Biomed. 2019, 9, 493. [Google Scholar] [CrossRef]
  94. Ekeuku, S.O.; Chin, K.-Y.; Ramli, N.Z.; Zarkasi, K.A.; Ahmad, F. Effect of Kelulut honey supplementation on bone health in male rats on high-carbohydrate high-fat diet. Trop. J. Pharm. Res. 2021, 20, 1185–1192. [Google Scholar] [CrossRef]
  95. Hasib, A.; Wahjuningrum, D.A.; Ibrahim, M.H.R.; Kurniawan, H.J.; Ernawati, R.; Hadinoto, M.E.K.; Mooduto, L. ALP (Alkaline Phosphatase) Expression in Simple Fracture Incident in Rat (Rattus norvegicus) Femur Bone Supplemented by Apis Mellifera Honey. J. Int. Dent. Med. Res. 2020, 13, 887–891. [Google Scholar]
  96. Abu-Serie, M.M.; Habashy, N.H. The ameliorating effect of the combined extract from Greek Thymus vulgaris and bee’s honey on the hydrocortisone-induced osteoporosis in rat bone cells via modulating the bone turnover, oxidative stress, and inflammation. RSC Adv. 2018, 8, 28341–28355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Arango-Ospina, M.; Lasch, K.; Weidinger, J.; Boccaccini, A.R. Manuka Honey and Zein Coatings Impart Bioactive Glass Bone Tissue Scaffolds Antibacterial Properties and Superior Mechanical Properties. Front. Mater. 2021, 7, 610889. [Google Scholar] [CrossRef]
  98. Tomasin, R.; Gomes-Marcondes, M.C.C. Oral administration of Aloe vera and honey reduces Walker tumour growth by decreasing cell proliferation and increasing apoptosis in tumour tissue. Phytother. Res. 2011, 25, 619–623. [Google Scholar] [CrossRef]
  99. Fukuda, M.; Kobayashi, K.; Hirono, Y.; Miyagawa, M.; Ishida, T.; Ejiogu, E.C.; Sawai, M.; Pinkerton, K.E.; Takeuchi, M. Jungle honey enhances immune function and antitumor activity. Evid. Based Complement. Altern. Med. 2011, 2011, 908743. [Google Scholar] [CrossRef] [PubMed]
  100. Tsiapara, A.V.; Jaakkola, M.; Chinou, I.; Graikou, K.; Tolonen, T.; Virtanen, V.; Moutsatsou, P. 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] [CrossRef]
  101. Gribel, N.V.; Pashinskiĭ, V.G. The antitumor properties of honey. Vopr. Onkol. 1990, 36, 704–709. [Google Scholar]
  102. Fauzi, A.N.; Norazmi, M.N.; Yaacob, N.S. 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. [Google Scholar] [CrossRef]
  103. Jaganathan, S.K. Honey Constituents and their apoptotic effect in colon cancer cells. J. ApiProduct ApiMedical Sci. 2009, 1, 29–36. [Google Scholar] [CrossRef]
  104. Samarghandian, S.; Afshari, J.T.; Davoodi, S. Honey induces apoptosis in renal cell carcinoma. Pharmacogn. Mag. 2011, 7, 46–52. [Google Scholar] [CrossRef] [Green Version]
  105. Ahmed, S.; Othman, N.H. Honey as a Potential Natural Anticancer Agent: A Review of Its Mechanisms. Evid. Based Complement. Altern. Med. 2013, 2013, e829070. [Google Scholar] [CrossRef] [Green Version]
  106. Afrin, S.; Haneefa, S.M.; Fernandez-Cabezudo, M.J.; Giampieri, F.; Al-Ramadi, B.K.; Battino, M. Therapeutic and preventive properties of honey and its bioactive compounds in cancer: An evidence-based review. Nutr. Res. Rev. 2020, 33, 50–76. [Google Scholar] [CrossRef]
  107. Syam, Y.; Prihantono, P.; Majid, S.; Sjattar, E.L.; Kana, M.; Usman, A.N. The effect of apis Dorsata honey as a complementary therapy to interleukin-6 (IL-6) levels and T lymphocytes of post-chemotherapy breast cancer patients. Breast Dis. 2021, 40, S97–S101. [Google Scholar] [CrossRef]
  108. Jaganathan, S.K.; Mandal, M. Antiproliferative effects of honey and of its polyphenols: A review. J. Biomed. Biotechnol. 2009, 2009, 830616. [Google Scholar] [CrossRef] [Green Version]
  109. Valko, M.; Leibfritz, D.; Moncol, J.; Cronin, M.T.D.; Mazur, M.; Telser, J. Free radicals and antioxidants in normal physiological functions and human disease. Int. J. Biochem. Cell Biol. 2007, 39, 44–84. [Google Scholar] [CrossRef] [PubMed]
  110. Jaganathan, S.K.; Mandal, M. Involvement of non-protein thiols, mitochondrial dysfunction, reactive oxygen species and p53 in honey-induced apoptosis. Investig. New Drugs 2010, 28, 624–633. [Google Scholar] [CrossRef] [PubMed]
  111. Choi, J.A.; Kim, J.Y.; Lee, J.Y.; Kang, C.M.; Kwon, H.J.; Yoo, Y.D.; Kim, T.W.; Lee, Y.S.; Lee, S.J. Induction of cell cycle arrest and apoptosis in human breast cancer cells by quercetin. Int. J. Oncol. 2001, 19, 837–844. [Google Scholar] [CrossRef] [PubMed]
  112. Ranganathan, S.; Halagowder, D.; Sivasithambaram, N.D. Quercetin Suppresses Twist to Induce Apoptosis in MCF-7 Breast Cancer Cells. PLoS ONE 2015, 10, e0141370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Kim, T.-K.; Fabisiak, A.; Brzeminski, P.; Reiter, R.J.; Slominski, A.T. Serotonin, melatonin and their precursors and metabolites and vitamin D3 derivatives in honey. Melatonin Res. 2022, 5, 374–380. [Google Scholar] [CrossRef]
  114. Florido, J.; Rodriguez-Santana, C.; Martinez-Ruiz, L.; López-Rodríguez, A.; Acuña-Castroviejo, D.; Rusanova, I.; Escames, G. Understanding the Mechanism of Action of Melatonin, Which Induces ROS Production in Cancer Cells. Antioxidants 2022, 11, 1621. [Google Scholar] [CrossRef] [PubMed]
  115. Moreira, A.J.; Ordoñez, R.; Cerski, C.T.; Picada, J.N.; García-Palomo, A.; Marroni, N.P.; Mauriz, J.L.; González-Gallego, J. Melatonin Activates Endoplasmic Reticulum Stress and Apoptosis in Rats with Diethylnitrosamine-Induced Hepatocarcinogenesis. PLoS ONE 2015, 10, e0144517. [Google Scholar] [CrossRef] [PubMed]
  116. Yun, S.-M.; Woo, S.H.; Oh, S.T.; Hong, S.-E.; Choe, T.-B.; Ye, S.-K.; Kim, E.-K.; Seong, M.K.; Kim, H.-A.; Noh, W.C.; et al. Melatonin enhances arsenic trioxide-induced cell death via sustained upregulation of Redd1 expression in breast cancer cells. Mol. Cell Endocrinol. 2016, 422, 64–73. [Google Scholar] [CrossRef]
  117. Guerra-Librero, A.; Fernandez-Gil, B.I.; Florido, J.; Martinez-Ruiz, L.; Rodríguez-Santana, C.; Shen, Y.-Q.; García-Verdugo, J.M.; López-Rodríguez, A.; Rusanova, I.; Quiñones-Hinojosa, A.; et al. Melatonin Targets Metabolism in Head and Neck Cancer Cells by Regulating Mitochondrial Structure and Function. Antioxidants 2021, 10, 603. [Google Scholar] [CrossRef] [PubMed]
  118. Yaacob, N.S.; Nengsih, A.; Norazmi, M.N. Tualang honey promotes apoptotic cell death induced by tamoxifen in breast cancer cell lines. Evid. Based Complement. Altern. Med. 2013, 2013, 989841. [Google Scholar] [CrossRef]
  119. Kadir, E.A.; Sulaiman, S.A.; Yahya, N.K.; Othman, N.H. Inhibitory effects of Tualang Honey on experimental breast cancer in rats: A preliminary study. Asian Pac. J. Cancer Prev. 2013, 14, 2249–2254. [Google Scholar] [CrossRef] [Green Version]
  120. Zakaria, Z.; Zainal Abidin, Z.F.; Gan, S.H.; Wan Abdul Hamid, W.Z.; Mohamed, M. Effects of honey supplementation on safety profiles among postmenopausal breast cancer patients. J. Taibah Univ. Med. Sci. 2018, 13, 535–540. [Google Scholar] [CrossRef]
  121. Hizan, N.S.; Hassan, N.H.M.; Haron, J.; Abubakar, M.B.; Mahdi, N.M.N.; Gan, S.H. Tualang honey adjunct with anastrozole improve parenchyma enhancement of breast tissue in breast cancer patients: A randomized controlled trial. Integr. Med. Res. 2018, 7, 322–327. [Google Scholar] [CrossRef]
  122. Aryappalli, P.; Al-Qubaisi, S.S.; Attoub, S.; George, J.A.; Arafat, K.; Ramadi, K.B.; Mohamed, Y.A.; Al-Dhaheri, M.M.; Al-Sbiei, A.; Fernandez-Cabezudo, M.J.; et al. The IL-6/STAT3 Signaling Pathway Is an Early Target of Manuka Honey-Induced Suppression of Human Breast Cancer Cells. Front. Oncol. 2017, 7, 167. [Google Scholar] [CrossRef] [Green Version]
  123. Aryappalli, P.; Shabbiri, K.; Masad, R.J.; Al-Marri, R.H.; Haneefa, S.M.; Mohamed, Y.A.; Arafat, K.; Attoub, S.; Cabral-Marques, O.; Ramadi, K.B.; et al. Inhibition of Tyrosine-Phosphorylated STAT3 in Human Breast and Lung Cancer Cells by Manuka Honey is Mediated by Selective Antagonism of the IL-6 Receptor. Int. J. Mol. Sci. 2019, 20, 4340. [Google Scholar] [CrossRef] [Green Version]
  124. Ahmed, S.; Sulaiman, S.A.; Othman, N.H. Oral Administration of Tualang and Manuka Honeys Modulates Breast Cancer Progression in Sprague-Dawley Rats Model. Evid. Based Complement. Altern. Med. 2017, 2017, 5904361. [Google Scholar] [CrossRef] [Green Version]
  125. Spilioti, E.; Jaakkola, M.; Tolonen, T.; Lipponen, M.; Virtanen, V.; Chinou, I.; Kassi, E.; Karabournioti, S.; Moutsatsou, P. Phenolic Acid Composition, Antiatherogenic and Anticancer Potential of Honeys Derived from Various Regions in Greece. PLoS ONE 2014, 9, e94860. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Seyhan, M.F.; Yılmaz, E.; Timirci-Kahraman, Ö.; Saygılı, N.; Kısakesen, H.İ.; Eronat, A.P.; Ceviz, A.B.; Bilgiç Gazioğlu, S.; Yılmaz-Aydoğan, H.; Öztürk, O. Anatolian honey is not only sweet but can also protect from breast cancer: Elixir for women from artemis to present. IUBMB Life 2017, 69, 677–688. [Google Scholar] [CrossRef]
  127. Kurniawan1, A.; Sampepajung1, D.; Syamsu1, S.A.; Prihantono1, P. Effectiveness of Dorsata Honey Suplement on Interleukin-3 Levels in Breast Cancer Patients Who Underwent Chemotherapy. Indian J. Public Health Res. Dev. 2020, 11, 947–952. [Google Scholar] [CrossRef]
  128. Yang, C.; Gong, G.; Jin, E.; Han, X.; Zhuo, Y.; Yang, S.; Song, B.; Zhang, Y.; Piao, C. Topical application of honey in the management of chemo/radiotherapy-induced oral mucositis: A systematic review and network meta-analysis. Int. J. Nurs. Stud. 2019, 89, 80–87. [Google Scholar] [CrossRef] [PubMed]
  129. Piredda, M.; Facchinetti, G.; Biagioli, V.; Giannarelli, D.; Armento, G.; Tonini, G.; De Marinis, M.G. Propolis in the prevention of oral mucositis in breast cancer patients receiving adjuvant chemotherapy: A pilot randomised controlled trial. Eur. J. Cancer Care 2017, 26, e12757. [Google Scholar] [CrossRef]
  130. Aghamohammadi, D.; Fakhari, S.; Bilehjani, E.; Hassanzadeh, S. The effects of honey and cinnamon mixture on improving the quality of life in breast cancer. Crescent J. Med. Biol. Sci. 2017, 4, 74–79. [Google Scholar]
Figure 1. The impact of RANKL, estrogen, ROS, and inflammation on the development of osteoporosis (A) and breast cancer (B) (created with BioRender.com, https://www.biorender.com/, accessed on 27 January 2023). Abbreviations: c-Myc—c-myelocytomatosis oncogene product; ERK—extracellular signal-regulated kinase; HSC—hematopoietic stem cell; IL-1—interleukin 1; IL-6—interleukin 6; JNK—c-Jun N-terminal kinase; K-ras—Kirsten rat sarcoma viral oncogene homolog; MSC—mesenchymal stem cell; N-ras—neuroblastoma RAS viral oncogene homolog; NF-κB—nuclear factor kappa-B; OPG—osteoprotegerin; p53—tumor protein p53; PKA—protein kinase A; RANKL—receptor activator of nuclear factor kappa-B ligand; ROS—reactive oxygen species; TNF-α—tumor necrosis factor-alpha; VEGF—vascular endothelial growth factor; ↑—increased; ↓—decreased.
Figure 1. The impact of RANKL, estrogen, ROS, and inflammation on the development of osteoporosis (A) and breast cancer (B) (created with BioRender.com, https://www.biorender.com/, accessed on 27 January 2023). Abbreviations: c-Myc—c-myelocytomatosis oncogene product; ERK—extracellular signal-regulated kinase; HSC—hematopoietic stem cell; IL-1—interleukin 1; IL-6—interleukin 6; JNK—c-Jun N-terminal kinase; K-ras—Kirsten rat sarcoma viral oncogene homolog; MSC—mesenchymal stem cell; N-ras—neuroblastoma RAS viral oncogene homolog; NF-κB—nuclear factor kappa-B; OPG—osteoprotegerin; p53—tumor protein p53; PKA—protein kinase A; RANKL—receptor activator of nuclear factor kappa-B ligand; ROS—reactive oxygen species; TNF-α—tumor necrosis factor-alpha; VEGF—vascular endothelial growth factor; ↑—increased; ↓—decreased.
Antioxidants 12 00567 g001
Table 1. Preclinical and clinical studies on the anti-osteoporotic potential of honey.
Table 1. Preclinical and clinical studies on the anti-osteoporotic potential of honey.
Research ModelsApplied TreatmentObtained ResultsReferences
OVX RatsTualang honey;
0.2 g/kg/day/2 weeks
↑BV/TV
↑Tb.Th
↑Tb.N
↓Tb.Sp
[85]
OVX RatsTualang honey;
0.2, 1.0, and 2.0 g/kg/2 weeks
Tibia: ↑BMD[87]
OVX RatsApis dorsata honey;
2 and 4 g/kg/12 weeks
↑Bone strength[30]
RatsKelulut honey;
200 and 400 mg/kg/day/2 months
↑BV/TV[93]
↑Tb.N
↓Tb.Sp
↑SOD activity
↓MDA activity
RatsKelulut honey↓Oc.S/BS[94]
1 g/kg/8 weeks↓OS/BS
RatsApis melifera honey;
1, 2, and 4 g/kg/2 weeks
↑ALP[95]
Bone cells
Rat/HC-induced bone damage
Greek thyme + honey↓ROS
↓Lipid peroxidation
Synergistic improving effect on parameters of bone turnover
[96]
Postmenopausal women (n = 39)Tualang honey;
20 mg/day/4 months
No difference in BMD and cardiovascular risk between honey and HRT groups[88]
Postmenopausal women (n = 78)Tualang honey;
20 g/day/16 weeks
↓Blood oxidative stress[89]
ALP—alkaline phosphatase; HRT—hormone replacement therapy; BMD—bone mineral density; BV/TV—bone volume per tissue volume; MDA—malondialdehyde; Oc.S/BS—osteoclast surface/bone surface; OS/BS—osteoid surface/bone surface; OVX—ovariectomized; ROS—reactive oxygen species; SOD—superoxide dismutase; Tb.Th—trabecular thickness, Tb.N—trabecular number; Tb.Sp—trabecular separation; ↑—increased; ↓—decreased.
Table 2. Preclinical and clinical studies on honey’s potential against breast cancer.
Table 2. Preclinical and clinical studies on honey’s potential against breast cancer.
Research ModelsApplied TreatmentObtained ResultsReferences
Cells
MCF-7; MDA-MB-231
HeLa
Tualang honey;
1–10%/72 h
↑Cytotoxicity
↑Cell death
↑Apoptosis
↓Δψm
↑Caspase-3/7 and -9
[102]
Cells
MCF-7; MDA-MB-231
Tualang honey;
10%/6, 24, 48, and 72 h
↑Apoptosis
↑Caspase-3/7 and -9
↓TAM-induced adverse effects
[118]
Cells
MDA-MB-231; MDA-MB-435; MCF-7
Manuka honey;
0.3–1.25 %/24–72 h
↓Viability of cancer cells
↑Caspase-dependent apoptosis
↑Bax protein expression
↑Apoptosis
↓IL-6/STAT3 signaling pathway
[122]
Cells
MCF-7
Greek honey extract↓Viability of MCF-7 cells[125]
Cells
MCF-7
Fir honey extract
0.2–125 μg/ml
↑Viability of MCF-7 cells[100]
Cells
MCF7, SKBR3, and MDAMB-231
Chestnut, pine, cedar, multifloral honey;
1, 2.5, 5, 7.5, and 10 µg/mL/
24, 48, and 72 h
↑Cytotoxic effect[126]
RatsTualang honey;
0.2, 1.0, and 2.0 g/kg/day/150 days
↓Tumor development
↓Tumor mean size
↓VEGF protein
[119]
RatsTualang honey, Manuka honey
1.0 g/kg/day/120 days
↓Cancer masses
↓Tumor size, weight, and multiplicity
↓Growth rate
↑Expression of pro-apoptotic proteins (Apaf-1, Caspase-9, IFN-γ, IFNGR1, and p53)
↓Expression of anti-apoptotic proteins (TNF-α, COX-2, and Bcl-xL 1)
[124]
Postmenopausal breast cancer women
(n = 72)
Tualang honey;
20 g/day/12 weeks
↓Alanine aminotransferase levels
↑Creatinine levels
↑Leukocyte counts
↑Platelet counts
[120]
Postmenopausal breast cancer women
(n = 40)
Tualang honey;
20 g/day/6 months
↓BPE[121]
Adult women with breast cancer
(n = 30)
Dorsata honey;
15 mL/3 times daily/15 days
↑IL-3[127]
Adult women with breast cancer
(n = 30)
Dorsata honey;
15 mL/3 times daily/15 days
↑T lymphocytes levels
No differences in IL-6 level
[107]
Adult women with breast cancer
(n = 117)
Honey + cinnamon powder;
30 g + 4 g/3 times daily/1 week
↑Overall quality of life[130]
Δψm—mitochondrial membrane potential; Apaf-1—apoptotic protease activating factor-1; Bcl-xL 1—B-cell lymphoma-extra large; BPE—background parenchymal enhancement; COX-2—cyclooxygenase-2; HeLa—cervical carcinoma; IFN-γ—interferon gamma; IFNGR1—interferon gamma receptor 1; IL-3—interleukin 3; IL-6/STAT3—interleukin-6/tyrosine-phosphorylated; MCF-7, MDA-MB-231, MDA-MB-435—human breast adenocarcinoma cell line; p53—tumor protein; SKBR-3—human breast cancer cell line; STAT3—signal transducer and activator of transcription 3; TAM—tamoxifen; TNF-α—tumor necrosis factor alpha; VEGF—vascular endothelial growth factor; ↑—increased; ↓—decreased.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Martiniakova, M.; Kovacova, V.; Mondockova, V.; Zemanova, N.; Babikova, M.; Biro, R.; Ciernikova, S.; Omelka, R. Honey: A Promising Therapeutic Supplement for the Prevention and Management of Osteoporosis and Breast Cancer. Antioxidants 2023, 12, 567. https://doi.org/10.3390/antiox12030567

AMA Style

Martiniakova M, Kovacova V, Mondockova V, Zemanova N, Babikova M, Biro R, Ciernikova S, Omelka R. Honey: A Promising Therapeutic Supplement for the Prevention and Management of Osteoporosis and Breast Cancer. Antioxidants. 2023; 12(3):567. https://doi.org/10.3390/antiox12030567

Chicago/Turabian Style

Martiniakova, Monika, Veronika Kovacova, Vladimira Mondockova, Nina Zemanova, Martina Babikova, Roman Biro, Sona Ciernikova, and Radoslav Omelka. 2023. "Honey: A Promising Therapeutic Supplement for the Prevention and Management of Osteoporosis and Breast Cancer" Antioxidants 12, no. 3: 567. https://doi.org/10.3390/antiox12030567

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop