Next Article in Journal
Boosting Electrocatalytic Oxidation of Formic Acid on Ir(IV)-Doped PdAg Alloy Nanodendrites with Sub-5 nm Branches
Next Article in Special Issue
Docking-Based Evidence for the Potential of ImmunoDefender: A Novel Formulated Essential Oil Blend Incorporating Synergistic Antiviral Bioactive Compounds as Promising Mpro Inhibitors against SARS-CoV-2
Previous Article in Journal
Outlook on Chronic Venous Disease Treatment: Phytochemical Screening, In Vitro Antioxidant Activity and In Silico Studies for Three Vegetal Extracts
Previous Article in Special Issue
Virtual Screening of a Library of Naturally Occurring Anthraquinones for Potential Anti-Fouling Agents
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Potential Effects of Geraniol on Cancer and Inflammation-Related Diseases: A Review of the Recent Research Findings

by
Rebai Ben Ammar
1,2
1
Department of Biological Sciences, College of Science, King Faisal University, Al-Ahsa 31982, Saudi Arabia
2
Laboratory of Aromatic and Medicinal Plants, Center of Biotechnology of Borj-Cedria, Technopole of Borj-Cedria, P.O. Box 901, Hammam-Lif 2050, Tunisia
Molecules 2023, 28(9), 3669; https://doi.org/10.3390/molecules28093669
Submission received: 5 March 2023 / Revised: 1 April 2023 / Accepted: 17 April 2023 / Published: 23 April 2023
(This article belongs to the Special Issue Bioactivities and In Silico Study of Phytochemicals)

Abstract

:
Geraniol (GNL), a natural monoterpene, is found in many essential oils of fruits, vegetables, and herbs, including lavender, citronella, lemongrass, and other medicinal and aromatic plants. GNL is commonly used by the cosmetic and food industries and has shown a wide spectrum of pharmacological activities including anti-inflammatory, anticancer, antimicrobial, antioxidant, and neuroprotective activities. It represents a potential anti-inflammatory agent and a promising cancer chemopreventive agent, as it has been found to be effective against a broad range of cancers, including colon, prostate, breast, lung, skin, kidney, liver, and pancreatic cancer. Moreover, GNL scavenges free radicals and preserves the activity of antioxidant enzymes. In addition, GNL induces apoptosis and cell cycle arrest, modulates multiple molecular targets, including p53 and STAT3, activates caspases, and modulates inflammation via transcriptional regulation. In the present study, different modes of action are described for GNL’s activity against cancer and inflammatory diseases. This compound protects various antioxidant enzymes, such as catalase, glutathione-S-transferase, and glutathione peroxidase. Experiments using allergic encephalomyelitis, diabetes, asthma, and carcinogenesis models showed that GNL treatment had beneficial effects with low toxicity. GNL has been shown to be effective in animal models and tumor cell lines, but there have not been any clinical studies carried out for it. The aim of the present review is to provide updated data on the potential effects of GNL on cancer and inflammation, and to enhance our understanding of molecular targets, involved pathways, and the possible use of GNL for clinical studies and therapeutic purposes in the treatment of cancer and inflammation-related diseases.

1. Introduction

Recently, molecules from natural products are gaining acceptance as potentially promising complementary and alternative medicines for the treatment of various diseases [1,2,3,4,5,6,7]. Generally, pure compounds could target proteins, DNA, mRNA, and even microRNA. Studies suggest that monoterpenes might offer new chemotherapy strategies for cancer and inflammatory diseases [8,9,10,11], and accumulating evidence in the last decades has indicated that GNL is a pure botanical compound without adverse effects, exerting diverse pharmacological activities by mainly regulating protein expression. The acyclic monoterpene geraniol ((2E)-3,7-dimethylocta-2,6-dien-1-ol) is frequently found in the essential oils of many plant species (Figure 1). Lemongrass, rose, lavender, and other aromatic plants have high concentrations of Geraniol, which was shown to possess various pharmacological properties, including antioxidant [12], anti-inflammatory [13], antimicrobial [14], and antitumor activities [15], through multiple signaling pathway regulation in diverse biological processes [16,17,18]. Moreover, many studies have found that GNL inhibits cancer cell growth in vitro via the signal transduction pathway, leading to apoptosis [19,20,21,22]. Geraniol exerts in vitro and in vivo antitumor activity against murine leukemia, hepatoma, and melanoma cells [23,24]. Furthermore, the biochemical, molecular, and histological studies conducted so far indicate that GNL has antioxidant and anti-inflammatory properties. Thus, it is believed that this compound, having a strong preventive potential, can protect against oxidative and inflammatory changes [25]. Further investigation demonstrated that GNL promotes the metabolism of inflammatory cells, increases GSH content, and stimulates antioxidant enzyme activities [1]. Ji et al. [26] investigated GNL’s immunosuppressive properties by using in vitro lymphocyte proliferation assays and an in vivo rat cardiac allograft transplant model, revealing that GNL can prevent acute allograft rejection. Although many publications have been published about its anticancer activity, the mechanism of GNL molecular activity has still not been fully explained. This review aims to extend previous review findings [1,18,19,25,27] by providing an updated overview of the literature on the potential effects of GNL on cancer and inflammation-related diseases, specifically focusing on experimental study findings which enhance our understanding of molecular targets and pathways involved in antioxidant/anti-inflammatory and anticancer effects of GNL, and the possible use of this natural monoterpene for the treatment of cancer and various inflammation related-diseases. Further, the present study aims to provide a mechanistic insight into GNL’s potential as an anti-inflammatory and anticancer agent, paving the way for future experimental and clinical studies, as well as for therapeutic purposes.

2. Research Methodology

To gather updated data on the potential effects of GNL on cancer and inflammation-related diseases, we used the most well-known scientific search engines: PubMed, Web of Science, ScienceDirect, Wiley Online, Google Scholar, SpringerLink, and Scopus. Data were mainly collected from several sources including journal articles, books, book chapters, and scientific reports. The search for this information was carried out between November 2022 to March 2023. The scientific literature published in the years 2020 to 2023 was searched. However, several papers published before 2020 were also included for insight in the introduction and explanation. We used Geraniol either alone or combined with ‘anti-inflammatory’, ‘antitumor’, ‘anticancer’, ‘antiproliferative’, ‘cytotoxicity’, and ‘antioxidant’ as the key words for the literature searches. The internet search generated 1147 articles in total. Duplicate articles and articles with similar results were excluded. Publications in other languages than English and those with limited raw data were also excluded. Only 104 articles were finally included in this study after reading the titles, abstracts, and whole papers.

3. Anti-Inflammatory and Antioxidant Properties of GNL

The immune system reacts non-specifically to injury or infection when inflammation occurs [25,28,29]. Research has revealed that chronic inflammation can cause diseases such as cancer, cardiovascular disease, and neurological disorders [30,31,32,33,34]. Since GNL has anti-inflammatory and antioxidant effects, scientists are studying its molecular mechanisms and potential applications in treating inflammation. El Azab and Abdulmalek [35] evaluated the ameliorative effect of GNL on age-related multiple inflammation and neuronal impairments in rats fed with high-fat diet (HFD). Their results revealed a drop in proinflammatory cytokines (TNF-α and IL-6) and leptin while boosting adiponectin in GNL-supplemented rats. The liver, kidney, and lipid profiles were improved in GNL-HFD-treated groups. In addition, GNL suppressed acetylcholinesterase (AChE) activity and alleviated oxidative stress by boosting neuronal reduced glutathione (GSH), catalase (CAT), glutathione-S-transferase (GST), and superoxide dismutase (SOD) activities. It lowered malondialdehyde concentration (TBARS), nitric oxide (NO), and xanthine oxidase (XO), and restored the structural damage to the brain tissue caused by HFD. Compared with model rats, GNL boosted learning and memory function and ameliorated the inflammation status in the brain by lowering the protein levels of IL-1β, iNOS, NF-κBp65, and COX-2 (Figure 2).
Furthermore, the supplementation of GNL reversed the oxidative and inflammation changes associated with aging. According to El Azab and Abdulmalek [35], dietary GNL supplementation was effective in modifying age-related neuroinflammation and oxidative stress in rats, suggesting the use of GNL as a noninvasive natural compound for controlling age- and diet-associated neuronal impairments and toxicity. In the same framework, AlAsmari et al. [36] investigated the potential effect of GNL on the modulation of doxorubicin-induced kidney damage in rats. A single doxorubicin injection triggered kidney impairment, as evidenced by the altered blood urea nitrogen, serum creatinine, and albumin parameters; it also caused kidney histological changes. Furthermore, doxorubicin increased lipid peroxidation while lowering catalase activity, reduced glutathione, and the expression of glutathione peroxidase and superoxide dismutase. Moreover, prophylactic treatment with GNL preserved most kidney histological characteristics in a dose-dependent manner, showing that GNL could protect against doxorubicin-mediated kidney dysfunction [36].

4. Anti-Free Radical Properties of GNL

GNL decreases the brain damage induced by ischemia/reperfusion in mice [29]. In RAW264.7 rat macrophages, GNL suppressed reactive oxygen species (ROS) levels induced by lipopolysaccharides [37]. COX-2 protein levels and mRNA levels were significantly attenuated, but the cytosolic degradation of IBα and upregulation of NF-κB p65 in the nucleus were reversed. LPS/D-GalN-induced FHF was inhibited by GNL through the inhibition of inflammation and an increase in PPARγ expression [38]. Another experiment showed the aforementioned inhibitory effects were mediated by the inhibition of iNOS and COX2 enzyme induction. The induction of iNOS and COX-2 has also been reported in cyclophosphamide-induced cystitis in rats [39]. In addition, iNOS and COX-2 inhibitors have been shown to reduce hyperactivity. Mohamed et al. inspected the GNL’s reno-protective effects against renal I/R damage with further analysis of embedded mechanisms of action through scrutinizing the Nrf-2/HO-1/NQO-1 and TLR2,4/MYD88/NFκB signaling pathways [40]. Their results showed that Renal I/R rats experienced severely compromised renal functions, histological alteration, oxidative stress status, escalated Nrf-2/HO-1/NQO-1, and amplified TLR2,4/MYD88/NFκB. GNL administration ameliorated renal function, alleviated histological changes, and enhanced Nrf-2/HO-1/NQO-1 with a subsequent intensification of antioxidant enzyme activities. GNL declined TLR2,4/MYD88/NFκB with subsequent TNF-α, IFN-γ, MCP-1 drop, Bax, caspase-3, and caspase-9 reduction IL-10 and Bcl-2 augmentation. GNL, therefore, might protect against renal I/R via the inhibition of the TLR2,4/MYD88/NFκB pathway, mediating anti-inflammation and activation of the Nrf2 pathway, this intervening in antioxidative activities [40]. In general, GNL has been shown to enhance the effect of antioxidant enzymes and inhibit inflammatory proteins.

5. Effects of GNL on Autoimmune Diseases

Inflammation triggers the immune system to react non-specifically against injury or infection. Chronic or continuous inflammatory diseases can result from uncontrolled inflammation [29]. Studies have shown that GNL can help with diabetes, arthritis, asthma, and other autoimmune diseases [41,42,43]. In a streptozotocin diabetic mouse model, GNL abrogated hyperglycemic and hypoinsulinemic responses [44]. Ischemia/reperfusion-induced brain damage was shown to be decreased by GNL in mice [45]; in this study, GNL suppressed TNF-α, iNOS, and COX-2 levels. Furthermore, GNL, Bax, and caspase-3 and -9 reduced antiapoptotic activity in liver tissue. The Nrf2/HO-1 antioxidant pathway is activated by GNL, making it a promising hepatoprotective agent. In streptozotocin or streptozotocin–nicotinamide diabetic rat models, GNL reversed both elevated glucose and low insulin levels [42,46]. GNL may have therapeutic effects in the treatment of diabetes by modulating serum glucose and insulin levels. Streptozotocin diabetic rats showed improvement in neuropathy and nephropathy with administration of GNL, suggesting that it could help to prevent diabetes-related diseases [47,48]. Additionally, this monoterpene reduced collagen-induced arthritis in mice [49]. In another study, GNL reduced serum nitric oxide, urea, and creatinine levels, as well as prevented kidney dysfunction, which is common in nephrotoxic animal models [50]. GNL has also been shown to decrease alkaline phosphatase expression [51]. A nonreceptor tyrosine kinase called Tyk2 was inhibited by GNL in mice induced by OVA [40]. Through modulating Tyk2-STAT1/3, GNL alleviates airway inflammation. LPS was also shown to stimulate STAT1/3 signaling while downregulating Tyk-inhibiting SOCS3 expression [52].

6. Glutathione Modulation by GNL

In a study by Stobiecka et al. [52], GNL was found to be a powerful scavenger of free radicals [52]. These findings are in line with [53], in which it was shown that GNL inhibits iron-dependent microsomal lipid peroxidation. These results suggest that GNL might have a role in oxidative stress prevention and treatment. Various sources of free radicals or superoxide radicals can be neutralized by antioxidant enzymes. In CCl4-induced oxidative stress rats, Mostafa, et al. [54] showed that GNL reversed the expression of catalase, glutathione peroxidase, and glutathione-S-transferase, and reduced glutathione in the liver tissues [54]. GNL also reverses isoproterenol’s suppression of catalase, glutathione peroxidase and glutathione-S-transferase activity, and mRNA expression [55]. Thus, GNL might be useful for reducing the adverse effects of inflammatory disorders caused by free radicals. Glutathione is a tripeptide used in drug detoxification that can prevent ROS damage [20,56]. Pronin et al. [57] reported that GNL improved experimental anti-encephalitis in vitro [57]. In addition, Kandeil et al. [58] noted that GNL reversed the reduced levels of glutathione, glutathione peroxidase, and catalase levels in rats treated with cisplatin [58]. GNL’s effects on glutathione levels in the body need to be investigated more closely, since such findings might explain its role in inflammation suppression or cancer prevention.

7. Transcriptional Effects of GNL on Inflammation

A wide range of stimuli, including stress, bacteria, viruses, cytokines, and free radicals, can trigger NF-ĸB. This transcription factor regulates the expression of a variety of genes, including enzymes, cytokines, and cell-cycle regulators [58,59,60]. In LPS-induced acute lung injury, Jiang et al. [61] reported that GNL inhibited the production of pro-inflammatory cytokines in BALB, including TNF-α, IL-1β, and IL-6, as well as the MAPKs and NF-kB signaling pathways [61]. Another study showed that monoterpene fractions suppress TNF-α-induced NF-kB activation [13]. A previous study suggested that GNL inhibited NF-kB translocation into the nucleus in Ox-LDL-stimulated inflammation in HUVECs [62]. In addition, Lee et al. [63] found that an extract of lemongrass abrogates LPS-induced NF-kB signaling in RAW264.7 cells by targeting IKKb [63]. Several inflammatory disease models have been studied and the benefits of modulating NF-kB with GNL have been demonstrated. Treatment with GNL had previously been shown to prevent or ameliorate Japanese encephalomyelitis, possibly by inhibiting NF-kB [64]. According to a recent study [27,43], GNL may protect against rheumatoid arthritis through inhibition of NFkB-p65, p38, and ERK1/2 phosphorylation (Figure 3). NF-kB suppression by GNL probably plays a significant role in its anti-inflammatory properties. Angiogenesis, metastasis, differentiation, proliferation, and apoptosis are all controlled by STAT3. Many carcinomas are constitutively activated by STAT3, and it interferes with tumor growth at different levels [65,66,67]. A reduction in survivin levels caused by GNL-induced apoptosis was shown by Kuzu et al. [68]. MG132 inhibits proteasomal degradation in GNL-treated cells, restoring survivin levels [69]. In other words, survivin attenuates GNL-induced apoptosis. GNL reduces survivin protein levels by downregulating phosphorylated STAT3. GNL also inhibits survivin by blocking serine/threonine kinase activity because active phosphorylated STAT3 increases survivin stability. GNL inhibits the inhibitors of apoptosis, such as Bclxl, Mcl and Bcl-2, which belong to the anti-apoptotic Bcl-2 family [70]. GNL also reduces IL-8 production by LPS-stimulated human pulmonary epithelial cells [71]. As well as its antioxidant properties, GNL has PARP-1-inhibiting activity [72].

8. Anticancer and Chemo Preventive Effects of GNL

Chemoprevention has emerged as a new strategy for fighting cancer, preventing and reducing cancer risk via the ingestion or administration of natural or synthetic chemicals. In the search for new cancer chemopreventive agents, many plant constituents have been evaluated for their chemopreventive activities against cancers over the past few years [73]. The possible mechanism of GNL in relation to antioxidant status is the inhibition of free radical formation and reduced cancer incidence. It was found that GNL treatment is effective during the post-initiation phase of carcinogenesis. An increase in the activity of antioxidants and a decrease in the level of marker enzymes suggest that GNL reduces the adverse effects of cancer. Furthermore, a reduction in the levels of glycoprotein components during treatment with GNL indicates that acyclic monoterpene alcohol has the ability to suppress malignancy by modulating cell transformation through controlling cell proliferation. GNL plays a very important cytoprotective role against B(a)P-induced lung carcinogenesis [74].
In many countries, cancer is one of the most common diseases, with an increasing incidence rate every year [75]. There is an urgent need for a cure to this disease, as treatment is expensive, and complications frequently lead to death. GNL has been shown to inhibit the in vivo growth of cancer in mice with benzo(a)pyrene-induced lung cancer [73]. The antioxidant properties of GNL have been linked to the prevention of chemical-induced cancer. Scientists are researching the molecular mechanism(s) involved and are evaluating GNL’s significance in treating cancer on the basis of its potential. A number of in vitro and in vivo studies have shown that GNL inhibits tumorigenesis and prevents carcinogenesis. Elsayad and Adeshina evaluated the antitumor effects of GNL on oral cancer [33]. They have shown that GNL treatment significantly suppressed oral squamous cell carcinoma (OSCC) cell proliferation and migration in vitro and tumor growth in vivo in a time- and dose-dependent manner. In addition, they have shown that GNL treatment significantly caused OSCC apoptosis and blocked Phosphatidylinositol-3-kinase/protein kinase B (PI3K/AKT) signaling activation concurrently. They also found that GNL administration did not affect the body weight on tumor-bearing mice, confirming the GNL safety and showing that this compound may serve as a promising anticancer drug for oral cancer treatment [33].

9. Effect of GNL on Cell Proliferation

Hyper-proliferative cancerous cells are well-established targets for cancer treatment [75]. GNL helps to identify transcription factors controlling the gene expression of HaCaT keratinocytes [76,77]. Several kinds of carcinoma, including glioma and glioblastoma, breast carcinoma, osteosarcoma, and colorectal carcinoma, are inhibited by this compound [27,33,50,78,79,80]. 4NQO-induced oral cancer was inhibited by GNL in rats. GNL inhibits phase I enzymes and blocks the bioactivation of 4NQO to 4HAQO, which is cancer-causing, leading to a significant reduction in tumor volume and number [81]. Thymidine kinase and thymidylate synthase were effectively reduced by GNL. In Swiss nu/nu mice with implanted human TC-118 cancer cells, simultaneous administration of 5-fluorouracyl and GNL resulted in 53% tumor volume reduction, versus 26% for GNL alone and no difference for 5-fluorouracyl alone [15]. When chronically exposed to Fe-NTA, rats treated with rat GNL showed a significant decrease in p53 protein levels and a significant increase in caspase-3, -8, and -9, some of which are classic markers of inflammation, proliferation, and apoptosis [82]. In hamsters treated with DMBA, GNL completely prevented the formation of tumors in the cheek mucosa, affected the expression of p53 and Bcl-2, and increased the levels of Bax protein and caspase-3 and -9. GNL prevents cell proliferation markers, inflammation, apoptosis, and angiogenesis at tumor sites after oral administration [67]. Duan et al. [83] studied the potential effect of a multi-bioresponsive self-assembled nano drug delivery system based on hyaluronic acid and GNL against liver cancer [83]. A multi-bioresponsive self-assembled nano-drug delivery system (HSSG) was constructed by conjugating GNL to hyaluronic acid (HA) via a disulfide bond. According to Duan et al. [68], results of fluorescence microscopy and flow cytometry showed that HSSG NPs were uptaken by hepatocellular carcinoma cell lines HepG2 and Huh7 via CD44 receptor-mediated internalization. Studies on H22 tumor-bearing mice revealed that HSSG NPs could effectively accumulate at the tumor site for a long period. In vitro and in vivo studies showed that HSSG NPs significantly promoted the death of cancer cells while reducing the toxicity. Therefore, the HSSG NPs have great potential in the treatment of tumors [83]. Based on these results, GNL may be helpful in treating different types of malignancy, while having limited effects on normal cells.

10. Cancer Cells Respond to GNL in A Pro-Apoptotic Way

The ability of chemotherapeutic drugs to induce apoptosis determines their effectiveness in cancer treatment. As a cancer preventative or therapeutic agent, GNL has been shown to have beneficial effects on various types of cancer in humans but does not affect normal physiology by regulating the cell cycle and/or inducing apoptosis. According to Carnesecchi et al. [84], GNL induced apoptosis and growth inhibition in the human HCT116 colon cancer cell line. GNL activated caspase-3 cleavage and released cytochrome c from mitochondria. Bcl-2 family proteins such as PUMA play a key role in this [19,85]. In HCT116 cells, GNL also phosphorylated ATM and H2AX, and inhibited ATM with a chemical inhibitor that abrogated the downstream apoptotic cascades [77]. GNL treatment decreased Bcl-xL protein levels in ovarian cancer cells, while Bax, p53, and Bad protein levels were upregulated [70,86]. GNL also induced more apoptosis than either agent used alone in human colon cancer SW480 cells [87]. By regulating pro-apoptotic and anti-apoptotic proteins in the intrinsic apoptosis pathway, GNL induces apoptosis in ovarian cancer cells. Following GNL treatment, Bclxl protein levels were decreased in ovarian cancer cells, while p53, Bad, and Bax protein levels were upregulated (Figure 4). In addition, Akt was inactivated, the mitochondrial phase of apoptosis was activated, cytochrome c was released, caspase 3 was activated, and cells death eventuated [33]. Duncan et al. [88] observed that GNL inhibits cyclin-dependent kinase 2, which regulates cell proliferation, anti-apoptotic gene products, and metastatic transcription factors [88]. According to Shoff et al. [12], GNL inhibits liver HMG-CoA, which participates in cholesterol biosynthesis in mammals, as well as the cell cycle. As a result, its inhibitors can effectively demonstrate an anticancer activity by stopping the cell cycle between the G1 and S phases [12]. Zhuang et al. [89] evaluated the GNL protective effect on Helicobacter pylori-induced human gastric cancer signaling by increasing peroxiredoxin-1 expression in human gastric epithelial cells (GES-1) [89]. Indeed, GNL inhibits H. pylori-induced gastric carcinogen signaling by preventing ROS formation, cytotoxicity, and apoptosis in GES-1 cells. In addition, GNL prevents H. pylori-induced antioxidant depletion induced by nuclear fragmentation, damage of reactive DNA, and malondialdehyde. Furthermore, GNL potentially reduced the expression of phosphorylated mitogen and activated protein kinases (MAPKs) proteins such as p38 MAPK, tumor necrosis factor-alpha (TNF-α), c-Jun N-terminal kinase (c-JNK), extracellular signal-regulated kinase-1 (ERK1), interleukin-6 (IL-6), and cyclooxygenase-2 (COX-2) in GES-1 infected with H. pylori. Thus, GNL protects against H. pylori-concomitant infection, and its resistance may be a possible method of preventing gastric cancer caused by H. pylori [89].
The antitumoral activities of GNL against different types of cancer and using different modes of study are summarized in Table 1.

11. Effect of GNL on Metastasis and Angiogenesis Inhibition

In numerous studies, GNL has been reported to inhibit cancer metastasis and angiogenesis. Researchers have found that monoterpene modulates apoptosis, angiogenesis, inflammation, and metastasis signaling pathways. GNL seems to inhibit cancer cell growth and angiogenesis and induce cancer cell apoptosis, while preserving normal cell viability, and in some cases even protecting it. Part of the purpose of this review was to summarize information about GNL and provide insight into its potential chemo-preventive effects. VEGF gene expression at both the mRNA and protein levels is significantly reduced in endothelial-like eEND2 cells by GNL [103]. GNL inhibited tumor growth in 7,12-dimethylbenz[a]anthracene (DMBA)-induced hamster mice [83]. El-Ella [104] found that in A549 lung cancer cells treated with GNL, downregulation of HIF-1alpha, a VEGF regulator, occurred [104]. With onset of hypoxia, GNL downregulates NF-ĸB and VEGF expression levels, resulting in the downregulation of inflammatory and angiogenic markers. GNL also decreases autophagy through downregulation of BNIP3 and beclin-1 expression, which increases apoptotic cell death through HIF-1α signaling.

12. An Overview of GNL Bioavailability

Using NCM460 cells derived from primary human colon mucosa cells, tests were performed to simulate GNL penetration through the in vitro intestinal barrier. Additionally, GNL can potentially be transported from the intestinal lumen into the bloodstream without any degradation in the digestive tract, which indicates high penetration through the monolayers of cells. Plant fibers absorbed geraniol at 16% bioavailability, so it can reach the colon through the intestines [25,102]. Even at concentrations as high as 300 g/mL, geraniol is excreted from the bloodstream within about 12 min after intravenous administration. Based on its binding to blood proteins and cellular components, and its penetration into lipid compartments, researchers have suggested that in this case, a high concentration of GNL could damage mitochondrial depletion, leading to apoptosis. In studies performed using Sprague Dawley rats, the absolute availability of GNL was 92% with administration of GNL emulsified in glycerol. The maximum blood concentration was found after 30 min and was about 270 µg/mL. It is worth emphasizing that this was similar to the value obtained after intravenous administration of the same dose of GNL [16]. The concentration of GNL in the cerebrospinal fluid of the rats dropped rapidly over time, similarly to the blood concentration [16].

13. Conclusions and Future Prospects

Different modes of action have been shown to occur with GNL application against cancer and inflammation. Various antioxidant enzymes, such as catalase, glutathione peroxidase, and glutathione-S-transferase, are protected by this compound, which scavenges free radicals and superoxide radicals. Experiments using allergic encephalomyelitis, diabetes, asthma, and carcinogenesis models have shown that GNL treatment has beneficial effects. There are several different anticancer mechanisms, including the inhibition of proliferation, the induction of cell cycle arrest, the induction of apoptosis, synergy with conventional medicines, and ROS generation. Additionally, GNL can attenuate the toxicity associated with conventional medicines without compromising their effectiveness. GNL has been shown to target a variety of molecules in cancer cell lines; however, animal models are needed to obtain more conclusive evidence of the molecular basis of its action. In addition, novel GNL analogs have been synthesized and found to have better anticancer, anti-inflammatory and antioxidant properties. It has been shown to be effective in animal models and tumor cell lines, but there have not yet been any clinical studies carried out to further assess these effects. More studies are needed before GNL can be developed into a drug for cancer treatment and the treatment of inflammatory diseases. For GNL to be commercialized as a cancer preventative and therapeutic agent, substantial evidence from epidemiological research and clinical trials will be required. Further, the present study provides a mechanistic insight into GNL’s potential as an anti-inflammatory and anticancer agent, paving the way for future experimental studies. Overall, these findings show that GNL modulates molecular targets and pathways to trigger apoptosis in cancer cells. GNL is rapidly moving into clinical trials, so these targets could be exploited for therapeutic purposes in future.

Funding

This work was supported by the Deanship of Scientific Research, Vice Presidency for Graduate Studies and Scientific Research, King Faisal University, Saudi Arabia (Grant 3284 /1128/AN000122).

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors thank the Deanship of Scientific Research, King Faisal University and College of Science.

Conflicts of Interest

The author declares no conflict of interest.

Sample Availability

Not applicable.

References

  1. Lei, Y.; Fu, P.; Jun, X.; Cheng, P. Pharmacological Properties of Geraniol—A Review. Planta Med. 2019, 85, 48–55. [Google Scholar] [CrossRef] [PubMed]
  2. Hegde, M.M.; Lakshman, K. Role of Polyphenols and Flavonoids as Anti-Cancer Drug Candidates: A Review. Pharmacognosy Res. 2023, 15, 206–216. [Google Scholar] [CrossRef]
  3. Chimento, A.; De Luca, A.; D’Amico, M.; De Amicis, F.; Pezzi, V. The Involvement of Natural Polyphenols in Molecular Mechanisms Inducing Apoptosis in Tumor Cells: A Promising Adjuvant in Cancer Therapy. Int. J. Mol. Sci. 2023, 24, 1680. [Google Scholar] [CrossRef] [PubMed]
  4. Jia, W.; Zhou, L.; Li, L.; Zhou, P.; Shen, Z. Nano-Based Drug Delivery of Polyphenolic Compounds for Cancer Treatment: Progress, Opportunities, and Challenges. Pharmaceuticals 2023, 16, 101. [Google Scholar] [CrossRef] [PubMed]
  5. Aloo, S.O.; Ofosu, F.K.; Kim, N.H.; Kilonzi, S.M.; Oh, D.H. Insights on Dietary Polyphenols as Agents against Metabolic Disorders: Obesity as a Target Disease. Antioxidants 2023, 12, 416. [Google Scholar] [CrossRef]
  6. Gupta, N.; Singh, S.; Chauhan, D.; Srivastava, R.; Singh, V.K. Exploring the Anticancer Potentials of Polyphenols: A Comprehensive Review of Patents in the Last Five Years. Recent Pat. Anticancer Drug Discov. 2023, 18, 3–10. [Google Scholar]
  7. Sahiner, M.; Yilmaz, A.S.; Gungor, B.; Sahiner, N. A Review on Phyto-Therapeutic Approaches in Alzheimer’s Disease. J. Funct. Biomater. 2023, 14, 50. [Google Scholar] [CrossRef]
  8. Yang, J.; Zhong, C.; Yu, J. Natural Monoterpenes as Potential Therapeutic Agents against Atherosclerosis. Int. J. Mol. Sci. 2023, 24, 2429. [Google Scholar] [CrossRef]
  9. Sajid, A.; Manzoor, Q.; Sajid, A.; Nazir, A.; Mumtaz, M.A.; Fatima, N.; Alshawwa, S.Z.; Iqbal, M.; Younas, U. Downregulation of NF-κB activation pathways using essential oil derived from Citrus pseudolimon: Anticancer and anti-inflammatory potential. Biocatal. Agric. Biotechnol. 2023, 47, 102599. [Google Scholar] [CrossRef]
  10. Abdul Ghani, M.A.; Ugusman, A.; Latip, J.; Zainalabidin, S. Role of Terpenophenolics in Modulating Inflammation and Apoptosis in Cardiovascular Diseases: A Review. Int. J. Mol. Sci. 2023, 24, 5339. [Google Scholar] [CrossRef]
  11. Abdul-Hameed, Z.H.; Bawakid, N.O.; Alorfi, H.S.; Sobahi, T.R.; Alburae, N.A.; Abdel-Lateff, A.; Elbehairi, S.E.I.; Alfaifi, M.Y.; Alhakamy, N.A.; Alarif, W.M. Monoterpene Indole Alkaloids from the Aerial Parts of Rhazya stricta Induce Cytotoxicity and Apoptosis in Human Adenocarcinoma Cells. Molecules 2022, 27, 1422. [Google Scholar] [CrossRef] [PubMed]
  12. Shoff, S.M.; Grummer, M.; Yatvin, M.B.; Elson, C.E. Concentration-dependent increase of murine P388 and B16 population doubling time by the acyclic monoterpene geraniol. Cancer Res. 1991, 51, 37–42. [Google Scholar] [PubMed]
  13. de Cássia da Silveira e Sá, R.; Andrade, L.N.; de Sousa, D.P. A review on antiinflammatory activity of monoterpenes. Molecules 2013, 18, 1227–1254. [Google Scholar] [CrossRef] [PubMed]
  14. Solorzano-Santos, F.; Miranda-Novales, M.G. Essential oils from aromatic herbs as antimicrobial agents. Curr. Opin. Biotechnol. 2012, 23, 136–141. [Google Scholar] [CrossRef]
  15. Carnesecchi, S.; Bras-Goncalves, R.; Bradaia, A.; Zeisel, M.; Gosse, F.; Poupon, M.F.; Raul, F. Geraniol, a component of plant essential oils, modulates DNA synthesis and potentiates 5-fluorouracil efficacy on human colon tumor xenografts. Cancer Lett. 2004, 215, 53–59. [Google Scholar] [CrossRef]
  16. Pavan, B.; Dalpiaz, A.; Marani, L.; Beggiato, S.; Ferraro, L.; Canistro, D.; Paolini, M.; Vivarelli, F.; Valerii, M.C.; Comparone, A. Geraniol pharmacokinetics, bioavailability and its multiple effects on the liver antioxidant and xenobiotic-metabolizing enzymes. Front. Pharmacol. 2018, 9, 18. [Google Scholar] [CrossRef]
  17. Turina, A.d.V.; Nolan, M.; Zygadlo, J.; Perillo, M. Natural terpenes: Self-assembly and membrane partitioning. Biophys. Chem. 2006, 122, 101–113. [Google Scholar] [CrossRef]
  18. Chen, W.; Viljoen, A.M. Geraniol—A review of a commercially important fragrance material. S. Afr. J. Bot. 2010, 76, 643–651. [Google Scholar] [CrossRef]
  19. Cho, M.; So, I.; Chun, J.N.; Jeon, J.-H. The antitumor effects of geraniol: Modulation of cancer hallmark pathways. Int. J. Oncol. 2016, 48, 1772–1782. [Google Scholar] [CrossRef]
  20. Valdivieso-Ugarte, M.; Gomez-Llorente, C.; Plaza-Díaz, J.; Gil, Á. Antimicrobial, antioxidant, and immunomodulatory properties of essential oils: A systematic review. Nutrients 2019, 11, 2786. [Google Scholar] [CrossRef]
  21. Mamur, S. Geraniol, a natural monoterpene, identifications of cytotoxic and genotoxic effects in vitro. J. Essent. Oil Res. 2022, 34, 54–64. [Google Scholar] [CrossRef]
  22. Polo, M.; Crespo, R.; De Bravo, M. Geraniol and simvastatin show a synergistic effect on a human hepatocarcinoma cell line. Cell Biochem. Funct. 2011, 29, 452–458. [Google Scholar] [CrossRef] [PubMed]
  23. Burke, Y.D.; Stark, M.J.; Roach, S.L.; Sen, S.E.; Crowell, P.L. Inhibition of pancreatic cancer growth by the dietary isoprenoids farnesol and geraniol. Lipids 1997, 32, 151–156. [Google Scholar] [CrossRef] [PubMed]
  24. Yu, S.G.; Hildebrandt, L.A.; Elson, C.E. Geraniol, an inhibitor of mevalonate biosynthesis, suppresses the growth of hepatomas and melanomas transplanted to rats and mice. J. Nutr. 1995, 125, 2763–2767. [Google Scholar]
  25. Mączka, W.; Wińska, K.; Grabarczyk, M. One Hundred Faces of Geraniol. Molecules 2020, 25, 3303. [Google Scholar] [CrossRef]
  26. Ji, P.; Si, M.-S.; Podnos, Y.; Imagawa, D.K. Monoterpene geraniol prevents acute allograft rejection. Transplant. Proc. 2002, 34, 1418–1419. [Google Scholar] [CrossRef]
  27. e Silva, G.d.S.; de Jesus Marques, J.N.; Linhares, E.P.M.; Bonora, C.M.; Costa, É.T.; Saraiva, M.F. Review of anticancer activity of monoterpenoids: Geraniol, nerol, geranial and neral. Chem.-Biol. Interact. 2022, 362, 109994. [Google Scholar] [CrossRef]
  28. Crespo, R.; Rodenak-Kladniew, B.E.; Castro, M.A.; Soberón, M.V.; Lavarías, S.M.L. Induction of oxidative stress as a possible mechanism by which geraniol affects the proliferation of human A549 and HepG2 tumor cells. Chem.-Biol. Interact. 2020, 320, 109029. [Google Scholar] [CrossRef]
  29. Rajendran, P.; Chen, Y.F.; Chen, Y.F.; Chung, L.C.; Tamilselvi, S.; Shen, C.Y.; Day, C.H.; Chen, R.J.; Viswanadha, V.P.; Kuo, W.W. The multifaceted link between inflammation and human diseases. J. Cell. Physiol. 2018, 233, 6458–6471. [Google Scholar] [CrossRef]
  30. Coussens, L.M.; Werb, Z. Inflammation and cancer. Nature 2002, 420, 860–867. [Google Scholar] [CrossRef]
  31. Singh, N.; Baby, D.; Rajguru, J.P.; Patil, P.B.; Thakkannavar, S.S.; Pujari, V.B. Inflammation and cancer. Ann. Afr. Med. 2019, 18, 121. [Google Scholar] [CrossRef] [PubMed]
  32. Farokhcheh, M.; Hejazian, L.; Akbarnejad, Z.; Pourabdolhossein, F.; Hosseini, S.M.; Mehraei, T.M.; Soltanpour, N. Geraniol improved memory impairment and neurotoxicity induced by zinc oxide nanoparticles in male wistar rats through its antioxidant effect. Life Sci. 2021, 282, 119823. [Google Scholar] [CrossRef] [PubMed]
  33. Elsayad, J.; Adeshina, N. Antitumor effects of geraniol on oral cancer. STEMedicine 2023, 4, e155. [Google Scholar] [CrossRef]
  34. Younis, N.S.; Elsewedy, H.S.; Soliman, W.E.; Shehata, T.M.; Mohamed, M.E. Geraniol isolated from lemon grass to mitigate doxorubicin-induced cardiotoxicity through Nrf2 and NF-κB signaling. Chem.-Biol. Interact. 2021, 347, 109599. [Google Scholar] [CrossRef]
  35. El Azab, E.F.; Abdulmalek, S. Amelioration of Age-Related Multiple Neuronal Impairments and Inflammation in High-Fat Diet-Fed Rats: The Prospective Multitargets of Geraniol. Oxid. Med. Cell. Longev. 2022, 2022, 4812993. [Google Scholar] [CrossRef] [PubMed]
  36. AlAsmari, A.F.; Ali, N.; Alharbi, M.; Alqahtani, F.; Alasmari, F.; Almoqbel, D.; AlSwayyed, M.; Alshammari, A.; Alanazi, M.M.; Alhoshani, A. Geraniol Ameliorates Doxorubicin-Mediated Kidney Injury through Alteration of Antioxidant Status, Inflammation, and Apoptosis: Potential Roles of NF-κB and Nrf2/Ho-1. Nutrients 2022, 14, 1620. [Google Scholar] [CrossRef]
  37. Su, Y.-W.; Chao, S.-H.; Lee, M.-H.; Ou, T.-Y.; Tsai, Y.-C. Inhibitory effects of citronellol and geraniol on nitric oxide and prostaglandin E2 production in macrophages. Planta Med. 2010, 76, 1666–1671. [Google Scholar] [CrossRef] [PubMed]
  38. Li, Y.; Wang, N.; Jiang, Y. Geraniol protects against lipopolysaccharide and D-galactosamine-induced fulminant hepatic failure by activating PPARγ. Microb. Pathog. 2019, 128, 7–12. [Google Scholar] [CrossRef]
  39. Mohamed, M.E.; Elmorsy, M.A.; Younis, N.S. Renal Ischemia/Reperfusion Mitigation via Geraniol: The Role of Nrf-2/HO-1/NQO-1 and TLR2,4/MYD88/NFκB Pathway. Antioxidants 2022, 11, 1568. [Google Scholar] [CrossRef]
  40. Mohammed, M.J.; Tadros, M.G.; Michel, H.E. Geraniol protects against cyclophosphamide-induced hepatotoxicity in rats: Possible role of MAPK and PPAR-γ signaling pathways. Food Chem. Toxicol. 2020, 139, 111251. [Google Scholar] [CrossRef]
  41. Xue, Z.; Zhang, X.-G.; Wu, J.; Xu, W.-C.; Li, L.-Q.; Liu, F.; Yu, J.-E. Effect of treatment with geraniol on ovalbumin-induced allergic asthma in mice. Ann. Allergy Asthma Immunol. 2016, 116, 506–513. [Google Scholar] [CrossRef] [PubMed]
  42. El-Bassossy, H.M.; Elberry, A.A.; Ghareib, S.A. Geraniol improves the impaired vascular reactivity in diabetes and metabolic syndrome through calcium channel blocking effect. J. Diabetes Complicat. 2016, 30, 1008–1016. [Google Scholar] [CrossRef] [PubMed]
  43. Wu, Y.; Wang, Z.; Fu, X.; Lin, Z.; Yu, K. Geraniol-mediated osteoarthritis improvement by down-regulating PI3K/Akt/NF-κB and MAPK signals: In vivo and in vitro studies. Int. Immunopharmacol. 2020, 86, 106713. [Google Scholar] [CrossRef] [PubMed]
  44. Babukumar, S.; Vinothkumar, V.; Sankaranarayanan, C.; Srinivasan, S. Geraniol, a natural monoterpene, ameliorates hyperglycemia by attenuating the key enzymes of carbohydrate metabolism in streptozotocin-induced diabetic rats. Pharm. Biol. 2017, 55, 1442–1449. [Google Scholar] [CrossRef] [PubMed]
  45. El-Emam, S.Z.; Soubh, A.A.; Al-Mokaddem, A.K.; Abo El-Ella, D.M. Geraniol activates Nrf-2/HO-1 signaling pathway mediating protection against oxidative stress-induced apoptosis in hepatic ischemia-reperfusion injury. Naunyn Schmiedebergs Arch. Pharmacol. 2020, 393, 1849–1858. [Google Scholar] [CrossRef] [PubMed]
  46. El Azab, E.F.; Mostafa, H.S. Geraniol ameliorates the progression of high fat-diet/streptozotocin-induced type 2 diabetes mellitus in rats via regulation of caspase-3, Bcl-2, and Bax expression. J. Food Biochem. 2022, 46, e14142. [Google Scholar] [CrossRef]
  47. Prasad, S.N. Protective effects of geraniol (a monoterpene) in a diabetic neuropathy rat model: Attenuation of behavioral impairments and biochemical perturbations. J. Neurosci. Res. 2014, 92, 1205–1216. [Google Scholar] [CrossRef]
  48. El-Said, Y.A.M.; Sallam, N.A.A.; Ain-Shoka, A.A.-M.; Abdel-Latif, H.A.-T. Geraniol ameliorates diabetic nephropathy via interference with miRNA-21/PTEN/Akt/mTORC1 pathway in rats. Naunyn Schmiedebergs Arch. Pharmacol. 2020, 393, 2325–2337. [Google Scholar] [CrossRef]
  49. Coelho, M.; Sabino, K.; Dalmau, S. Immunomodulatory effects of sucupira (Pterodon pubescens) seed infusion on collagen-induced arthritis. Clin. Exp. Rheumatol. 2004, 22, 213–218. [Google Scholar]
  50. Zhi, H.; Cui, J.; Yang, H.; Zhang, Y.; Zhu, M. Research progress of geraniol in tumor therapy. Proc. Anticancer Res. 2021, 5, 26–30. [Google Scholar] [CrossRef]
  51. Van Aller, R.; Nes, W. The phosphorylation of geraniol in germinating peas. Phytochemistry 1968, 7, 85–88. [Google Scholar] [CrossRef]
  52. Stobiecka, A. Comparative study on the free radical scavenging mechanism exerted by geraniol and geranylacetone using the combined experimental and theoretical approach. Flavour Fragr. J. 2015, 30, 399–409. [Google Scholar] [CrossRef]
  53. Tiwari, M.; Kakkar, P. Plant derived antioxidants–geraniol and camphene protect rat alveolar macrophages against t-BHP induced oxidative stress. Toxicol. In Vitro 2009, 23, 295–301. [Google Scholar] [CrossRef] [PubMed]
  54. Mostafa, A.M.; Allam, E.A.; Alghriany, A.A. Role of Psyllium Husk (Plantago ovata) on Liver Function Alterations Induced by Carbon Tetrachloride (CCl4) in Adult Male Albino Rats. Egypt. Acad. J. Biol. Sci. 2022, 14, 47–60. [Google Scholar] [CrossRef]
  55. Younis, N.S.; Abduldaium, M.S.; Mohamed, M.E. Protective effect of geraniol on oxidative, inflammatory and apoptotic alterations in isoproterenol-induced cardiotoxicity: Role of the Keap1/Nrf2/HO-1 and PI3K/Akt/mTOR pathways. Antioxidants 2020, 9, 977. [Google Scholar] [CrossRef]
  56. Ozkaya, A.; Sahin, Z.; Gorgulu, A.O.; Yuce, A.; Celik, S. Geraniol attenuates hydrogen peroxide-induced liver fatty acid alterations in male rats. J. Intercult. Ethnopharmacol. 2017, 6, 29. [Google Scholar] [CrossRef]
  57. Ozherelkov, S.; Timofeev, A.; Novikova, G.; Deeva, A.; Narovlianskiĭ, A.; Sanin, A.; Pronin, A. Protective effect of a new antiviral preparation of phosprenyl in experimental tick-borne encephalitis. Vopr. Virusol. 2000, 45, 33–37. [Google Scholar]
  58. Kandeil, M.A.; Mahmoud, M.O.; Abdel-Razik, A.-R.H.; Gomaa, S.B. Thymoquinone and geraniol alleviate cisplatin-induced neurotoxicity in rats through downregulating the p38 MAPK/STAT-1 pathway and oxidative stress. Life Sci. 2019, 228, 145–151. [Google Scholar] [CrossRef]
  59. Nennig, S.; Schank, J. The role of NFkB in drug addiction: Beyond inflammation. Alcohol Alcohol. 2017, 52, 172–179. [Google Scholar] [CrossRef]
  60. Hunter, C.J.; De Plaen, I.G. Inflammatory signaling in NEC: Role of NFKB and cytokines. Pathophysiol. Off. J. Int. Soc. Pathophysiol. ISP 2014, 21, 55. [Google Scholar]
  61. Jiang, K.; Zhang, T.; Yin, N.; Ma, X.; Zhao, G.; Wu, H.; Qiu, C.; Deng, G. Geraniol alleviates LPS-induced acute lung injury in mice via inhibiting inflammation and apoptosis. Oncotarget 2017, 8, 71038. [Google Scholar] [CrossRef] [PubMed]
  62. Ben Ammar, R.; Mohamed, M.E.; Alfwuaires, M.; Abdulaziz Alamer, S.; Bani Ismail, M.; Veeraraghavan, V.P.; Sekar, A.K.; Ksouri, R.; Rajendran, P. Anti-Inflammatory Activity of Geraniol Isolated from Lemon Grass on Ox-LDL-Stimulated Endothelial Cells by Upregulation of Heme Oxygenase-1 via PI3K/Akt and Nrf-2 Signaling Pathways. Nutrients 2022, 14, 4817. [Google Scholar] [CrossRef] [PubMed]
  63. Lee, H.J.; Jeong, H.S.; Kim, D.J.; Noh, Y.H.; Yuk, D.Y.; Hong, J.T. Inhibitory effect of citral on NO production by suppression of iNOS expression and NF-κB activation in RAW264. 7 cells. Arch. Pharm. Res. 2008, 31, 342–349. [Google Scholar] [CrossRef]
  64. Sharma, R.; Rao, R.; Kumar, S.; Mahant, S.; Khatkar, S. Therapeutic potential of citronella essential oil: A review. Curr. Drug Discov. Technol. 2019, 16, 330–339. [Google Scholar] [CrossRef]
  65. Rajendran, P.; Li, F.; Shanmugam, M.K.; Kannaiyan, R.; Goh, J.N.; Wong, K.F.; Wang, W.; Khin, E.; Tergaonkar, V.; Kumar, A.P. Celastrol Suppresses Growth and Induces Apoptosis of Human Hepatocellular Carcinoma through the Modulation of STAT3/JAK2 Signaling Cascade In Vitro and In Vivo Celastrol Inhibits STAT3 Signaling in HCC. Cancer Prev. Res. 2012, 5, 631–643. [Google Scholar] [CrossRef] [PubMed]
  66. Rajendran, P.; Li, F.; Shanmugam, M.K.; Vali, S.; Abbasi, T.; Kapoor, S.; Ahn, K.S.; Kumar, A.P.; Sethi, G. Honokiol inhibits signal transducer and activator of transcription-3 signaling, proliferation, and survival of hepatocellular carcinoma cells via the protein tyrosine phosphatase SHP-1. J. Cell. Physiol. 2012, 227, 2184–2195. [Google Scholar] [CrossRef] [PubMed]
  67. Rajendran, P.; Li, F.; Manu, K.A.; Shanmugam, M.K.; Loo, S.Y.; Kumar, A.P.; Sethi, G. γ-Tocotrienol is a novel inhibitor of constitutive and inducible STAT3 signalling pathway in human hepatocellular carcinoma: Potential role as an antiproliferative, pro-apoptotic and chemosensitizing agent. Br. J. Pharmacol. 2011, 163, 283–298. [Google Scholar] [CrossRef]
  68. Kuzu, B.; Cüce, G.; Ayan, İ.Ç.; Gültekin, B.; Canbaz, H.T.; Dursun, H.G.; Şahin, Z.; Keskin, İ.; Kalkan, S.S. Evaluation of apoptosis pathway of geraniol on Ishikawa cells. Nutr. Cancer 2021, 73, 2532–2537. [Google Scholar] [CrossRef]
  69. Sundin, T.; Peffley, D.M.; Gauthier, D.; Hentosh, P. The isoprenoid perillyl alcohol inhibits telomerase activity in prostate cancer cells. Biochimie 2012, 94, 2639–2648. [Google Scholar] [CrossRef]
  70. Zhang, L.; Lu, Z.; Zhao, X. Targeting Bcl-2 for cancer therapy. Biochim. Biophys. Acta Rev. Cancer BBA-REV CANCER 2021, 1876, 188569. [Google Scholar] [CrossRef]
  71. El Azab, E.F.; Saleh, A.M.; Yousif, S.O.; Mazhari, B.B.Z.; Abu Alrub, H.; Elfaki, E.M.; Hamza, A.; Abdulmalek, S. New insights into geraniol’s antihemolytic, anti-inflammatory, antioxidant, and anticoagulant potentials using a combined biological and in silico screening strategy. Inflammopharmacology 2022, 30, 1811–1833. [Google Scholar] [CrossRef] [PubMed]
  72. Jia, S.-S.; Xi, G.-P.; Zhang, M.; Chen, Y.-B.; Lei, B.; Dong, X.-S.; Yang, Y.-M. Induction of apoptosis by D-limonene is mediated by inactivation of Akt in LS174T human colon cancer cells. Oncol. Rep. 2013, 29, 349–354. [Google Scholar] [CrossRef] [PubMed]
  73. Ahmad, S.T.; Arjumand, W.; Seth, A.; Nafees, S.; Rashid, S.; Ali, N.; Sultana, S. Preclinical renal cancer chemopreventive efficacy of geraniol by modulation of multiple molecular pathways. Toxicology 2011, 290, 69–81. [Google Scholar] [CrossRef] [PubMed]
  74. Veena, M.S.P.; Mourisha, P.; Nithya, G.; Dhivya, S.; Ilakkia, A.; Sakthisekaran, D. Biochemical Studies on the Cytoprotective Efficacy of Geraniol in Benzo(a)pyrene Induced Experimental Lung Cancer. Am. J. PharmTech Res. 2012, 2, 360–371. [Google Scholar]
  75. Rajendran, P.; Abdelsalam, S.A.; Renu, K.; Veeraraghavan, V.; Ben Ammar, R.; Ahmed, E.A. Polyphenols as Potent Epigenetics Agents for Cancer. Int. J. Mol. Sci. 2022, 23, 11712. [Google Scholar] [CrossRef] [PubMed]
  76. Pavithra, P.; Mehta, A.; Verma, R.S. Essential oils: From prevention to treatment of skin cancer. Drug Discov. Today 2019, 24, 644–655. [Google Scholar] [CrossRef] [PubMed]
  77. Faraji, P.; Araj-Khodaei, M.; Ghaffari, M.; Dolatabadi, J.E.N. Anticancer Effects of Melissa officinalis: A Traditional Medicine. Pharm. Sci. 2021, 28, 355–364. [Google Scholar] [CrossRef]
  78. Magalhaes, M.; Manadas, B.; Efferth, T.; Cabral, C. Chemoprevention and therapeutic role of essential oils and phenolic compounds: Modeling tumor microenvironment in glioblastoma. Pharmacol. Res. 2021, 169, 105638. [Google Scholar] [CrossRef]
  79. Machado, T.Q.; da Fonseca, A.C.; Duarte, A.; Robbs, B.K.; de Sousa, D.P. A Narrative Review of the Antitumor Activity of Monoterpenes from Essential Oils: An Update. Biomed. Res. Int. 2022, 2022, 6317201. [Google Scholar] [CrossRef] [PubMed]
  80. Yang, W.; Chen, X.; Li, Y.; Guo, S.; Wang, Z.; Yu, X. Advances in pharmacological activities of terpenoids. Nat. Prod. Commun. 2020, 15, 1934578X20903555. [Google Scholar] [CrossRef]
  81. Madankumar, A.; Jayakumar, S.; Gokuladhas, K.; Rajan, B.; Raghunandhakumar, S.; Asokkumar, S.; Devaki, T. Geraniol modulates tongue and hepatic phase I and phase II conjugation activities and may contribute directly to the chemopreventive activity against experimental oral carcinogenesis. Eur. J. Pharmacol. 2013, 705, 148–155. [Google Scholar] [CrossRef] [PubMed]
  82. Vinothkumar, V.; Manoharan, S.; Sindhu, G.; Nirmal, M.R.; Vetrichelvi, V. Geraniol modulates cell proliferation, apoptosis, inflammation, and angiogenesis during 7, 12-dimethylbenz [a] anthracene-induced hamster buccal pouch carcinogenesis. Mol. Cell. Biochem. 2012, 369, 17–25. [Google Scholar] [CrossRef] [PubMed]
  83. Duan, S.; Xia, Y.; Tian, X.; Cui, J.; Zhang, X.; Yang, Q.; Zhao, T.; Lin, Y.; Zhang, F.; Zhang, X. A multi-bioresponsive self-assembled nano drug delivery system based on hyaluronic acid and geraniol against liver cancer. Carbohydr. Polym. 2023, 310, 120695. [Google Scholar] [CrossRef] [PubMed]
  84. Carnesecchi, S.; Bradaia, A.; Fischer, B.; Coelho, D.; Schöller-Guinard, M.; Gosse, F.; Raul, F. Perturbation by geraniol of cell membrane permeability and signal transduction pathways in human colon cancer cells. J. Pharmacol. Exp. Ther. 2002, 303, 711–715. [Google Scholar] [CrossRef] [PubMed]
  85. Li, M.; Tang, D.; Yang, T.; Qian, D.; Xu, R. Apoptosis triggering, an important way for natural products from herbal medicines to treat pancreatic cancers. Front. Pharmacol. 2022, 12, 3835. [Google Scholar] [CrossRef]
  86. Mukarram, M.; Choudhary, S.; Khan, M.A.; Poltronieri, P.; Khan, M.M.A.; Ali, J.; Kurjak, D.; Shahid, M. Lemongrass essential oil components with antimicrobial and anticancer activities. Antioxidants 2021, 11, 20. [Google Scholar] [CrossRef]
  87. Pu, Z.; Yang, F.; Wang, L.; Diao, Y.; Chen, D. Advancements of compounds targeting Wnt and Notch signalling pathways in the treatment of inflammatory bowel disease and colon cancer. J. Drug Target. 2021, 29, 507–519. [Google Scholar] [CrossRef]
  88. Duncan, R.E.; Lau, D.; El-Sohemy, A.; Archer, M.C. Geraniol and β-ionone inhibit proliferation, cell cycle progression, and cyclin-dependent kinase 2 activity in MCF-7 breast cancer cells independent of effects on HMG-CoA reductase activity. Biochem. Pharmacol. 2004, 68, 1739–1747. [Google Scholar] [CrossRef]
  89. Zhuang, K.; Tang, H.; Guo, H.; Yuan, S. Geraniol prevents Helicobacterium pylori-induced human gastric cancer signalling by enhancing peroxiredoxin-1 expression in GES-1 cells. Microb. Pathog. 2023, 174, 105937. [Google Scholar] [CrossRef]
  90. Chaudhary, S.C.; Siddiqui, M.S.; Athar, M.; Alam, M.S. Geraniol inhibits murine skin tumorigenesis by modulating COX-2 expression, Ras-ERK1/2 signaling pathway and apoptosis. J. Appl. Toxicol. 2013, 33, 828–837. [Google Scholar] [CrossRef]
  91. Fatima, K.; Wani, Z.A.; Meena, A.; Luqman, S. Geraniol exerts its antiproliferative action by modulating molecular targets in lung and skin carcinoma cells. Phytother. Res. 2021, 35, 3861–3874. [Google Scholar] [CrossRef] [PubMed]
  92. Manoharan, S.; Selvan, M.V. Chemopreventive potential of geraniol in 7, 12-dimethylbenz (a) anthracene (DMBA) induced skin carcinogenesis in Swiss albino mice. J. Environ. Biol. 2012, 33, 255. [Google Scholar] [PubMed]
  93. Jin, X.; Sun, J.; Miao, X.; Liu, G.; Zhong, D. Inhibitory effect of geraniol in combination with gemcitabine on proliferation of BXPC-3 human pancreatic cancer cells. Int. J. Med. Res. 2013, 41, 993–1001. [Google Scholar] [CrossRef]
  94. Lee, S.; Park, Y.R.; Kim, S.H.; Park, E.J.; Kang, M.J.; So, I.; Chun, J.N.; Jeon, J.H. Geraniol suppresses prostate cancer growth through down-regulation of E2F8. Cancer Med. 2016, 5, 2899–2908. [Google Scholar] [CrossRef] [PubMed]
  95. Qi, F.; Yan, Q.; Zheng, Z.; Liu, J.; Chen, Y.; Zhang, G. Geraniol and geranyl acetate induce potent anticancer effects in colon cancer Colo-205 cells by inducing apoptosis, DNA damage and cell cycle arrest. J. BUON 2018, 23, 346–352. [Google Scholar] [PubMed]
  96. Thapa, D.; Richardson, A.J.; Zweifel, B.; Wallace, R.J.; Gratz, S.W. Genoprotective effects of essential oil compounds against oxidative and methylated DNA damage in human colon cancer cells. J. Food Sci. 2019, 84, 1979–1985. [Google Scholar] [CrossRef] [PubMed]
  97. Crespo, R.; Montero Villegas, S.; Abba, M.C.; de Bravo, M.G.; Polo, M.P. Transcriptional and posttranscriptional inhibition of HMGCR and PC biosynthesis by geraniol in 2 Hep-G2 cell proliferation linked pathways. Biochem. Cell Biol. 2013, 91, 131–139. [Google Scholar] [CrossRef]
  98. Queiroz, T.; Santos, G.; Ventura, S.; Hiruma-Lima, C.; Gaivã, I.; Maistro, E. Cytotoxic and genotoxic potential of geraniol in peripheral blood mononuclear cells and human hepatoma cell line (HepG2). Genet. Mol. Res. 2017, 16, gmr16039777. [Google Scholar] [CrossRef]
  99. Sawada, S.; Okano, J.-i.; Imamoto, R.; Yasunaka, Y.; Abe, R.; Koda, M.; Murawaki, Y.; Isomoto, H. Preventive effect of geraniol on diethylnitrosamine-induced hepatocarcinogenesis in rats. Yonago Acta Med. 2016, 59, 37. [Google Scholar]
  100. Galle, M.; Crespo, R.; Rodenak Kladniew, B.; Montero Villegas, S.; Polo, M.; de Bravo, M.G. Suppression by geraniol of the growth of A549 human lung adenocarcinoma cells and inhibition of the mevalonate pathway in culture and in vivo: Potential use in cancer chemotherapy. Nutr. Cancer 2014, 66, 888–895. [Google Scholar] [CrossRef]
  101. Rodenak-Kladniew, B.; Gambaro, R.; Cisneros, J.S.; Huck-Iriart, C.; Padula, G.; Castro, G.R.; Chain, C.Y.; Islan, G.A. Enhanced anticancer activity of encapsulated geraniol into biocompatible lipid nanoparticles against A549 human lung cancer cells. J. Drug Deliv. Sci. Technol. 2023, 80, 104159. [Google Scholar] [CrossRef]
  102. De Fazio, L.; Spisni, E.; Cavazza, E.; Strillacci, A.; Candela, M.; Centanni, M.; Ricci, C.; Rizzello, F.; Campieri, M.; Valerii, M.C. Dietary geraniol by oral or enema administration strongly reduces dysbiosis and systemic inflammation in dextran sulfate sodium-treated mice. Front. Pharmacol. 2016, 7, 38. [Google Scholar] [CrossRef] [PubMed]
  103. Wittig, C.; Scheuer, C.; Parakenings, J.; Menger, M.D.; Laschke, M.W. Geraniol suppresses angiogenesis by downregulating vascular endothelial growth factor (VEGF)/VEGFR-2 signaling. PLoS ONE 2015, 10, e0131946. [Google Scholar] [CrossRef] [PubMed]
  104. El-Ella, D.M.A. Autophagy/apoptosis induced by geraniol through HIF-1α/BNIP3/beclin-1 signaling pathway in A549 CoCl2 treated cells. Adv. Pharm. Bull. 2022, 12, 155. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Chemical structure of Geraniol ((2E)-3,7-dimethylocta-2,6-dien-1-ol).
Figure 1. Chemical structure of Geraniol ((2E)-3,7-dimethylocta-2,6-dien-1-ol).
Molecules 28 03669 g001
Figure 2. GNL enhances antioxidant enzymes and inhibits inflammatory proteins.
Figure 2. GNL enhances antioxidant enzymes and inhibits inflammatory proteins.
Molecules 28 03669 g002
Figure 3. GNL inhibits inflammation by blocking NF-ĸB.
Figure 3. GNL inhibits inflammation by blocking NF-ĸB.
Molecules 28 03669 g003
Figure 4. Mechanisms of action of GNL in cancer treatment.
Figure 4. Mechanisms of action of GNL in cancer treatment.
Molecules 28 03669 g004
Table 1. Antitumor activities of GNL.
Table 1. Antitumor activities of GNL.
Type of CancerMode of StudyCell Lines/Animal ModelTargetEffectReferences
Skin cancerIn vivo/In vitroSwiss albino mice
PC-3, A431, and A549 cells
Swiss albino mice
Ras/Raf/ERK1/2
ornithine decarboxylase, LOX-5, and hyaluronidase
Phase II and Antioxidants
Apoptosis
Anti-proliferation
Chemoprevention
[90,91,92]
Pancreatic cancerIn vitroBXPC-3 cellsDNA damageApoptosis, Anti-proliferative[93,94]
Prostate cancerIn vitroPC-3E2F8 expressionAnti-proliferative[95]
Colon cancerIn vitroHT-29DNA damageApoptosis[96]
Liver cancerIn vitro/In vivoHepG2/RatsMevalonate pathway, HMGCR, DNA damage and ERK, NFkBApoptosis; anti-proliferative[83,97,98,99,100]
Lung CancerIn vitro/In vivoA549, Albino miceMevalonate pathway, DNA damage, ChemopreventionApoptosis; anti-proliferative[101,102]
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

Ben Ammar, R. Potential Effects of Geraniol on Cancer and Inflammation-Related Diseases: A Review of the Recent Research Findings. Molecules 2023, 28, 3669. https://doi.org/10.3390/molecules28093669

AMA Style

Ben Ammar R. Potential Effects of Geraniol on Cancer and Inflammation-Related Diseases: A Review of the Recent Research Findings. Molecules. 2023; 28(9):3669. https://doi.org/10.3390/molecules28093669

Chicago/Turabian Style

Ben Ammar, Rebai. 2023. "Potential Effects of Geraniol on Cancer and Inflammation-Related Diseases: A Review of the Recent Research Findings" Molecules 28, no. 9: 3669. https://doi.org/10.3390/molecules28093669

Article Metrics

Back to TopTop