Journal of Food Bioactives, ISSN 2637-8752 print, 2637-8779 online
Journal website www.isnff-jfb.com

Review

Volume 18, June 2022, pages 67-76


Ginger–a potential source of therapeutic and pharmaceutical compounds

Ravindra Verma*, Prakash S. Bisen

School of Studies in Biotechnology, Jiwaji University, Gwalior 474001, India
*Corresponding author: Ravindra Verma, School of Studies in Biotechnology, Jiwaji University, Gwalior 474001, India. Tel: 91-11-7999212685; E-mail: vermaravindra917@gmail.com
DOI: 10.31665/JFB.2022.18309

Received: March 30, 2022
Revised received & accepted: June 24, 2022

Abstract▴Top 

Ginger is traditionally known for its therapeutic and pharmaceutical properties. It has been used widely to treat various health problems such as high blood pressure, coughs, colds, swelling, nausea, rheumatic disorders, vomiting, bronchitis, indigestion, gastric ulcers, and behavioral problems. Shogaol and Gingerol are anti-inflammatory, anti-fever, anti-pain, and anti-cough compounds that may help treat a cold. This review provides an up-to-date understanding of the impact of ginger and its active compounds on human health. Various ginger compounds such as gingerol, shogaols, zingiberene, zingerone, paradols and zingerone are receiving attention for their clinical applications and pharmaceutical properties. Studies indicate that ginger is anti-inflammatory, anti-tumor, antimicrobial, antiemetic, hepatoprotective, and neuroprotective. During the inflammatory response, ginger inhibits (NF-κB) and immune system activation in addition to many other cellular processes. Ginger has shown benefits in preclinical and clinical studies for neurology, cardiovascular disease, and cancer. These findings indicate the necessity for further in vivo and clinical studies.

Keywords: Ginger; Immunity; Pharmaceutical; Phytocompounds; Therapeutic

1. Introduction▴Top 

Ginger is a spice widely used in traditional medicine and as an ingredient in food. Zingiber officinale is a plant belonging to the Zingiberaceae family. Originally, it was mentioned in Confucius’s Analects (475-221 BC) as a spice for medicinal purposes (Pickersgill, 2005). Scientists have examined the effects of ginger on diseases such as asthma, stroke, diabetes, constipation, nervous disorders, etc., due to its antiviral and antioxidant properties (Butt and Sultan, 2011; Karna, et al., 2012; Mashhadi, et al., 2013). It is an underground rhizome that grows up to 75 cm in height. Ginger grows mainly in tropical and subtropical regions (Vijayan et al., 2020). China is the largest exporter, followed by the Netherlands, Thailand, Peru, and India, while the US is the major importer, followed by Japan, the Netherlands, Pakistan, and Germany. It contains mainly dietary fiber, vitamin E, vitamin B6, iron, magnesium, manganese, potassium, and selenium. In South Asian countries, it is used in many traditional formulations, especially in Ayurveda, a traditional Indian medical system. Therapeutically, it has been used in many Ayurvedic formulations from the ancient period and is also known as “Maha Aushadhi-A Great Medicine” (Narayana, et al., 2000; Abdulwase, et al., 2020). The United States and European countries registered and sold it as a nutraceutical against nausea, motion sickness, and migraine. It is an approved herb by the German Commission E Monographs for dyspepsia and prevents motion sickness (Vasala, 2012; www.herbalgram.org). Ginger is known by different brand names, like African ginger, black ginger, cochin ginger, Imber, Jamaica ginger, race ginger, rhizoma zingerberis, rhizome, sheng jiang, shokyo, zingibain, Zingiber officinale, and Zingiberis. A controlled trial on abdominal distention in post-cesarean women found that the quality of life and the number of patients who could eat was higher in the ginger group than the others. This study confirms that ginger is efficacious in abdominal distention, low-priced herbal folk medicine with minimum side effects (Tianthong and Phupong, 2018). A trial on postoperative nausea and vomiting observed that 1 g of ginger prevents vomiting and nausea. A minor side effect of abdominal discomfort was seen (Chaiyakunapruk et al., 2006). Its anti-inflammatory and antioxidant properties proved its worth in the pharmaceutical industry. Its phytocompounds like gingerols modulate the biochemical pathways and specific biomarkers activated in chronic inflammation (Yusof, 2016). The antioxidant properties due to 6-gingerol protect cell membrane lipids during oxidation and scavenging free radicals (Grzanna, et al., 2005; Han et al., 2022). The most important constituents are 6-gingerol, 6-paradol, and shogaol and zingerone, which are mainly responsible for the anticancer properties of ginger (Shukla and Singh, 2007; Zhang et al., 2021b). The scientific literature supports the use of ginger in the treatment of a variety of ailments, including osteoarthritis, neurological disorders, rheumatoid arthritis, diabetes, respiratory distress, liver diseases, and primary dysmenorrhea (Mahomoodally et al., 2021; Kiyama, 2020; Huang, et al., 2019; Khan et al., 2016; Jafarzadeh and Nemati, 2018; Bhaskar et al., 2020).

2. Pharmaceutical potential of ginger▴Top 

The ginger constituents vary depending on origin and whether the rhizomes are fresh or dry. A ginger root is largely composed of carbohydrates, fats, proteins, fiber, vitamins, minerals, water, ash, and volatile oils. The phytocompounds observed in fresh ginger are zingerone, gingerol, zingiberene, β-sesquiphellandrene, shogaol, bisabolene (α-form), β-phellandrene, farnesene, 1, 4-cineole, citral, camphene, 6-paradol, curcumene, terpineol (α-form), borneol, β-elemene, zingiberenol, limonene, geraniol, linalool (Jolad, et al., 2005; Mbaveng and Kuete, 2017; Ali et al., 2008). Ginger is receiving attention for its clinical applications and pharmaceutical properties due to gingerol, shogaols, zingiberene, zingerone, paradols, etc. Among these phytocompounds, gingerols are the primary components of several bioactivities (Kubra and Rao, 2012). The structure of selected phytocompounds is shown in Figure 1.


Click for large image
Figure 1. Chemical structure of important phytocompounds.

The potential pharmacological activities of phytocompounds increase the significant investigations and clinical trials on the health benefits and ginger efficacy. In dry ginger, 6-gingerol transformed to 6-shogaol, which is more stable and has more powerful pharmacological effects. Further, 6-shogaol biotransformed to 6-Paradol (Jafarzadeh et al., 2021). The fiery taste of ginger is due to zingerone, the major phytochemical used for medicinal purposes. Ginger is also used as a primary food additive due to its fragrant and spicy characteristics (Srinivasan, 2017).

2.1. Gingerols

The gingerols are an important group of polyphenols isolated from the fresh root of ginger. Ginger rhizomes are one of the richest sources of ginger-derived bioactives (6-gingerol, 8-gingerol, and 10-gingerol). 6-Gingerol contains a 5-hydroxydecan-3-one moiety that is substituted by a 4-hydroxy-3-methoxyphenyl group at position 1; thought to inhibit adipogenesis. This beta-hydroxy ketone belongs to the group of plant metabolites known as guaiacols. It is an antineoplastic agent. It is rich in antioxidants, cardiotonics, and cardioprotective agents, including [8]-gingerol (Xue, et al., 2021b). Inhibition of leukotriene A (4) hydrolase expression and induction of G2/M arrest have been shown to be biochemical mechanisms by which 6-gingerol plays an anti-colorectal cancer role (Kumara et al., 2017).

2.2. Shogaol

Shogaols are bioactive compounds found in ginger that have gastroprotective and neuroprotective properties. [6]-Shogaol is a monomethoxybenzene, which belongs to the phenols and enones. Using a Mannich reaction, Mase and his colleagues synthesized shogaol using dimethylammonium dimethylcarbamate (DIMCARB), an ionic liquid (Mase et al., 2010). 6-Shogaol inhibits the infiltration of leukocytes into inflamed tissue, reducing edema swelling. 6-Shogaol affects pathways such as NFκB and MAPK, and regulates the cytoprotective HO-1 in both vitro and in vivo (Bischoff-Kont and Fürst, 2021). In addition to scavenging free radicals, the 6-, 8-, and 10-shogaols also exhibit strong anti-proliferative activity against human lung cancer cell lines (Sang et al., 2009). Previous studies suggest that 6-shogaol inhibits glial cell activation and has anti-oxidant properties against neurological disorders (Moon et al., 2014).

2.3. Paradols

[6]-paradol belongs to the class of phenols, a monomethoxybenzene, and ketone. It controls several obesity-related genes without triggering AMPK. 6-paradol decreases body weight gain and visceral and subcutaneous fat in mice after 2 weeks. It decreased liver cholesterol, triglyceride production, fatty acid synthesis, and lipid transport, as well as adipocyte differentiation, both in liver and adipose tissue (Hattori, et al., 2021). 6-shogaol is almost completely metabolized into 6-paradol, despite the fact that 6-paradol is only a minor component of ginger, mainly derived from 6-gingerol (Chen et al, 2012). According to a study evaluating the concentration-dependent effects of 6-paradol on glucose utilization, 6-paradol inhibits the synthesis of lipid in 3T3-L1 cells, reducing cellular lipid accumulation in a concentration-dependent manner, in addition to reducing insulin-induced lipid accumulation (Wei et al, 2017).

2.4. Zingerone

Zignerone is a 4-phenylbutan-2-one that contains a methoxy and hydroxy group at position 3 and 4 of the phenyl ring, respectively. It is responsible for the pungent flavor in ginger and may be used as an antioxidant, as an anti-inflammatory agent, as an antiemetic, as a flavouring, as a fragrance, and as a plant metabolite (Baliga, et al., 2011). Zingerone reduced oxidative stress and ameliorated inflammation, and levels of antioxidant enzymes. Oral administration was suggested as a treatment for rheumatoid arthritis. It significantly lowered levels of NF-κB, TGF-β, TNF-α, IL-1β, IL-6 and Hs-CRP while significantly increasing levels of IL-10 (Bashir et al., 2021). By regulating AMPK, Gingerone prevents hepatic inflammation, oxidative stress, and apoptosis (Mohammed, 2022).

2.5. Zingiberene

It is a sesquiterpene and a cyclohexadiene that contains a hydrogen at the 5 position that is substituted by a 6-methyl-hept-5-en-2-yl group (R configuration). It is found in dried ginger rhizomes, Zingiber officinale (indonesian ginger). Researchers observed that ginger rhizomes contained terpenes like zingiberene that inhibited MAO-A enzyme activity (Kukula-Koch et al., 2018). Experimental observations indicate that zingiberene can be exploited as a natural and novel therapeutic in preventing oxidative damage in neurodegenerative disorders (Togar et al., 2015a). Researchers have examined the cytotoxic, genotoxic, and antioxidant properties of zingiberene in an in vitro culture of rat brain cells and discovered that this compound could be potentially used as a natural anticancer agent (Togar et al., 2015b).

2.6. Zerumbone

Zerumbone is a sesquiterpene and cyclic ketone derived from (1E,4E,8E)-alpha-humulene, which can be obtained by steam distillation from ginger grown in Southeast Asia. As a plant metabolite, it is also useful as an anti-inflammatory drug for gliomas, as well as being an inhibitor of certain oncogenes (Rahman, et al., 2013). Researchers have found that zerumbone can prevent Zearalenone-induced liver injury and have presented the molecular basis for potential uses of zerumbone in treating liver injuries from Zearalenone (AbuZahra et al., 2021).

Recent studies have also shown that Zerumbone inhibits inflammation by inhibiting NF-κB and TLR. Zerumbone could potentially be used for the treatment and prevention of diabetes and its complications (Kim et al., 2022). Researchers found that zerumbone increased BAX, caspase-7, and caspase-9 expression and decreased BCL-2 expression, leading to apoptotic cell death induced by paclitaxel and proapoptotic proteins. By enhancing intracellular ROS-mediated oxidative stress, zerumbone promotes resensitization of breast cancer cells to PTX (Li et al., 2022).

3. Therapeutic potential▴Top 

A potential source of functional food additives for cancer prevention and treatment is ginger (Köngül and Şeker 2021). Gingerol is a phytocompound that is capable of resisting the proteases required for Coronavirus entry and replication (Oso, et al., 2020). Ginger extract reduces nausea and vomiting through inhibition of the cholinergic M3 receptors and the serotonergic 5-HT3 receptors associated with breast cancer (Pertz, et al., 2011). The effects of ginger on lipid profiles are noteworthy. Studies have shown that ginger has favorable effects on triacylglycerol (TAG) and low-density lipoprotein cholesterol (LDL-C) (Pourmasoumi et al., 2018; Jafarnejad, et al., 2017; Mazidi, et al., 2016). The phytocompounds in ginger extract and their associate diseases and disorders with molecular target are mentioned in Table 1.


Click to view
Table 1.

The phytocompounds in ginger extract and their associate diseases and disorders with molecular target
 

3.1. In cardiovascular

The world’s leading cause of death is cardiovascular disease. Diabetes, high cholesterol, triglycerides, and high blood pressure causes these problems. Rat serum samples tested for the effects of ginger doses (500 mg/kg) revealed a significant reduction in cholesterol levels, but no change in triglycerides (Thomson et al., 2002). A study found a dose of 5 grams or more is required to show significant antiplatelet activity (Nicoll and Henein, 2009). Ginger inhibits reactive oxygen species (ROS), inducible nitric oxide synthase (iNOS), superoxide dismutase (SODs), glutathione, and heme oxygenase (Roudsari et al., 2021). 6-gingerol (20 μM) is a novel phytocompound from ginger that improves cardiac function and alleviates pressure overload-induced cardiac changes in a p38-dependent manner (Ma et al., 2021). 6-gingerol is a promising drug for the treatment of pathobiological cardiac remodeling (Xu et al., 2018; Lv et al., 2018; Ren et al., 2019). CVD inflammation triggers proinflammatory cytokines such as tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6). Studies have shown that ginger reduces TNF-α values, while 6-gingerol reduces inflammatory factors and nitric oxide synthase enzyme levels (Morvaridzadeh et al., 2020). The increase in evidence for Nrf2 and its downstream targets is likely responsible for preventing CVD development, including the development of endothelial dysfunction and atherosclerosis induced by oxidative stress. In the early stages of atherosclerosis, endothelium dysfunction is marked by an increase in blood flow, cellular permeability, LDL oxidation, monocyte adherence, platelet activation, vascular inflammation, and the proliferation and infiltration of smooth muscle cells from the media to the arteries (da Costa et al., 2019; Zhang et al., 2021a). The reduction of antioxidant enzyme protein expression was found in LDLR−/− mice models transplanted with NRF2-deficient bone marrow cells. Migratory macrophages, inflammatory cytokines, and atherosclerotic lesions were also observed after transplant of Nrf2-deficient bone marrow cells (Mimura and Itoh, 2015; Ooi et al., 2018). In the meantime, silencing Nrf2 in U937 monocytic cells led to elevations in proinflammatory cytokines such as interleukin-1β (IL-1β ), interleukin-6 (IL-6), tumour necrosis factor alpha (TNF-α), monocyte chemotactic protein-1 (MCP-1) and endoplasmic reticulum (ER) stress markers. Nrf2 deficiency also increased the production of pro-inflammatory cytokines such as (MCP-1, IL-6, and TNF-α) within macrophages, which led to foam cell formation (Ooi et al., 2018). A study found that bioactive molecules in ginger compounds inhibited the expression of Keap1, which resulted in a reduction in Nrf2, which subsequently elevated the levels of Nrf2 and other downstream antioxidant enzymes (Gao et al., 2022). Several other aspects of cardiovascular disease can also be treated with ginger. A study indicates that aqueous ginger extract lowers blood pressure and reduces heart palpitations by stimulating muscarinic receptors and blocking Ca++ channels (Ghayur, et al., 2005). A daily supplement of ginger in specific amounts has been found beneficial for heart health. Ginger’s extract exhibits moderate inhibitory properties against the Monoamine oxidase-A (MAO-A) enzyme, which is an effective antidepressant, as well as a panic disorder agent (Kukula-Koch et al., 2018). An investigation examined the cardioprotective effects of 8-Gingerol in isoproterenol (ISO)-induced MI. in rats ISO (85 mg/kg/d) was subcutaneously injected over 2 consecutive days to induce an acute MI model. There was an association between 8-Gin and relaxation of oxidative stress, cardiomyocyte apoptosis inhibition, and regulation of Ca2+ homeostasis through modulation of ICa-L (Xue et al., 2021a). Another study investigated the possible mechanisms by which 8-Gin (10 and 20 mg/kg/d) reduced J-point elevation and heart rate in mice induced with isoproterenol (ISO-10mg/kg/d) for 2 weeks. It was found that 8-Gin exerted cardioprotective effects on ISO-induced MF, which is likely associated with its inhibition of ROS generation, apoptosis, and autophagy by modulating the PI3K/Akt/mTOR signaling pathway (Xue et al., 2021b).

3.2. Anticancer agent

The term “cancer” refers to a group of disorders characterized by abnormal cell growth that may invade and spread. The cancer death rate is 15.7% (eighteen million deaths a year) (Wang, et al., 2016). A study demonstrated that 6-gingerol had anti-tumorigenic properties by affecting cell cycle regulation and apoptosis by increasing caspase activity during the G0/G1 phase of the cell cycle (Park et al., 2006; Wang et al., 2014). 8-gingerol is a derivative classified according to its ability to inhibit the growth of cancerous cells and blood vessel formation as well as its ability to suppress platelet growth. A further class of bioactive derivative is 10-gingerol, which decreases cell proliferation and inhibits cellular expression (Kubra and Rao, 2012; Zaid et al., 2022). In a pilot study that investigated ginger’s effects on proliferation, apoptosis, and differentiation in normal-appearing colonic mucosa; it was found that 2g of ginger reduced proliferation and increased apoptosis and differentiation relative to proliferation in the crypt differentiation zone (Citronberg, et al., 2013). Ginger aqueous extract contains polyphenols and flavonoids-flavonols that have anticancer properties (Choudhury, et al., 2010). In colorectal cancer (CRC), cyclooxygenase (COX)-produced prostaglandin E2 (PGE2) tissues increase, which is an early sign of the disease. The ginger root significantly reduced the expression of COX-1 protein, which is responsible for an increased risk of CRC. It shows that ginger has the potential to reduce eicosanoid levels and to reduce inflammation in the colon (Jiang, et al., 2013; Zick, et al., 2011). Ginger is a chemopreventive (Zick et al., 2015). Ginger has been found to have anti-skin-cancer chemopreventive effects. Ginger ethanol extract pre-applied to the skin of Sensitivity to carcinogenesis mice model significantly inhibited tissue plasminogen activator (TPA)-induced cyclooxygenase and lipoxygenase activities, ODC mRNA expression, and epidermal edema and hyperplasia (Katiyar, et al., 1996). Research has established that gingerol is able to scavenge the peroxyl radicals produced during pulse radiolysis (Park et al., 1998; Ahmad et al., 2001; Chauhan et al., 2021). 6-gingerol inhibits tumor growth and metastasis through the inhibition of cyclooxygenase-2 (COX-2) and the p38 MAPK-NF-κB pathway (mitogen-activated protein kinase-necrosis factor-kappa B) (Kim et al., 2005a; Kim et al., 2005b). The possible mechanisms of gingerol’s effects on treating liver cancer were studied by network pharmacology, in-silico experiments, as well as in-vitro experiments on human liver cancer HepG2 cells. Molecular docking revealed that gingerol derivatives had good PI3K and Akt binding activity. Researchers report that gingerol derivatives and compound gingerol (compound gingerol is composed of 6gingerol, 8gingerol, and 10gingerol in a ratio of 7:1.5:1.5) inhibit HepG2 cell proliferation, and each administration group can significantly increase the apoptosis rate of HepG2 cells and the fluorescence intensity of the nucleus, thereby blocking the cell cycle. In Western Blot and real-time quantitative PCR experiments, gingerol derivatives and compound gingerol had a significant effect on decreasing the expression of Akt and p-Akt and increasing the expression of Bax/Bcl-2 (Su, et al., 2022). Cancer can be prevented and treated with ginger. Clinical trials are needed to support the efficacy of the drug. In colon cancer, the signaling pathway PI3K-Akt involves the serine/threonine kinase Akt, as well as the genes AKT1, PIK3CA, MAPK3, and TP53. Human colon cancer is associated with a misregulation of the PI3K-Akt pathway, the most commonly activated signaling pathway in cancer (Wang et al., 2020b; Li et al., 2020). In the study, phosphoinositol 3-kinase signaling was found to be an active biological process in ginger in controlling cancer-related pathways through interactions with PI3K/Akt (Kiyama, 2020). Ginger’s anticancer mechanism is directly influenced by its PI3K-Akt signaling pathways and EGFR kinase inhibitor resistance (Zhang et al., 2021b).

3.3. Neurodegenerative agent

Several studies have shown ginger to be analgesic (Phillips et al., 1993; Black, et al., 2010; Rondanelli, et al., 2020). In Alzheimer’s disease (AD), abnormal aggregation of neurons and synaptic damage is caused by abnormal accumulation of amyloid beta (Aβ) and tau proteins (Busche and Hyman, 2020). Aβ is considered by many to be a key pathogen that contributes to AD pathogenesis, but the exact mechanisms are still unclear. Several studies have found protective effects of ginger on amyloid beta (Aβ) neurotoxicity. The ginger treatment inhibited the inflammatory markers NF-κB and interleukin (IL)-1β resulting in behavioral dysfunction and neuronal death caused by Aβ1–40 plaques as well as increasing antioxidant enzymes superoxide dismutase and catalase (SOD and CAT) (Choi, et al., 2018). There is evidence that 6-gingerone, 6-shogaol, 6-paradol, and dehydrozingerone may provide benefits in diseases related to aging and neurological disorders (Ma et al., 2021; Srinivasan et al., 2019). Researchers assessed acetylcholinesterase (AChE) as the most promising molecular target for ginger compounds with multiple molecular targets in Alzheimer’s disease (Azam, et al., 2014; Wheeler, 2003). Additionally, ginger compounds inhibit the activity of butyrylcholinesterase (BChE) by binding to Trp82 and Tyr332 residues (Cuya and França, 2020; Cuya et al., 2018; Schepici, et al., 2021). A reduction in clinical symptoms of mice with Experimental autoimmune encephalomyelitis (EAE) as well as a reduction of expression of IL-27 and IL-33 in their spinal cords is also observed (Jafarzadeh et al, 2014). In studies on migraine headaches, ginger was observed to have both abortive and prophylactic effects without causing any side effects (Mustafa and Srivastava, 1990). In a meta-analysis of placebo-controlled randomized controlled trials (RCTs), ginger was shown to be very effective and safe in treating migraine patients with pain outcomes as measured after two hours (Chen and Cai, 2021).

3.4. Antiviral

The antiviral and immune-modulating properties of herbal extracts have been extensively studied (Das, et al., 2021; Yasmin et al., 2020; Mukhtar, et al., 2008). Studies have reported that allicin from ginger has anti-influenza properties (Hornung, et al., 1994; Sahoo et al., 2016). The Dengue virus infection significantly enhanced matrix metalloproteinase (MMP)-2 proteolytic activity, but not matrix metalloproteinase-9. Ethylenediaminetetraacetic acid (EDTA-MMP inhibitor) reduced this enhancement. An aqueous extract from Zingiber officinale Roscoe was confirmed to modulate expression levels of MMP-2, MMP-9, TIMP-1, and TIMP-2 in a concentration-dependent manner. Gingerols and shogaols, which are naturally occurring compounds in Zingiber officinale, have been associated with its anti-MMP activity (Sharma, et al., 2015). In vitro, the anti-chikungunya effect of Zingiber officinale (62.50 μg/mL) aquatic plant extract was tested using the Vero cell line. Additionally, Zingiber officinale is used as an antiviral treatment against Chikungunya virus that prevents drug resistance (Kaushik, et al., 2020). Fresh ginger blocks the human respiratory syncytial virus (HRSV) (San Chang et al., 2013). Ginger chemicals as antiviral agents against a variety of viruses, as well as their mechanisms of action, Table 2.


Click to view
Table 2.

Ginger extract as an antiviral agent
 

Recently, several studies have focused their attention on the interaction between ginger compounds and the spike protein of Coronaviruses. PLPro (papain-like protease) and 3CLPro (chymotrypsin-like protease) are essential for the survival of SARS-CoV-2 in humans (Li, et al., 2020). Natural products are being investigated as a potential targeted therapy to treat SARS-CoV-2. A structure-based molecular docking study found that ginger phytocompounds like 8-Gingerol, 10-Gingerol, and 6-Gingerol are potent inhibitors of PLpro, with high affinity for binding (Goswami, et al., 2020). Ginger components were shown to have high binding affinity to the protease (Mpro) of CoV-2, the virus that causes SARS (Jahan et al., 2021).

4. Conclusion▴Top 

The promising target profiles of natural compounds enable them to be considered as ideal drug development candidates since they contain signature events that are dispersed throughout. There are several compounds in ginger extract that have antiviral and antibiotic properties. Ginger extract can be used as a treatment for various diseases. Ginger is considered a natural alternative to treat cancer and other diseases due to its phytochemical properties and pharmacological effects. It is a popular healthy food because of its phytochemical properties and pharmacological effects. Ginger compounds can relieve fevers, reduce pain, and suppress coughs, making them helpful to minimize the effects of a winter cold. It has been shown to be effective in treating several ailments such as high blood pressure, cough, cold, swelling, nausea, rheumatoid disorders, vomiting, bronchitis, indigestion, gastric ulcers, infectious diseases, and poisonings. There is evidence that ginger is anti-inflammatory, anti-cancer, anti-tumor, anti-microbial, antiemetic, hepatoprotective, and neuroprotective. Clinical trials show that ginger can reduce nausea and vomiting in women during early pregnancy. Ginger inhibits proinflammatory cytokines, including nuclear factor kappa B (NF-κB) and immune system activation, as well as many other cellular processes. Ginger inhibits reactive oxygen species, inducible nitric oxide synthase, superoxide dismutase, glutathione, heme oxygenase, and inducible nitric oxide synthase. Research on ginger and its bioactive components, including both preclinical and clinical studies, is shown to have positive effects on cardiovascular disease, neurology, and cancer. These results reinforce the need for further In vivo and clinical studies.

Acknowledgments

None.

None.

Conflict of interest

The authors declare no conflict of interest.


References▴Top 
Abdulwase, R., Abbas, A.B., and Yan, S. (2020). Ginger as a commercial product in China. MOJ Biol. Med. 5(1): 1–2.
AbuZahra, H.M., Rajendran, P., and Ismail, M.B. (2021). Zerumbone Exhibit Protective Effect against Zearalenone In-Duced Toxicity via Ameliorating Inflammation and Oxidative Stress Induced Apoptosis. Antioxidants 10(10): 1593.
Ahmad, N., Katiyar, S.K., and Mukhtar, H. (2001). Antioxidants in chemoprevention of skin cancer. Curr. Probl. Dermatol. 29: 128–139.
Ali, B.H., Blunden, G., Tanira, M.O., and Nemmar, A. (2008). Some phytochemical, pharmacological and toxicological properties of ginger (Zingiber officinale Roscoe): a review of recent research. Food Chem. Toxicol. 46(2): 409–420.
Azam, F., Amer, A.M., Abulifa, A.R., and Elzwawi, M.M. (2014). Ginger components as new leads for the design and development of novel multi-targeted anti-Alzheimer’s drugs: A computational investigation. Drug Des. Devel. Ther. 8: 2045.
Baliga, M.S., Haniadka, R., Pereira, M.M., D’Souza, J.J., Pallaty, P.L., Bhat, H.P., and Popuri, S. (2011). Update on the chemopreventive effects of ginger and its phytochemicals. Crit. Rev. Food Sci. Nutr. 51(6): 499–523.
Bashir, N., Ahmad, S.B., Rehman, M.U., Muzamil, S., Bhat, R.R., Mir, M.U.R., Shazly, G.A., Ibrahim, M.A., Elossaily, G.M., Sherif, A.Y., and Kazi, M. (2021). Zingerone (4-(four-hydroxy-3-methylphenyl) butane-two-1) modulates adjuvant-induced rheumatoid arthritis by regulating inflammatory cytokines and antioxidants. Redox Rep. 26(1): 62–70.
Bhaskar, A., Kumari, A., Singh, M., Kumar, S., Kumar, S., Dabla, A., Chaturvedi, S., Yadav, V., Chattopadhyay, D., and Prakash Dwivedi, V. (2020). [6]-Gingerol exhibits potent anti-mycobacterial and immunomodulatory activity against tuberculosis. Int. Immunopharmacol. 87: 106809.
Bischoff-Kont, I., and Fürst, R. (2021). Benefits of ginger and its constituent 6-shogaol in inhibiting inflammatory processes. Pharmaceuticals 14(6): 571.
Black, C.D., Herring, M.P., Hurley, D.J., and O’Connor, P.J. (2010). Ginger (Zingiber officinale) reduces muscle pain caused by eccentric exercise. J. Pain 11(9): 894–903.
Busche, M.A., and Hyman, B.T. (2020). Synergy between amyloid-β and tau in Alzheimer’s disease. Nat. Neurosci. 23(10): 1183–1193.
Butt, M.S., and Sultan, M.T. (2011). Ginger and its health claims: molecular aspects. Crit. Rev. Food Sci. Nutr. 51(5): 383–393.
Chaiyakunapruk, N., Kitikannakorn, N., Nathisuwan, S., Leeprakobboon, K., and Leelasettagool, C. (2006). The efficacy of ginger for the prevention of postoperative nausea and vomiting: a meta-analysis. Am. J. Obstet. Gynecol. 194(1): 95–99.
Chakraborty, D., Mukherjee, A., Sikdar, S., Paul, A., Ghosh, S., and Khuda-Bukhsh, A.R. (2012). [6]-Gingerol isolated from ginger attenuates sodium arsenite induced oxidative stress and plays a corrective role in improving insulin signaling in mice. Toxicol. Lett. 210(1): 34–43.
Chauhan, A., Islam, A., Prakash, H., and Singh, S. (2021). Phytochemicals targeting NF-κB signaling: Potential anti-cancer interventions. J. Pharm. Biomed. Anal.
Chen, H., Lv, L., Soroka, D., Warin, R.F., Parks, T.A., Hu, Y., Zhu, Y., Chen, X., and Sang, S. (2012). Metabolism of [6]-shogaol in mice and in cancer cells. Drug Metab. Dispos. 40(4): 742–753.
Chen, L., and Cai, Z. (2021). The efficacy of ginger for the treatment of migraine: A meta-analysis of randomized controlled studies. Am. J. Emerg. Med. 46: 567–571.
Choi, J.G., Kim, S.Y., Jeong, M., and Oh, M.S. (2018). Pharmacotherapeutic potential of ginger and its compounds in age-related neurological disorders. Pharmacol. Ther. 182: 56–69.
Choudhury, D., Das, A., Bhattacharya, A., and Chakrabarti, G. (2010). Aqueous extract of ginger shows antiproliferative activity through disruption of microtubule network of cancer cells. Food Chem. Toxicol. 48(10): 2872–2880.
Citronberg, J., Bostick, R., Ahearn, T., Turgeon, D.K., Ruffin, M.T., Djuric, Z., Sen, A., Brenner, D.E., and Zick, S.M. (2013). Effects of ginger supplementation on cell-cycle biomarkers in the normal-appearing colonic mucosa of patients at increased risk for colorectal cancer: results from a pilot, randomized, and controlled trial. Cancer Prev. Res. (Phila) 6(4): 271–281.
Cuya, T., and França, T.C.C. (2020). A molecular modeling study of components of the ginger (zingiber officinale) extract inside human butyrylcholinesterase: Implications for Alzheimer disease. J. Biomol. Struct. Dyn. 38(9): 2809–2815.
Cuya, T., Baptista, L., and Celmar Costa França, T. (2018). A molecular dynamics study of components of the ginger (Zingiber officinale) extract inside human acetylcholinesterase: Implications for alzheimer disease. J. Biomol. Struct. Dyn. 36(14): 3843–3855.
da Costa, R.M., Rodrigues, D., Pereira, C.A., Silva, J.F., Alves, J.V., Lobato, N.S., and Tostes, R.C. (2019). Nrf2 as a potential mediator of cardiovascular risk in metabolic diseases. Front. Pharmacol. 10: 382.
Das, G., Heredia, J.B., de Lourdes Pereira, M., Coy-Barrera, E., Rodrigues Oliveira, S.M., Gutierrez-Grijalva, E.P., Cabanillas-Bojorquez, L.A., Shin, H.S., and Patra, J.K. (2021). Korean traditional foods as antiviral and respiratory disease prevention and treatments: A detailed review. Trends Food Sci. Technol. 116: 415–433.
Eid, B.G., Mosli, H., Hasan, A., and El-Bassossy, H.M. (2017). Ginger ingredients alleviate diabetic prostatic complications: Effect on oxidative stress and fibrosis. Evid. Based Complement. Alternat. Med. 2017: 6090269.
El-Adawi, H., El-Demellawy, M., and El-Wahab, A.A. (2011). Some medicinal plant extracts exhibit potency against viral hepatitis C. J. Biosci. Tech. 2(1): 223–231.
Gan, F.F., Ling, H., Ang, X., Reddy, S.A., Lee, S.S., Yang, H., Tan, S.H., Hayes, J.D., Chui, W.K., and Chew, E.H. (2013). A novel shogaol analog suppresses cancer cell invasion and inflammation, and displays cytoprotective effects through modulation of NF-κB and Nrf2-Keap1 signaling pathways. Toxicol. Appl. Pharmacol. 272(3): 852–862.
Ganaie, M.A., Al Saeedan, A., Madhkali, H., Jan, B.L., Khatlani, T., Sheikh, I.A., Rehman, M.U., and Wani, K. (2019). Chemopreventive efficacy zingerone (4-[4-hydroxy-3-methylphenyl] butan-2-one) in experimental colon carcinogenesis in Wistar rats. Environ. Toxicol. 34(5): 610–625.
Gao, Z., Ren, Y., Liu, B., Ma, R., Li, F., Li, D., and Wang, Y. (2022). Ginger constituents ameliorated B (α) P-induced toxicity via modulation of antioxidants and xenobiotic-metabolising enzymes in mice. Int. Food Res. J. 29(2): 433–445.
GBD 2015 Mortality and Causes of Death Collaborators. (2016). Global, regional, and national life expectancy, all-cause mortality, and cause-specific mortality for 249 causes of death, 1980–2015: a systematic analysis for the Global Burden of Disease Study 2015. Lancet 388(10053): 1459–1544.
Ghayur, M.N., Gilani, A.H., Afridi, M.B., and Houghton, P.J. (2005). Cardiovascular effects of ginger aqueous extract and its phenolic constituents are mediated through multiple pathways. Vascul. Pharmacol. 43(4): 234–241.
Goswami, D., Kumar, M., Ghosh, S.K., and Das, A. (2020). Natural product compounds in alpinia officinarum and ginger are potent SARS-CoV-2 papain-like protease inhibitors. Chemrix .
Grzanna, R., Lindmark, L., and Frondoza, C.G. (2005). Ginger – an herbal medicinal product with broad anti-inflammatory actions. J. Med. Food 8(2): 125–132.
Habib, S.H.M., Makpol, S., Hamid, N.A.A., Das, S., Ngah, W.Z.W., and Yusof, Y.A.M. (2008). Ginger extract (Zingiber officinale) has anti-cancer and anti-inflammatory effects on ethionine-induced hepatoma rats. Clinics 63(6): 807–813.
Han, X., Liu, P., Zheng, B., Zhang, M., Zhang, Y., Xue, Y., Liu, C., Chu, X., Wang, X., Sun, S., and Chu, L. (2022). 6-Gingerol exerts a protective effect against hypoxic injury through the p38/Nrf2/HO-1 and p38/NF-kappaB pathway in H9c2 cells. J. Nutr. Biochem. 104: 108975.
Haridas, M., Sasidhar, V., Nath, P., Abhithaj, J., Sabu, A., and Rammanohar, P. (2021). Compounds of Citrus medica and Zingiber officinale for COVID-19 inhibition: in silico evidence for cues from Ayurveda. Futur J Pharm Sci 7: 13.
Hattori, H., Mori, T., Shibata, T., Kita, M., and Mitsunaga, T. (2021). 6-Paradol Acts as a Potential Anti-obesity Vanilloid from Grains of Paradise. Mol. Nutr. Food Res. 65(16): 2100185.
Ho, Y.C., Lee, S.S., Yang, M.L., Huang-Liu, R., Lee, C.Y., Li, Y.C., and Kuan, Y.H. (2017). Zerumbone reduced the inflammatory response of acute lung injury in endotoxin-treated mice via Akt-NFκB pathway. Chem. Biol. Interact. 271: 9–14.
Hornung, B., Amtmann, E., and Sauer, G. (1994). Lauric acid inhibits the maturation of vesicular stomatitis virus. J. Gen. Virol. 75(2): 353–361.
Huang, F.Y., Deng, T., Meng, L.X., and Ma, X.L. (2019). Dietary ginger as a traditional therapy for blood sugar control in patients with type 2 diabetes mellitus: A systematic review and meta-analysis. Medicine 98(13): e15054.
Jafarnejad, S., Keshavarz, S.A., Mahbubi, S., Saremi, S., Arab, A., Abbasi, S., and Djafarian, K. (2017). Effect of ginger (Zingiber officinale) on blood glucose and lipid concentrations in diabetic and hyperlipidemic subjects: a meta-analysis of randomized controlled trials. J. Funct. Foods 29: 127–134.
Jafarzadeh, A., and Nemati, M. (2018). Therapeutic potentials of ginger for treatment of Multiple sclerosis: A review with emphasis on its immunomodulatory, anti-inflammatory and anti-oxidative properties. J. Neuroimmunol. 324: 54–75.
Jafarzadeh, A., Jafarzadeh, S., and Nemati, M. (2021). Therapeutic potential of ginger against COVID-19: Is there enough evidence? J. Tradit. Chin. Med. Sci. 8(4): 267–279.
Jafarzadeh, A., Mohammadi-Kordkhayli, M., Ahangar-Parvin, R., Azizi, V., Khoramdel-Azad, H., Shamsizadeh, A., Ayoobi, A., Nemati, M., Hassan, Z.M., Moazeni, S.M., and Khaksari, M. (2014). Ginger extracts influence the expression of IL-27 and IL-33 in the central nervous system in experimental autoimmune encephalomyelitis and ameliorates the clinical symptoms of disease. J. Neuroimmunol. 276(1-2): 80–88.
Jahan, R., Paul, A.K., Bondhon, T.A., Hasan, A., Jannat, K., Mahboob, T., Nissapatorn, V., Pereira, M.L., Wiart, C., Wilairatana, P., and Rahmatullah, M. (2021). Zingiber officinale: Ayurvedic uses of the plant and in silico binding studies of selected phytochemicals with Mpro of SARS-CoV-2. Nat. Prod. Commun. 16(10): 1934578X211031766.
Jeon, M., Han, J., Nam, S.J., Lee, J.E., and Kim, S. (2016). Elevated IL-1β expression induces invasiveness of triple negative breast cancer cells and is suppressed by zerumbone. Chem. Biol. Interact. 258: 126–133.
Jiang, X., Wang, J., Chen, P., He, Z., Xu, J., Chen, Y., Liu, X., and Jiang, J. (2021). [6]-Paradol suppresses proliferation and metastases of pancreatic cancer by decreasing EGFR and inactivating PI3K/AKT signaling. Cancer Cell Int. 21(1): 420.
Jiang, Y., Turgeon, D.K., Wright, B.D., Sidahmed, E., Ruffin, M.T., Brenner, D.E., Sen, A., and Zick, S.M. (2013). Effect of ginger root on cyclooxygenase-1 and 15-hydroxyprostaglandin dehydrogenase expression in colonic mucosa of humans at normal and increased risk for colorectal cancer. Eur. J. Cancer Prev. 22(5): 455–460.
Jolad, S.D., Lantz, R.C., Chen, G.J., Bates, R.B., and Timmermann, B.N. (2005). Commercially processed dry ginger (Zingiber officinale): composition and effects on LPS-stimulated PGE2 production. Phytochemistry 66(13): 1614–1635.
Karna, P., Chagani, S., Gundala, S.R., Rida, P.C., Asif, G., Sharma, V., Gupta, M.V., and Aneja, R. (2012). Benefits of whole ginger extract in prostate cancer. Br. J. Nutr. 107(4): 473–484.
Katiyar, S.K., Agarwal, R., and Mukhtar, H. (1996). Inhibition of tumor promotion in SENCAR mouse skin by ethanol extract of Zingiber officinale rhizome. Cancer Res. 56(5): 1023–1030.
Kaushik, S., Jangra, G., Kundu, V., Yadav, J.P., and Kaushik, S. (2020). Anti-viral activity of Zingiber officinale (Ginger) ingredients against the Chikungunya virus. Virusdisease 31(3): 270–276.
Khan, S., Pandotra, P., Qazi, A.K., Lone, S.A., Muzafar, M., Gupta, A.P., and Gupta, S. (2016). Medicinal and nutritional qualities of zingiber officinale. Fruits, vegetables, and herbs. Academic Press, pp. 525–550.
Kim, A., Gwon, M.H., Lee, W., Moon, H.R., and Yun, J.M. (2022). Zerumbone suppresses high glucose and LPS-induced inflammation in THP-1-derived macrophages by inhibiting the NF-κB/TLR signaling pathway. Nutr. Res. 100: 58–69.
Kim, E.C., Min, J.K., Kim, T.Y., Lee, S.J., Yang, H.O., Han, S., Kim, Y.M., and Kwon, Y.G. (2005a). [6]-Gingerol, a pungent ingredient of ginger, inhibits angiogenesis in vitro and in vivo. Biochem. Biophys. Res. Commun. 335(2): 300–308.
Kim, S.O., Kundu, J.K., Shin, Y.K., Park, J.H., Cho, M.H., Kim, T.Y., and Surh, Y.J. (2005b). [6]-Gingerol inhibits COX-2 expression by blocking the activation of p38 MAP kinase and NF-κB in phorbol ester-stimulated mouse skin. Oncogene 24(15): 2558–2567.
Kiyama, R. (2020). Nutritional implications of ginger: chemistry, biological activities and signaling pathways. J. Nutr. Biochem. 86: 108486.
Köngül Şafak, E., and Şeker Karatoprak, G. (2021). Ginger (Gingerols and 6-Shogaol) Against Cancer. Nutraceuticals and Cancer Signaling. Springer, Cham, pp. 291–321.
Kubra, I.R., and Rao, L.J.M. (2012). An impression on current developments in the technology, chemistry, and biological activities of ginger (Zingiber officinale Roscoe). Crit. Rev. Food Sci. Nutr. 52(8): 651–688.
Kukula-Koch, W., Koch, W., Czernicka, L., Glowniak, K., Asakawa, Y., Umeyama, A., Marzec, Z., and Kuzuhara, T. (2018). MAO-A Inhibitory Potential of Terpene Constituents from Ginger Rhizomes–A Bioactivity Guided Fractionation. Molecules 23(6): 1301.
Kumara, M., Shylajab, M.R., Nazeemc, P.A., and Babu, T. (2017). 6-Gingerol is the most potent anticancerous compound in ginger (Zingiber officinale Rosc.). J. Dev. Drugs 6(1): 1–6.
Lee, H.S., Seo, E.Y., Kang, N.E., and Kim, W.K. (2008). [6]-Gingerol inhibits metastasis of MDA-MB-231 human breast cancer cells. J. Nutr. Biochem. 19(5): 313–319.
Lee, W., Hwang, M.H., Lee, Y., and Bae, J.S. (2018). Protective effects of zingerone on lipopolysaccharide-induced hepatic failure through the modulation of inflammatory pathways. Chem. Biol. Interact. 281: 106–110.
Li, G., and De Clercq, E. (2020). Therapeutic options for the 2019 novel coronavirus (2019-nCoV). Nat. Rev. Drug Discov. 19(3): 149–150.
Li, J., Wang, L., Sun, Y., Wang, Z., Qian, Y., Duraisamy, V., and Antary, T.M.A. (2022). Zerumbone-induced reactive oxygen species-mediated oxidative stress re-sensitizes breast cancer cells to paclitaxel. Biotechnol. Appl. Biochem.
Li, X., Tian, R., Liu, L., Wang, L., He, D., Cao, K., Ma, J.K., and Huang, C. (2020). Andrographolide enhanced radiosensitivity by downregulating glycolysis via the inhibition of the PI3K-Akt-mTOR signaling pathway in HCT116 colorectal cancer cells. J. Int. Med. Res. 48(8): 300060520946169.
Lv, X., Xu, T., Wu, Q., Zhou, Y., Huang, G., Xu, Y., and Zhong, G. (2018). 6-Gingerol activates PI3K/Akt and inhibits apoptosis to attenuate myocardial ischemia/reperfusion injury. Evid. Based Complement. Alternat. Med. 2018: 9024034.
Ma, R.H., Ni, Z.J., Zhu, Y.Y., Thakur, K., Zhang, F., Zhang, Y.Y., Hu, F., Zhang, J.G., and Wei, Z.J. (2021). A recent update on the multifaceted health benefits associated with ginger and its bioactive components. Food Funct. 12(2): 519–542.
Ma, S.Q., Guo, Z., Liu, F.Y., Hasan, S.G., Yang, D., Tang, N., An, P., Wang, M.Y., Wu, H.M., Yang, Z., Fan, D., and Tang, Q.Z. (2021). 6-Gingerol protects against cardiac remodeling by inhibiting the p38 mitogen-activated protein kinase pathway. Acta Pharmacol Sin. 42(10): 1575–1586.
Mahomoodally, M.F., Aumeeruddy, M.Z., Rengasamy, K.R.R., Roshan, S., Hammad, S., Pandohee, J., Hu, X., and Zengin, G. (2021). Ginger and its active compounds in cancer therapy: From folk uses to nano-therapeutic applications. Semin. Cancer Biol. 69: 140–149.
Mase, N., Kitagawa, N., and Takabe, K. (2010). Protection-, Salt-, and Metal-Free Syntheses of [n]-Shogaols by Use of Dimethylammonium Dimethyl Carbamate (DIMCARB) without Protecting Groups. Synlett 2010(01): 93–96.
Mashhadi, N.S., Ghiasvand, R., Askari, G., Hariri, M., Darvishi, L., and Mofid, M.R. (2013). Anti-oxidative and anti-inflammatory effects of ginger in health and physical activity: review of current evidence. Int. J. Prev. Med. 4(Suppl 1): S36.
Mazidi, M., Gao, H.K., Rezaie, P., and Ferns, G.A. (2016). The effect of ginger supplementation on serum C-reactive protein, lipid profile and glycaemia: a systematic review and meta-analysis. Food Nutr. Res. 60(1): 32613.
Mbaveng, A.T., and Kuete, V. (2017). Zingiber officinale. Medicinal Spices and Vegetables from Africa. Academic Press, pp. 627–639.
Mimura, J., and Itoh, K. (2015). Role of Nrf2 in the pathogenesis of atherosclerosis. Free Radic. Biol. Med. 88: 221–232.
Mohammed, H.M. (2022). Zingerone ameliorates non-alcoholic fatty liver disease in rats by activating AMPK. J. Food Biochem. e14149.
Mohd Zaid, N.A., Sekar, M., Bonam, S.R., Gan, S.H., Lum, P.T., Begum, M.Y., Mat Rani, N., Vaijanathappa, J., Wu, Y.S., Subramaniyan, V., Fuloria, N.K., and Fuloria, S. (2022). Promising Natural Products in New Drug Design, Development, and Therapy for Skin Disorders: An Overview of Scientific Evidence and Understanding Their Mechanism of Action. Drug Des. Devel. Ther. 16: 23–66.
Moon, M., Kim, H.G., Choi, J.G., Oh, H., Lee, P.K., Ha, S.K., Kim, S.Y., Park, Y., Huh, Y., and Oh, M.S. (2014). 6-Shogaol, an active constituent of ginger, attenuates neuroinflammation and cognitive deficits in animal models of dementia. Biochem. Biophys. Res. Commun. 449(1): 8–13.
Morvaridzadeh, M., Fazelian, S., Agah, S., Khazdouz, M., Rahimlou, M., Agh, F., Potter, E., Heshmati, S., and Heshmati, J. (2020). Effect of ginger (Zingiber officinale) on inflammatory markers: A systematic review and meta-analysis of randomized controlled trials. Cytokine 135: 155224.
Mukhtar, M., Arshad, M., Ahmad, M., Pomerantz, R.J., Wigdahl, B., and Parveen, Z. (2008). Antiviral potentials of medicinal plants. Virus Res. 131(2): 111–120.
Mustafa, T., and Srivastava, K.C. (1990). Ginger (Zingiber officinale) in migraine headache. J. Ethnopharmacol. 29(3): 267–273.
Narayana, D.B.A., Brindavanam, N.B., Dobriyal, R.M., and Katuyar, K.C. (2000). Indian Spices: An overview with special reference to neutraceuticals. J. Medicinal Aromat. Plants Sci. 22(1B): 236–246.
Nicoll, R., and Henein, M.Y. (2009). Ginger (Zingiber officinale Roscoe): a hot remedy for cardiovascular disease? Int. J. Cardiol. 131(3): 408–409.
Okamoto, M., Irii, H., Tahara, Y., Ishii, H., Hirao, A., Udagawa, H., Hiramoto, M., Yasuda, K., Takanishi, A., Shibata, S., and Shimizu, I. (2011). Synthesis of a new [6]-gingerol analogue and its protective effect with respect to the development of metabolic syndrome in mice fed a high-fat diet. J. Med. Chem. 54(18): 6295–6304.
Ooi, B.K., Chan, K.G., Goh, B.H., and Yap, W.H. (2018). The role of natural products in targeting cardiovascular diseases via Nrf2 pathway: novel molecular mechanisms and therapeutic approaches. Front. Pharmacol. 9: 1308.
Oso, B.J., Adeoye, A.O., and Olaoye, I.F. (2022). Pharmacoinformatics and hypothetical studies on allicin, curcumin, and gingerol as potential candidates against COVID-19-associated proteases. J. Biomol. Struct. Dyn. 40(1): 389–400.
Park, K.K., Chun, K.S., Lee, J.M., Lee, S.S., and Surh, Y.J. (1998). Inhibitory effects of [6]-gingerol, a major pungent principle of ginger, on phorbol ester-induced inflammation, epidermal ornithine decarboxylase activity and skin tumor promotion in ICR mice. Cancer lett. 129(2): 139–144.
Park, Y.J., Wen, J., Bang, S., Park, S.W., and Song, S.Y. (2006). [6]-Gingerol induces cell cycle arrest and cell death of mutant p53-expressing pancreatic cancer cells. Yonsei Med. J. 47(5): 688–697.
Pertz, H.H., Lehmann, J., Roth-Ehrang, R., and Elz, S. (2011). Effects of ginger constituents on the gastrointestinal tract: role of cholinergic M3 and serotonergic 5-HT3 and 5-HT4 receptors. Planta Med. 77(10): 973–978.
Phillips, S., Ruggier, R., and Hutchinson, S.E. (1993). Zingiber officinale (ginger)–an antiemetic for day case surgery. Anaesthesia 48(8): 715–717.
Pickersgill, B (2005). In: Prance, G, and Nesbitt, M (Ed.). The Cultural History of Plants. Routledge, pp. 163–164.
Pourmasoumi, M., Hadi, A., Rafie, N., Najafgholizadeh, A., Mohammadi, H., and Rouhani, M.H. (2018). The effect of ginger supplementation on lipid profile: A systematic review and meta-analysis of clinical trials. Phytomedicine 43: 28–36.
Rahman, H.S., Abdullah, R., Abdul, A.B., Allaudin, Z.N., Namvar, F., Othman, H.H., Yeap, S.K., and How, C.W. (2013). Zerumbone, a Natural Dietary Sesquiterpene from Zingiber Zerumbet for Leukaemia Therapy In Vitro. Open Conf. Proc. J. 4(1): 67.
Ren, Q., Zhao, S., and Ren, C. (2019). 6-Gingerol protects cardiocytes H9c2 against hypoxia-induced injury by suppressing BNIP3 expression. Artif. Cells Nanomed. Biotechnol. 47(1): 2016–2023.
Rondanelli, M., Fossari, F., Vecchio, V., Gasparri, C., Peroni, G., Spadaccini, D., Riva, A., Petrangolini, G., Iannello, G., Nichetti, M., Infantino, V., and Perna, S. (2020). Clinical trials on pain lowering effect of ginger: A narrative review. Phytother. Res. 34(11): 2843–2856.
Roudsari, N.M., Lashgari, N.A., Momtaz, S., Roufogalis, B., Abdolghaffari, A.H., and Sahebkar, A. (2021). Ginger: A complementary approach for management of cardiovascular diseases. BioFactors 47(6): 933–951.
Sahoo, M., Jena, L., Rath, S.N., and Kumar, S. (2016). Identification of suitable natural inhibitor against influenza A (H1N1) neuraminidase protein by molecular docking. Genomics Inform. 14(3): 96.
San Chang, J., Wang, K.C., Yeh, C.F., Shieh, D.E., and Chiang, L.C. (2013). Fresh ginger (Zingiber officinale) has anti-viral activity against human respiratory syncytial virus in human respiratory tract cell lines. J. Ethnopharmacol. 145(1): 146–151.
Sang, S., Hong, J., Wu, H., Liu, J., Yang, C.S., Pan, M.H., Badmaev, V., and Ho, C.T. (2009). Increased growth inhibitory effects on human cancer cells and anti-inflammatory potency of shogaols from Zingiber officinale relative to gingerols. J. Agric. Food Chem. 57(22): 10645–10650.
Schepici, G., Contestabile, V., Valeri, A., and Mazzon, E. (2021). Ginger, a Possible Candidate for the Treatment of Dementias? Molecules 26(18): 5700.
Sharma, B.K., Klinzing, D.C., and Ramos, J.D. (2015). Zingiber officinale roscoe aqueous extract modulates matrixmetalloproteinases and tissue inhibitors of metalloproteinases expressions in dengue virus-infected cells: Implications for prevention of vascular permeability. Trop. J. Pharm. Res. 14(8): 1371–1381.
Shim, S., and Kwon, J. (2012). Effects of [6]-shogaol on cholinergic signaling in HT22 cells following neuronal damage induced by hydrogen peroxide. Food Chem. Toxicol. 50(5): 1454–1459.
Shukla, Y., and Singh, M. (2007). Cancer preventive properties of ginger: a brief review. Food Chem. Toxicol. 45(5): 683–690.
Srinivasan, K. (2017). Ginger rhizomes (Zingiber officinale): A spice with multiple health beneficial potentials. PharmaNutrition 5(1): 18–28.
Srinivasan, K., Adhya, P., and Sharma, S.S. (2019). Nutraceutical Potential of Ginger. Nutraceuticals in Veterinary Medicine. Springer, Cham, pp. 51–70.
Su, M., Wang, X., Cao, G., Sun, L., Ho, R.J.Y., Han, Y., Hong, Y., and Wu, D. (2022). Prediction of the potential mechanism of compound gingerol against liver cancer based on network pharmacology and experimental verification. J. Pharm. Pharmacol. 74(6): 869–886.
Sun, Y., Ren, J., and Wang, F. (2021). [6]-Gingerol impedes 7, 12-dimethylbenz (a) anthracene-induced inflammation and cell proliferation-associated hamster buccal pouch carcinogenesis through modulating Nrf2 signaling events. J. Biochem. Mol. Toxicol. 35(4): e22689.
Taha, M.M.E., Abdul, A.B., Abdullah, R., Ibrahim, T.A.T., Abdelwahab, S.I., and Mohan, S. (2010). Potential chemoprevention of diethylnitrosamine-initiated and 2-acetylaminofluorene-promoted hepatocarcinogenesis by zerumbone from the rhizomes of the subtropical ginger (Zingiber zerumbet). Chem. Biol. Interact. 186(3): 295–305.
Thomson, M., Al-Qattan, K.K., Al-Sawan, S.M., Alnaqeeb, M.A., Khan, I., and Ali, M. (2002). The use of ginger (Zingiber officinale Rosc.) as a potential anti-inflammatory and antithrombotic agent. Prostaglandins Leukot. Essent. Fatty Acids 67(6): 475–478.
Tianthong, W., and Phupong, V. (2018). A randomized, double-blind, placebo-controlled trial on the efficacy of ginger in the prevention of abdominal distention in post cesarean section patients. Sci. Rep. 8(1): 1–5.
Togar, B.A.S.A.K., Türkez, H., Stefano, A.D., Tatar, A., and Cetin, D.A.M.L.A. (2015a). Zingiberene attenuates hydrogen peroxide-induced toxicity in neuronal cells. Hum. Exp. Toxicol. 34(2): 135–144.
Togar, B., Turkez, H., Tatar, A., Hacimuftuoglu, A., and Geyikoglu, F. (2015b). Cytotoxicity and genotoxicity of zingiberene on different neuron cell lines in vitro. Cytotechnology 67(6): 939–946.
Tzeng, T.F., Lu, H.J., Liou, S.S., Chang, C.J., and Liu, I.M. (2014). Lipid-lowering effects of zerumbone, a natural cyclic sesquiterpene of Zingiber zerumbet Smith, in high-fat diet-induced hyperlipidemic hamsters. Food Chem. Toxicol. 69: 132–139.
Vasala, P.A. (2012). Ginger. Handbook of herbs and spices. Woodhead Publishing, pp. 319–335.
Vijayan, A.K., Gudade, B.A., Gautam, A., Deka, T.N., Bora, S.S., Dhanapal, K., and Remashree, A.B. (2020). Cultivation of ginger in Sikkim under an organic system. Ginger Cultivation and Its Antimicrobial and Pharmacological Potentials. IntechOpen.
Wang, J., Prinz, R.A., Liu, X., and Xu, X. (2020a). In vitro and in vivo antiviral activity of gingerenone A on influenza a virus is mediated by targeting Janus kinase 2. Viruses 12(10): 1141.
Wang, L., Bo, X., Yi, X., Xiao, X., Zheng, Q., Ma, L., and Li, B. (2020b). Exosome-transferred LINC01559 promotes the progression of gastric cancer via PI3K/AKT signaling pathway. Cell Death. Dis. 11(9): 1–14.
Wang, S., Zhang, C., Yang, G., and Yang, Y. (2014). Biological properties of 6-gingerol: a brief review. Nat. Prod. Commun. 9(7): 1934578X1400900736.
Wei, C.K., Tsai, Y.H., Korinek, M., Hung, P.H., El-Shazly, M., Cheng, Y.B., Wu, Y.C., Hsieh, T.J., and Chang, F.R. (2017). 6-Paradol and 6-Shogaol, the Pungent Compounds of Ginger, Promote Glucose Utilization in Adipocytes and Myotubes, and 6-Paradol Reduces Blood Glucose in High-Fat Diet-Fed Mice. Int. J. Mol. Sci. 18(1): 168.
Wheeler, D.L. (2003). Egypt: Building an information society for international development. Rev. Afr. Polit. Econ. 30(98): 627–642.
Xu, S., Zhang, H., Liu, T., Wang, Z., Yang, W., Hou, T., Wang, X., He, D., and Zheng, P. (2021). 6-Gingerol suppresses tumor cell metastasis by increasing YAPser127 phosphorylation in renal cell carcinoma. J. Biochem. Mol. Toxicol. 35(1): e22609.
Xu, T., Qin, G., Jiang, W., Zhao, Y., Xu, Y., and Lv, X. (2018). 6-Gingerol protects heart by suppressing myocardial ischemia/reperfusion induced inflammation via the PI3K/Akt-dependent mechanism in rats. Evid. Based Complement. Alternat. Med. 2018: 6209679.
Xue, Y., Zhang, M., Liu, M., Liu, Y., Li, L., Han, X., Sun, Z., and Chu, L. (2021b). 8-Gingerol Ameliorates Myocardial Fibrosis by Attenuating Reactive Oxygen Species, Apoptosis, and Autophagy via the PI3K/Akt/mTOR Signaling Pathway. Front. Pharmacol. 12: 711701.
Xue, Y., Zhang, M., Zheng, B., Zhang, Y., Chu, X., Liu, Y., Li, Z., Han, X., and Chu, L. (2021a). [8]-Gingerol exerts anti-myocardial ischemic effects in rats via modulation of the MAPK signaling pathway and L-type Ca(2+) channels. Pharmacol. Res. Perspect. 9(5): e00852.
Yasmin, A.R., Chia, S.L., Looi, Q.H., Omar, A.R., Noordin, M.M., and Ideris, A. (2020). Herbal extracts as antiviral agents. Feed additives. Academic Press, pp. 115–132.
Yusof, Y.A.M. (2016). Gingerol and its role in chronic diseases. Drug discovery from mother nature. pp. 177–207.
Zhang, Q., Liu, J., Duan, H., Li, R., Peng, W., and Wu, C. (2021a). Activation of Nrf2/HO-1 signaling: An important molecular mechanism of herbal medicine in the treatment of atherosclerosis via the protection of vascular endothelial cells from oxidative stress. J. Adv. Res. 34: 43–63.
Zhang, M.M., Wang, D., Lu, F., Zhao, R., Ye, X., He, L., Ai, L., and Wu, C.J. (2021b). Identification of the active substances and mechanisms of ginger for the treatment of colon cancer based on network pharmacology and molecular docking. BioData Min. 14(1): 1.
Zick, S.M., Turgeon, D.K., Vareed, S.K., Ruffin, M.T., Litzinger, A.J., Wright, B.D., Alrawi, S., Normolle, D.P., Djuric, Z., and Brenner, D.E. (2011). Phase II study of the effects of ginger root extract on eicosanoids in colon mucosa in people at normal risk for colorectal cancer. Cancer. Prev. Res. (Phila) 4(11): 1929–1937.
Zick, S.M., Turgeon, D.K., Ren, J., Ruffin, M.T., Wright, B.D., Sen, A., Djuric, Z., and Brenner, D.E. (2015). Pilot clinical study of the effects of ginger root extract on eicosanoids in colonic mucosa of subjects at increased risk for colorectal cancer. Mol. Carcinog. 54(9): 908–915.