Next Article in Journal
Genes Encoding Structurally Conserved Serpins in the Wheat Genome: Identification and Expression Profiles during Plant Development and Abiotic and Biotic Stress
Next Article in Special Issue
Genistein and Procyanidin B2 Reduce Carcinogen-Induced Reactive Oxygen Species and DNA Damage through the Activation of Nrf2/ARE Cell Signaling in Bronchial Epithelial Cells In Vitro
Previous Article in Journal
Exploration of Novel Scaffolds Targeting Cytochrome b of Pyricularia oryzae
Previous Article in Special Issue
Identification of Novel Artemisinin Hybrids Induce Apoptosis and Ferroptosis in MCF-7 Cells
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Health Benefits of Coffee Consumption for Cancer and Other Diseases and Mechanisms of Action

1
Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA
2
Master of Biotechnology Program, Texas A&M University, College Station, TX 77843, USA
3
Program in Integrative Nutrition and Complex Diseases, Department of Nutrition, Texas A&M University, College Station, TX 77843, USA
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(3), 2706; https://doi.org/10.3390/ijms24032706
Submission received: 19 December 2022 / Revised: 24 January 2023 / Accepted: 26 January 2023 / Published: 31 January 2023
(This article belongs to the Special Issue Cancer Targeted Small Molecules)

Abstract

:
Coffee is one of the most widely consumed beverages worldwide, and epidemiology studies associate higher coffee consumption with decreased rates of mortality and decreased rates of neurological and metabolic diseases, including Parkinson’s disease and type 2 diabetes. In addition, there is also evidence that higher coffee consumption is associated with lower rates of colon and rectal cancer, as well as breast, endometrial, and other cancers, although for some of these cancers, the results are conflicting. These studies reflect the chemopreventive effects of coffee; there is also evidence that coffee consumption may be therapeutic for some forms of breast and colon cancer, and this needs to be further investigated. The mechanisms associated with the chemopreventive or chemotherapeutic effects of over 1000 individual compounds in roasted coffee are complex and may vary with different diseases. Some of these mechanisms may be related to nuclear factor erythroid 2 (Nrf2)-regulated pathways that target oxidative stress or pathways that induce reactive oxygen species to kill diseased cells (primarily therapeutic). There is evidence for the involvement of receptors which include the aryl hydrocarbon receptor (AhR) and orphan nuclear receptor 4A1 (NR4A1), as well as contributions from epigenetic pathways and the gut microbiome. Further elucidation of the mechanisms will facilitate the potential future clinical applications of coffee extracts for treating cancer and other inflammatory diseases.

1. Introduction

Coffee is among the most widely consumed beverages in the world, and it is estimated that over two billion cups of coffee are consumed daily [1,2]. Coffee intake is highly variable with respect to different countries, ages, and sex, and there appears to be a continuing increase in consumption which parallels, in part, the increasing number of specialty coffee shops in many countries. Coffee intake is often associated with the stimulant caffeine, which is a major component of coffee, and the average caffeine intake in the United States is 135 milligrams per day, which is equivalent to about 1.5 cups per day. Many individuals consume up to 6 cups of coffee per day and much higher amounts of caffeine. Although roasted coffee beans and brewed coffee contain high levels of caffeine, there are several hundred individual phytochemical-derived compounds in coffee, and these include chlorogenic acid/lignans, alkaloids, polyphenolics, terpenoids, melanoidins, vitamins, and metals [3].
Figure 1 illustrates some examples of the compounds identified in coffee, and these include the flavonoid quercetin, chlorogenic acid, caffeine, the alkaloid norharman (β-carboline), and the terpenoid cafestrol. The health impacts of coffee consumption have been extensively investigated and are associated with lower all-cause mortality, diabetes mellitus, dementia, Parkinson’s disease, cardiovascular disease, and many types of cancer [2,3,4,5,6]. The effects of coffee on mortality and other diseases have been extensively investigated in many countries and in groups of individuals that are both “normal” or have specific health problems. The results of recent and past studies clearly show the overall health benefits of higher coffee consumption compared to lower consumption; however, there are also many studies that do not correlate and, in some cases, report conflicting results. The reasons for these differences in some cases may be the failure to examine the effects of sex dependency; however, many other potential unknown confounders may be involved and these need to be further investigated.

2. Coffee and Health Benefits: Non-Cancer

2.1. Mortality

A number of recent studies (2020–present) have demonstrated that the higher consumption of coffee is associated with decreased mortality in both men and women. For example, in the UK Biobank study [2], the coffee intake of 395,539 individuals was collected between 2006–2010, and their overall disease-specific mortality was followed through 2020 (a median follow-up of 11.8 years). High levels of coffee intake (≥4 cups/day) were inversely associated with mortality from 30 of 31 diseases, with HRs ranging from 0.61–0.94, and these inverse associations were more predominant in women vs. men [2]. Interestingly the association between a high vs. low consumption of coffee in this population and mortality from various diseases was dependent on multiple variables, which include the sex of the individual, specific diseases, regular vs. decaffeinated coffee, and consumption levels. Examples of male vs. female differences with respect to a high vs. low consumption of coffee were associated with female/male HR values of 0.73/1.0 (digestive disorders), diabetes mellitus (0.74/0.92), and gout (0.71/0.59). In this study, there were “2 distinct clusters of medical conditions affecting mainly the cardiometabolic and gastrointestinal systems” [2]. In contrast, other studies did not observe inverse associations between coffee consumption and decreased mortality from neurogenerative diseases. Other studies in Korea [4], the United States [7,8,9,10], an Asia cohort [11], and an adult Mediterranean population [12] also reported that the higher consumption of coffee is associated with decreased mortality. Thus, the overall effect of coffee on mortality is comparable to previous and ongoing studies on other groups, including Seventh Day Adventists and the consumption of a Mediterranean diet, where high intakes of vegetables are also associated with decreased mortality [13,14].

2.2. Cardiovascular Diseases (CVDs)

In the Biobank study noted above [2], there was a strong association between decreased mortality from cardiometabolic diseases and the higher consumption of coffee [6], and this was also observed in a recent study on the Biobank population [15]. In contrast, a meta-analysis of 32 prospective cohorts reported that various studies showed that higher coffee consumption was associated with increased, decreased, or no effect on CVD [16]. In the UK Biobank studies, the higher consumption of coffee was associated with higher total cholesterol and LDL-cholesterol levels, and this was highest in the expresso coffee drinkers [17]. A meta-analysis of studies showed that higher coffee consumption was associated with “an increased risk of CHD in men and a potentially decreased risk in women” [16], and an increased risk was also observed in another analysis of the UK Biobank population [18]. Another recent report indicated that, among individuals with grade 2–3 hypertension, the HR mortality values were 0.98 (<1 cup/day), 0.74 (1 cup/day), and 2.05 (≥2 cups/day) compared to noncoffee drinkers [19]. In contrast, it was also reported that the medium–high consumption of coffee (3–5 cups/day) had a beneficial or neutral impact on hypertension and blood pressure [20]. Thus, the relationship between the higher consumption of coffee and mortality from cardiovascular diseases is somewhat variable between studies, and the factors that modulate the impact of the effects of coffee on this disease need to be determined.

2.3. Neurological Diseases

The linkage between higher coffee consumption and decreased mortality for neurologic diseases such as dementia, Parkinson’s disease, and Alzheimer’s disease have been extensively investigated, and the results has been variable. It was recently confirmed that high coffee consumption is associated with decreased risks of neurological disorders, including dementia, stroke, and Parkinson’s disease [16,17,19,21,22,23]. These results were also observed in the UK Biobank prospective study [24]. As noted above, another report using the UK Biobank did not observe any correlation between coffee consumption and decreased mortality from neurodegenerative diseases [2]. A negative finding was the association between the early age of onset of Huntington’s disease with increased coffee consumption, and this outlier might be due to the strong genetic origins of this debilitating autosomal dominant disease [25]. However, there is evidence for the improvement of specific neurologic conditions with coffee consumption. For example, the moderate consumption of mocha coffee in an elderly population was associated with higher cognitive and mood status [26]; coffee consumption enhanced the age at onset of Parkinson’s disease in Ashkenazi Jewish patients [27]; the results of a meta-analysis showed that coffee consumption reduced the risk of overall stroke, hemorrhagic, and ischemic stroke [28]. Thus coffee–neurological disease interactions are variable in terms of mortality; however, there is evidence of protection from nonlethal neurological diseases that also needs to be considered and more fully investigated.

2.4. Metabolic Diseases including Diabetes

The higher consumption of coffee is also associated with a decreased risk of metabolic diseases and type 2 diabetes [29,30,31,32,33,34]. An analysis of plasma biomarkers in nondrinkers vs. individuals consuming ≥4 cups/day showed that in the latter group, the changes in biomarkers were consistent with favorable outcomes [31]. For example, higher concentrations of sex hormone binding globulin (SHBG) (5.0%), total testosterone (7.3 and 5.3% in women and men, respectively), and total (9.3%) and high molecular weight (17.2%) adiponectin were increased in the coffee drinkers. In contrast, the group consuming high amounts of coffee exhibited lower levels of inflammatory markers, such as interleukin-6 (−8.1%), soluble tumor necrosis factor receptor (−5.8%), and C-reactive protein (−16.6%) [31]. These results were obtained from two large prospective studies: the Nurses Health Study and the Health Professionals Follow-up Study in the United States [31]. There is also evidence that coffee consumption interacts with other factors that modify the association between coffee and metabolic diseases. For example, in patients with rheumatoid arthritis, coffee intake was associated with lower metabolic syndrome scores [33]; increased caffeinated and noncaffeinated coffee intake protected against nonalcoholic fatty liver disease (NAFLD) severity in individuals with type 2 diabetes [35], and similar results were observed in a normal population [36]. The protective effects of coffee on metabolic disorders were also observed in individuals with a history of gestational diabetes [37], diabetic retinopathy [38], and hepatitis B viral infection [39]. These results are complemented by several recent studies showing that high coffee consumption is also associated with protection from inflammatory bowel disease and gut recovery in gynecological patients from surgery [40,41,42].

2.5. Sex-Dependent Effects of Coffee Consumption

The sex-dependent development of some diseases has been described and is thought to be due to multiple factors; there is some evidence for differences with respect to the effects of coffee on males and females [43]. The higher consumption of coffee decreased the prevalence of metabolic syndrome in an adult Taiwanese population, and the protective effects were more pronounced in women [44]. In a UK Biobank study [2], the overall effects of high coffee consumption among subgroups of diseases were higher for women than men and this was particularly evident for functional digestive disorders and diabetes mellitus, whereas men were more protected from gout [2]. A meta-analysis of the risk of coronary heart disease also showed that coffee consumption was associated with lower risks for coronary heart disease in women than in men [16]. A recent review summarized the sex-dependent differences in several neurological and psychiatric disorders and used estimated caffeine consumption data to compare the association between these disorders [43]. Caffeine is more effective in women than in men for improving depression and Parkinson’s disease, and caffeine enhances anxiety in men more than in women. In this brief introduction to the association between coffee and a decreased risk of some diseases, we have primarily used references published from 2020–the present, and a similar selection of more recent articles will be used to review the association of high coffee consumption to decreased risks of cancer.

3. Coffee and Cancer

Coffee consumption and a decreased risk of cancer have been extensively investigated, and multiple studies confirm the inverse association between high coffee consumption and a decreased cancer risk [6,7,11,12,45,46]. Reports on the effects of coffee consumption on the overall and specific cancer risks are extensive and sometimes contradictory, and this review will primarily focus on the results of recent studies (i.e., 2020–the present) unless data for a particular type of cancer have not been published recently.

3.1. Gastrointestinal Tract

3.1.1. Liver Cancer

A recent comprehensive review summarizes the lack of association between coffee consumption and cancers of the different digestive organs, pointing to the powerful protective effect of coffee against the risk of hepatocellular carcinoma [47]. Two studies using the UK Biobank participants confirmed that the higher consumption of all coffee types (including decaffeinated) decreased the risk of chronic liver disease [48] and liver cancer [49] but no other digestive cancers [50]. Similar results were observed for liver cancer in a Japanese population [49] and meta-analysis of other studies [51].

3.1.2. Colon and Rectal Cancer

Two recent studies have confirmed previous reports in which higher coffee consumption did not decrease the risk of colon cancer [52,53,54], whereas another report indicated that ≥2 cups/day of decaffeinated coffee lowered the risk of colon cancer [55]. Two additional papers reported that higher coffee consumption was associated with decreased risk of colon cancer [56,57]. Moreover, in both studies, the interaction between genetic polymorphisms was observed, including the variant aryl hydrocarbon (AhR) rs2066853 gene [52]. It was also reported that higher coffee consumption was associated with some decreased risks of rectal cancer.

3.1.3. Other Gastrointestinal Cancers

There was no association between high coffee consumption and the risk for pancreatic cancer [58,59] or gastric cancer [54,60,61]. Similar results were observed for overall esophageal cancer [62,63]. However, in a European prospective cohort, there was a decreased risk of esophageal squamous cell carcinoma [63], and a decreased risk was observed for East-Asian but not European participants, whereas an analysis of the UK Biobank data suggested that coffee intake had increased the risk of esophageal cancer [45].

3.2. Genitourinary Cancers

The effects of coffee consumption on genitourinary cancers have not provided definitive associations in epidemiological studies, and research in this area is ongoing.

3.2.1. Prostate and Bladder Cancer

Some studies show that there is no association between high coffee consumption and decreased risk of prostate cancer [64,65,66], while other reports show an inverse relationship [67,68,69,70,71]. A recent study of a large Japanese cohort showed no association between higher coffee consumption and prostate cancer risk [72]; clearly, these studies are inconclusive, and other factors may be involved. For bladder cancer, individual studies and meta-analyses do not show an association between coffee consumption and bladder cancer [73,74].

3.2.2. Renal Cancer

A recent Mendelian randomization study did not observe a decreased risk of renal cancer with increased coffee consumption [75]. These results were in contrast to a meta-analysis of several cohorts [76] and another large study [77], where a 20% reduced risk was observed in patients with coffee intakes of ≥2 cups/day. These effects may be due, in part, to coffee constituents, such as kahweol and cafestrol which inhibit the growth and migration of renal cancer cells [78].

3.2.3. Endocrine Cancers

The endocrine status of an organ modulates the effects of various exogenous substances, including diet and beverages, such as coffee, and this has been observed for breast and endometrial cancers.

3.2.4. Breast Cancer

A recent review concluded that “there is no association between coffee intake and breast cancer risk or a slight protective effect even at the higher dosages” [79]. However, breast cancer is a highly complex disease and is observed in both pre and postmenopausal women, and these factors may modify the effects of coffee. An analysis of the data from the Women’s Health Initiative [80,81] did not show an association between high coffee intake and decreased risk of breast cancer, and similar results were observed with the Cancer Prevention Study-II Nutrition cohort [82]. In contrast, there is also evidence that higher coffee consumption decreases breast cancer risk in postmenopausal and European women [83,84], women expressing the minor allele of the bcl-2 gene haplotype [85], and other bcl-2 polymorphisms.

3.2.5. Endometrial Cancers

A meta-analysis of several studies showed no association between coffee consumption and the risk for endometrial cancer, whereas another large study demonstrated that coffee consumption decreased the risk for endometrial cancer, particularly among women with a body mass of ≥25 kg/m2 [86,87].

3.2.6. Other Cancers

Results from lung cancer studies showed that coffee consumption increased [88,89] or decreased [90] the risk; coffee consumption had no effect [91] or decreased [92,93] the risk for glioma and decreased the risk of head and neck cancers [94]. Maternal consumption of coffee was either not associated with or increased the risk of childhood cancers [95,96]. This is an area that needs to be further investigated since the effects of in utero exposures on the offspring are being studied for many other chemicals, including endocrine-disrupting compounds and phytochemicals.

4. Chemopreventive and Chemotherapeutic Effects of Coffee

The dietary consumption of foods, including coffee, and specific diets, such as the health-promoting Mediterranean diet, contain many of the same classes of phytochemicals and are associated with some of the same health benefits linked to non-cancer and cancer endpoints. These benefits are usually derived from the long-term consumption of specific foods and beverages and are chemopreventive; namely, they decrease the risks of developing a disease. Too few studies are designed to examine their chemotherapeutic activities, which are the effects observed after disease diagnosis. Soldato and coworkers investigated the effects of coffee consumption on breast cancer patient outcomes from years 1–4 after an initial diagnosis. Based on their patterns of coffee consumption, there was no association with the clinical outcomes [97]. In contrast, the postdiagnosis effects of high coffee consumption by women in the Nurses’ Health Study were associated with lower breast cancer-specific mortality [98]. Another report showed that in a cohort of patients being treated for advanced metastatic colon cancer, coffee consumption was associated with a decreased risk of subsequent cancer progression and death [99]. For example, patients “who consumed at least 4 cups of coffee per day have a multivariate HR for OS of 0.64 (95% CI, 0.46–0.87) and for PFS of 0.78 (95% CI, 0.59–1.05)”. Significant associations were noted for both caffeinated and decaffeinated coffee [99]. These results suggest potential therapeutic applications for coffee, which may be of value for treating multiple cancers. Thus, either coffee or coffee extracts should be more extensively evaluated as cancer therapeutics that can be used in combination with ongoing therapies to enhance the overall survival from this deadly disease. This should also be investigated in other cancers.

5. Mechanisms of Coffee-Mediated Anticancer Activities

This review of cancer studies has primarily focused on coffee consumption and its anticancer activities, which can be both chemopreventive (before cancer diagnosis) and chemotherapeutic (after cancer diagnosis). The subset of coffee compounds that are chemopreventive and chemotherapeutic will not necessarily be the same compounds, and for those compounds that are both chemopreventive and therapeutic, their mechanisms of action for these two responses may also differ. Chemopreventive mechanisms are difficult to establish in human and rodent models. However, it is assumed that the compounds that reduce the formation of oxidative stress and other stressors and decrease radical formation and inflammation play a role in disease prevention. These chemopreventive pathways are also associated with the Mediterranean diet [100,101], which is enriched in phytochemicals similar to those observed in coffee. Figure 1 illustrates examples of some of the major classes of phytochemicals in coffee and include caffeine, quercetin, chlorogenic acid, cafestrol and norharman (β-carboline).

Activation of Nrf2 by Coffee

A recent study examined the activities of the phytochemicals in coffee and concluded that their overall effects were not sufficient to account for the required radical scavenging anti-inflammatory activity observed in human and laboratory animal studies. It was proposed that the activation of nuclear factor erythroid 2 (Nrf2) and its protective pathways (Figure 2) play a major role in mediating the beneficial health effects of coffee [102]. For example, coffee/coffee phytochemicals induce or activate Nrf2 in cells under some oxidative stress [102,103,104,105]. Among the important Nrf2-regulated genes associated with antioxidant activity are glutathione peroxidase haem oxygenase-1, glutathione reductases, superoxide dismutase, quinone oxidoreductase, and several thioreductases. Nrf2 exists as a cytosolic dimer with Keap2, and it also interacts with Cullin 3-based ubiquitin ligase and this complex maintains basal cytosolic levels of the Nrf2-Keap heterodimer. A combination of factors, including those that inhibit Keap-Nrf2 interactions or enhance Keap degradation, results in the nuclear uptake of Nrf2, which binds to small musculoaponeurotic fibrosarcoma (sMaF) protein to form the Nrf2-sMaF heterodimer, which then interacts with the cis-acting antioxidant response element (ARE) in target gene promoters to activate gene expression. This results in the activation of antioxidant genes (e.g., glutathione reductase, glutathione peroxidase), multiple redox family genes, (e.g., catalase, haem oxygenase (1)), anti-inflammatory genes (e.g., interleukins, interferons, tumor necrosis factor), drug metabolism enzymes (e.g., epoxide hydrolase, UDP–glucuronyl transferases, CYP1B1) and many other genes/pathways. All of these induced, Nrf2-dependent pathways/genes play diverse roles as cellular protective proteins. In addition, several reports demonstrate that the AhR and its ligands co-operatively enhance Nrf2 pathways [105,106,107,108]. One of these pathways involves the aryl hydrocarbon receptor (AhR)-dependent induction of CYP1A1 and CYP1A1-dependent substrate metabolism, which, in turn, enhances Nrf2 expression. The AhR also directly binds to the Nrf2 promoter and induces the levels of this protein [106].
The protective properties of Nrf2 are dependent on the induction of Nrf2 and Nrf2-dependent genes, as illustrated in Figure 2, and there is evidence that many of the constituents in coffee, including chlorogenic acids, phenolics, caffeine, cafestrol, and kahweol, are inducers of Nrf2 [109,110,111,112,113,114,115,116,117,118,119]. Moreover, a recent study showed that aqueous coffee extracts induced Nrf2 and Nrf2-dependent genes in various cell lines, and the co-operative role of the coffee-induced activation of AhR/Nrf2 had been reported in several in vitro and in vivo studies involving coffee extracts [120,121,122,123,124,125]. The activation of Nrf2 and Nrf2 protective genes by coffee and its individual components is consistent with their activity to protect nontransformed cells from oxidative stress. However, there is also evidence that the cytotoxicity of coffee components can occur by multiple pathways, including the disruption of the mitochondrial membrane, and this results in a loss of mitochondrial membrane potential and ROS-dependent cytotoxicity [125,126,127,128,129,130,131] (Figure 3). This pathway has been observed for many ROS-inducing phytochemicals and includes the inactivation of cMyc and cMyc-regulated microRNAs (miRs), the induction of ZBTB genes, which repress Sp1-, Sp2-, and Sp3-regulated genes/pathways [131]. Thus, coffee components activate cell context-dependent antioxidant and oxidative stress pathways that are primarily but not exclusively linked to chemopreventive and chemotherapeutic pathways in non-cancer and cancer cells, respectively.
Although the activation of Nrf2 plays an important role in alleviating oxidative stress, the increased levels of Nrf2 in cancer cells are linked to malignant progression and drug resistance [132]. Coffee extract-induced Nrf2 and the similar effects of other phytochemical-enriched diets may be beneficial in normal cells that require low levels of ROS; however, in cancer cells, this can result in enhanced carcinogenesis [132,133]. Nrf2 levels in cancer cells are enhanced due to overexpressed mutations that activate Nrf2 or mutations in Keap, and this enhances the pro-oncogenic pathways and the development of resistance to anticancer agents [133,134]. For example, in hepatic progenitor cells, Nrf2 induces malignant transformation due to the activation of the wnt-β-catenin pathway [135]; suppression of Nrf2 by histone lysine methyltransferase SETDB2 inhibits the progression of lung adenocarcinoma cells, whereas the decreased SETDB2 in these cells enhances Nrf2-mediated tumorigenesis [136]. It has also been reported that the chemically or genetic-mediated downregulation of Nrf2 increases ROS-medicated cell death and reverses some drug resistance [137,138,139]. Interestingly, coffee extracts contain trigonelline, which inhibits Nrf2 activity and thereby modulates the potentially beneficial and harmful effects of NRF2. The inhibitor of Nrf2 trigonelline and other coffee components inhibit cancer cell growth, migration, and drug resistance [140,141,142,143,144,145]. In contrast, Nrf2 inhibitors, such as trigonelline, can interfere with Nrf2-mediated antioxidant activity and thereby reverse some of the protective effects of Nrf2. Thus, coffee and some of its key chemical components (chlorogenic acid and phenolics trigonelline) and pathways (Nrf2 and ROS) exhibit cell context-dependent opposing effects. This suggests that other cell-specific factors must also contribute to the regulation of other pathways/genes that are associated with the health benefits of this beverage. Moreover, the issue of coffee and other phytochemicals and their beneficial effects need to be further evaluated with respect to cancer, where Nrf2 may not be beneficial.

6. Receptor-Mediated Responses

The thousands of membrane-associated and intracellular receptors are the key genes that are the sensors of dietary and intracellular cues and are required for maintaining cellular homeostasis and are involved in pathophysiology. PubMed lists over 40,000 manuscripts under the heading “Receptors and aging”, and these include papers on most of the age-related responses, such as decreased mortality and improved neurological responses, which are associated with the higher consumption of coffee. However, the direct linkages between the individual compounds in coffee extracts to the specific receptors that play a significant role in coffee extract-induced health benefits have been understudied. This review identifies some receptors that may serve as targets for coffee components and their associated beneficial health effects.

6.1. Aryl Hydrocarbon Receptor (AhR)

As indicated above, Nrf2 and the AhR are co-operatively activated by coffee extracts, and studies in our laboratories have primarily focused on the direct effects of coffee extracts as AhR ligands in the intestine (Figure 4A) [146]. AhR plays an important role in multiple organs/tissues, and there is extensive evidence that AhR is involved in aging and is related to diseases, suggesting that AhR and its ligands can affect these conditions [147,148,149,150,151]. Aqueous coffee extracts induced the Ah- responsive CYP1A1, CYP1B1, and UGT1A1 genes in Caco-2 colon cancer cells and YAMC mouse colonocytes, and these responses were abrogated in their corresponding AhR knockout (AhR-KO) cell lines. In addition, Ah responsiveness was primarily observed in a chloroform extract of aqueous coffee extracts, and chromatographic and cell culture analysis indicated that caffeine was not an AhR-active compound. The chloroform extracts from coffee were separated by this layer chromatography into three bands: a less polar band, a caffeine band, and a polar band. The extracts of the polar and less polar bands were AhR-active [146]. A comparison between aqueous extracts of unroasted and roasted ground coffee demonstrated that the high levels of AhR-active extracts in brewed coffee were primarily due to the roasting process. In vivo studies showed that coffee extracts inhibited DSS-induced colonic inflammation and inhibited the growth of organoids enriched in colonic stem cells. This latter response is consistent with anticancer activity since colonic stem cells are precursors for the development of colon tumors. We observed some of the same responses for the alkaloid norharman(β-carboline), which is one of the AhR agonists in coffee extracts that contributes to the AhR-dependent responses observed in the study. These results for coffee were observed in wild type but not AhR-KO cells and animal models and are consistent with previous studies showing a protective role for AhR and its ligands in colonic inflammation and cancer [152,153,154,155,156,157]. The identities of most AhR ligands in coffee extracts have not been determined; however, it should be pointed out that AhR and its ligands can be both beneficial and harmful, and these responses are organ/tissue-context-dependent [158]. The beneficial effects of AhR ligands in colon cancer are well supported; however, AhR is also a pro-oncogenic factor in other tumor types, including head and neck cancer [159]. Thus, at least some of the health-promoting effects of coffee are mediated through its binding and activation in AhR signaling.

6.2. Nuclear Receptor 4A1 (NR4A1, Nur77)

NR4A1, NR4A2 (Nurr1), and NR4A3 (Nor1) are orphan nuclear receptors and the immediate, early genes that respond to stress and play a role in maintaining cellular homeostasis and pathophysiology [160,161]. These receptors are often increased in the diseased cell types that are associated with stress, and this includes many solid tumors and derived cancer cell lines [162]. It was hypothesized that NR4A orphan nuclear receptors were the potential targets for anti-aging interventions [163] due to the functions of these receptors in the immune system and stress-related diseases, and this has now been confirmed by several studies [164,165,166,167]. For example, age-related cognitive impairment in mice results in decreased NR4A expression, and treatment with a bis-indole-derived NR4A2 ligand enhanced long-term spatial memory and rescued memory deficits in mouse models [166]. Age-related renal fibrosis was also suppressed by NR4A1 [165], and there are other reports showing that NR4A is involved in age-related diseases. Recent studies reported that polyphenolics, including flavonoids such as quercetin and kaempferol that occur in coffee, bind to NR4A1 in cancer cells, act as receptor antagonists, and inhibit pro-oncogenic NR4A1-regulated genes and pathways (Figure 4B) [168]. When using Rh30 rhabdomyosarcoma cells as a model, it was reported that quercetin and kaempferol decreased NR4A1-dependent transactivation, inhibited Rh30 cell growth, survival, and invasion, decreased tumor growth, and also decreased mTOR signaling and β1-integrin expression. All of these effects were also observed after NR4A1 knockdown or after treatment with other NR4A1 antagonists [168]. Ongoing studies have identified multiple polyphenols that are present in coffee extracts and in Mediterranean diets. Thus, it is likely that NR4A1 and its phytochemical ligands may play important roles in the health-promoting effects of coffee and phytochemical-enriched diets, and this needs to be further investigated.

7. Other Coffee-Induced Pathways

7.1. Changes in DNA Methylation

Based on the health benefits of coffee consumption, an epigenome-wide association meta-analysis of DNA methylation with coffee drinkers was reported [169]. The data were generated from epigenome-wide association studies of coffee (and tea) consumption among 15,789 European and African American participants from 15 cohorts. An analysis of the data identified several CpGs that were related to coffee consumption, and these included the AHRR, F2RL3, FLJ43663, HDAC4, GFL1, and PHGDH genes. The relationship between these genes and the effects of coffee is uncertain, although it was suggested that PHGDH might be associated with protection from liver damage. In addition, AHRR represses AhR activation and could also affect AhR activity. The nuclear receptor NR4A1 regulates the methyltransferase gene G9A [168], and, possibly, the other methyltransferases and the epigenetic effects of coffee could be due, in part, to NR4A1 ligands, such as flavonoids and other polyphenolics in coffee extracts. Thus, the linkage between coffee extract-induced epigenetic changes as causal factors needs to be further investigated, along with parallel comparisons with animal models.

7.2. Coffee-Induced Microbiome Changes and Health

The gut microbial populations and their metabolites strongly influence intestinal health and also affect distal organs, and there is extensive evidence that the influence of diet on health may be due, in part, to changes in the gut microbiome [170,171,172,173]. The effects of coffee on the gastrointestinal system have recently been reviewed, and it is clear that coffee modulates the composition of intestinal microbiota and microbial metabolites [55]. Moreover, in mouse models, caffeine-induced sleep restriction affects the composition of the gut microbiome and fecal metabolites [174]. For example, the nondigestible polysaccharides in coffee are rapidly metabolized to short-chain fatty acids in the gut, and this results in increased levels of Bacteroides/Prevotella species [175,176]. Another study also reported higher levels of Bacteroides-Prevotella-Porphyromonas in high consumers of coffee [177], and this was also accompanied by lower levels of lipoperoxidation and the increased microbial production of short-chain fatty acids that are chemoprotective in the gut [178]. It was also observed in a human dietary study that a high fiber/coffee (no red meat) diet improved insulin sensitivity in type 2 diabetes and decreased the expression of the pro-inflammatory marker interleukin-18 [179]. It was reported that Prevotella induced IL-18 and other inflammatory cytokines in the colon of SPF mice, whereas inflammation (DSS), induced colonic inflammation, and cytokine production were inhibited by Prevotella, and this was consistent with the overall beneficial effects of Prevotella [180], which is enhanced in coffee drinkers. The results of several other studies indicate that coffee induces bacterial species and their metabolites that are known to have beneficial effects [181,182,183]. However, due to the variability in the diet-induced changes of the intestinal microbes and their metabolites [172,184], the role of these coffee-induced effects on changes in microbial populations and microbial metabolites is not yet fully understood.

8. Summary

Coffee is not only the most consumed beverage worldwide, but it joins the Mediterranean diet as being among those dietary components that extend life, protect against neurological and liver diseases, and protect against the diseases of other organs. There is also an association between higher coffee consumption and overall anti-inflammatory effects and protection against some cancers, whereby coffee acts as both a chemopreventive and chemotherapeutic agent. The mechanisms of action of coffee are dependent on the effects of its constituents, including chlorogenic acids, polyphenolics, terpenoids, alkaloids, and other phytochemicals. Caffeine may contribute to some coffee-induced responses, but there are studies showing similar health benefits in individuals consuming caffeinated or decaffeinated coffee. There is evidence that the antioxidant activity of coffee, which activates Nrf2, may be an important mechanism of action. However, since Nrf2 exhibits both health-protective and drug-resistant activities, other cell context-dependent factors may also be important. There is also evidence that the protective effects of coffee in the gut and decreased colon cancer risk may be due to its activity as an AhR ligand. Moreover, some of the components of coffee bind the orphan nuclear receptor NR4A1 to the interactions with this receptor, and as of yet, unidentified receptors may also be important. Overall, these mechanisms, in concert with possible epigenetic pathways and the modulation of gut microbiota/microbial metabolites, contribute to the health benefits of higher coffee consumption, and this suggests that clinical applications of coffee extracts, particularly for treating some cancers, should be considered.

Author Contributions

Conceptualization, S.S. and R.S.C.; Experiment Design, S.S. and R.S.C.; investigation, S.S., J.K., S.U. and A.H.; writing—original draft preparation, S.S. and L.A.D.; writing—review and editing, S.S., R.S.C. and L.A.D.; funding acquisition, S.S. and R.S.C. All authors have read and agreed to the published version of the manuscript.

Funding

Funding was provided by Texas AgriLife Research, the Sid Kyle Chair Endowment, the Allen Endowed Chair in Nutrition & Chronic Disease Prevention, the Cancer Prevention Research Institute of Texas (RP160589), and the National Institutes of Health (R01-ES025713, R01-CA202697, R35-CA197707, P30-ES029067, and T32-CA090301).

Institutional Review Board Statement

All experiments involving mice were approved by Texas A&M University’s Animal Care and Use Committee.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data available in a publicly accessible repository.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Chu, Y.F. Coffee: Emerging Health Effects and Disease Prevention. In Coffee: Emerging Health Effects and Disease Prevention; John Wiley & Sons, Inc.: Hoboken, NJ, USA, 2012. [Google Scholar] [CrossRef]
  2. Hou, C.; Zeng, Y.; Chen, W.; Han, X.; Yang, H.; Ying, Z.; Hu, Y.; Sun, Y.; Qu, Y.; Fang, F.; et al. Medical conditions associated with coffee consumption: Disease-trajectory and comorbidity network analyses of a prospective cohort study in UK Biobank. Am. J. Clin. Nutr. 2022, 116, 730–740. [Google Scholar] [CrossRef] [PubMed]
  3. Van Dam, R.M.; Hu, F.B.; Willett, W.C. Coffee, Caffeine, and Health. N. Engl. J. Med. 2020, 383, 369–378. [Google Scholar] [CrossRef] [PubMed]
  4. Kim, S.A.; Tan, L.J.; Shin, S. Coffee Consumption and the Risk of All-Cause and Cause-Specific Mortality in the Korean Population. J. Acad. Nutr. Diet. 2021, 121, P2221–P2232.e4. [Google Scholar] [CrossRef]
  5. Di Maso, M.; Boffetta, P.; Negri, E.; la Vecchia, C.; Bravi, F. Caffeinated Coffee Consumption and Health Outcomes in the US Population: A Dose-Response Meta-Analysis and Estimation of Disease Cases and Deaths Avoided. Adv. Nutr. 2021, 12, 1160–1176. [Google Scholar] [CrossRef] [PubMed]
  6. Liu, D.; Li, Z.H.; Shen, D.; Zhang, P.D.; Song, W.-Q.; Zhang, W.-T.; Huang, Q.-M.; Chen, P.L.; Zhang, X.R.; Mao, C. Association of Sugar-Sweetened, Artificially Sweetened, and Unsweetened Coffee Consumption With All-Cause and Cause-Specific Mortality: A Large Prospective Cohort Study. Ann. Intern. Med. 2022, 175, 909–917. [Google Scholar] [CrossRef]
  7. Lin, P.; Liang, Z.; Wang, M. Caffeine consumption and mortality in populations with different weight statuses: An analysis of NHANES 1999–2014. Nutrition 2022, 102, 111731. [Google Scholar] [CrossRef]
  8. Wang, T.; Wu, Y.; Wang, W.; Zhang, D. Association between coffee consumption and functional disability in older US adults. Br. J. Nutr. 2021, 125, 695–702. [Google Scholar] [CrossRef]
  9. Feng, J.; Wang, J.; Jose, M.; Seo, Y.; Feng, L.; Ge, S. Association between Caffeine Intake and All-Cause and Cause-Specific Mortality: An Analysis of the National Health and Nutrition Examination Survey (NHANES) 1999–2014 Database. Nurs. Rep. 2021, 11, 901–912. [Google Scholar] [CrossRef]
  10. Doepker, C.; Movva, N.; Cohen, S.S.; Wikoff, D.S. Benefit-risk of coffee consumption and all-cause mortality: A systematic review and disability adjusted life year analysis. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2022, 170, 113472. [Google Scholar] [CrossRef]
  11. Shin, S.; Lee, J.E.; Loftfield, E.; Shu, X.O.; Abe, S.K.; Rahman, M.S.; Saito, E.; Islam, M.R.; Tsugane, S.; Sawada, N.; et al. Coffee and tea consumption and mortality from all causes, cardiovascular disease and cancer: A pooled analysis of prospective studies from the Asia Cohort Consortium. Int. J. Epidemiol. 2022, 51, 626–640. [Google Scholar] [CrossRef]
  12. Torres-Collado, L.; Compañ-Gabucio, L.M.; González-Palacios, S.; Notario-Barandiaran, L.; Oncina-Cánovas, A.; Vioque, J.; García-de la Hera, M. Coffee Consumption and All-Cause, Cardiovascular, and Cancer Mortality in an Adult Mediterranean Population. Nutrients 2021, 13, 1241. [Google Scholar] [CrossRef] [PubMed]
  13. Fraser, G.E.; Cosgrove, C.M.; Mashchak, A.D.; Orlich, M.J.; Altekruse, S.F. Lower rates of cancer and all-cause mortality in an Adventist cohort compared with a US Census population. Cancer 2020, 126, 1102–1111. [Google Scholar] [CrossRef] [PubMed]
  14. Shan, Z.; Wang, F.; Li, Y.; Baden, M.Y.; Bhupathiraju, S.N.; Wang, D.D.; Sun, Q.; Rexrode, K.M.; Rimm, E.B.; Qi, L.; et al. Healthy Eating Patterns and Risk of Total and Cause-Specific Mortality. JAMA Intern. Med. 2023; ahead of print. [Google Scholar] [CrossRef]
  15. Chen, Y.; Zhang, Y.; Zhang, M.; Yang, H.; Wang, Y. Consumption of coffee and tea with all-cause and cause-specific mortality: A prospective cohort study. BMC Med. 2022, 20, 449. [Google Scholar] [CrossRef] [PubMed]
  16. Park, Y.; Cho, H.; Myung, S.K. Effect of Coffee Consumption on Risk of Coronary Heart Disease in a Systematic Review and Meta-Analysis of Prospective Cohort Studies. Am. J. Cardiol. 2023, 186, 17–29. [Google Scholar] [CrossRef]
  17. Cornelis, M.C.; van Dam, R.M. Habitual coffee and tea consumption and cardiometabolic biomarkers in the UK biobank: The role of beverage types and genetic variation. J. Nutr. 2020, 150, 2772–2788. [Google Scholar] [CrossRef]
  18. Zhou, A.; Hyppönen, E. Long-term coffee consumption, caffeine metabolism genetics, and risk of cardiovascular disease: A prospective analysis of up to 347,077 individuals and 8368 cases. Am. J. Clin. Nutr. 2019, 109, 509–516. [Google Scholar] [CrossRef] [PubMed]
  19. Teramoto, M.; Yamagishi, K.; Muraki, I.; Tamakoshi, A.; Iso, H. Coffee and Green Tea Consumption and Cardiovascular Disease Mortality Among People With and Without Hypertension. J. Am. Heart Assoc. 2022, 12, e026477. [Google Scholar] [CrossRef]
  20. Borghi, C. Coffee and blood pressure: Exciting news! Blood Press. 2022, 31, 284–287. [Google Scholar] [CrossRef]
  21. Ruggiero, E.; di Castelnuovo, A.; Costanzo, S.; Persichillo, M.; de Curtis, A.; Cerletti, C.; Donati, M.B.; de Gaetano, G.; Iacoviello, L.; Bonaccio, M. Daily Coffee Drinking Is Associated with Lower Risks of Cardiovascular and Total Mortality in a General Italian Population: Results from the Moli-sani Study. J. Nutr. 2021, 151, 395–404. [Google Scholar] [CrossRef]
  22. Zhang, Y.; Yang, H.; Li, S.; Li, W.D.; Wang, Y. Consumption of coffee and tea and risk of developing stroke, dementia, and poststroke dementia: A cohort study in the UK Biobank. PLoS Med. 2021, 18, e1003830. [Google Scholar] [CrossRef]
  23. Tran, A.; Zhang, C.Y.; Cao, C. The Role of Coffee in the Therapy of Parkinson’s Disease. J. Alzheimer’s Dis. Park. 2016, 5, 203. [Google Scholar] [CrossRef]
  24. Herden, L.; Weissert, R. The Impact of Coffee and Caffeine on Multiple Sclerosis Compared to Other Neurodegenerative Diseases. Front. Nutr. 2018, 5, 133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Wang, M.; Cornelis, M.C.; Zhang, Z.; Liu, D.; Lian, X. Mendelian randomization study of coffee consumption and age at onset of Huntington’s disease. Clin. Nutr. 2021, 40, 5615–5618. [Google Scholar] [CrossRef] [PubMed]
  26. Fisicaro, F.; Lanza, G.; Pennisi, M.; Vagli, C.; Cantone, M.; Pennisi, G.; Ferri, R.; Bella, R. Moderate Mocha Coffee Consumption Is Associated with Higher Cognitive and Mood Status in a Non-Demented Elderly Population with Subcortical Ischemic Vascular Disease. Nutrients 2021, 13, 536. [Google Scholar] [CrossRef]
  27. Yahalom, G.; Rigbi, A.; Israeli-Korn, S.; Krohn, L.; Rudakou, U.; Ruskey, J.A.; Benshimol, L.; Tsafnat, T.; Gan-Or, Z.; Hassin-Baer, S.; et al. Age at Onset of Parkinson’s Disease Among Ashkenazi Jewish Patients: Contribution of Environmental Factors, LRRK2 p.G2019S and GBA p.N370S Mutations. J. Park. Dis. 2010, 10, 1123–1132. [Google Scholar] [CrossRef]
  28. Chan, L.; Hong, C.T.; Bai, C.H. Coffee consumption and the risk of cerebrovascular disease: A meta-analysis of prospective cohort studies. BMC Neurol. 2021, 21, 380. [Google Scholar] [CrossRef]
  29. Kolb, H.; Martin, S.; Kempf, K. Coffee and lower risk of type 2 diabetes: Arguments for a causal relationship. Nutrients 2021, 13, 1144. [Google Scholar] [CrossRef]
  30. Imamura, F.; Schulze, M.B.; Sharp, S.J.; Guevara, M.; Romaguera, D.; Bendinelli, B.; Salamanca-Fernández, E.; Ardanaz, E.; Arriola, L.; Aune, D.; et al. Estimated Substitution of Tea or Coffee for Sugar-Sweetened Beverages Was Associated with Lower Type 2 Diabetes Incidence in Case-Cohort Analysis across 8 European Countries in the EPIC-InterAct Study. J. Nutr. 2019, 149, 1985–1993. [Google Scholar] [CrossRef] [Green Version]
  31. Hang, D.; Kværner, A.S.; Ma, W.; Hu, Y.; Tabung, F.K.; Nan, H.; Hu, Z.; Shen, H.; Mucci, L.A.; Chan, A.T.; et al. Coffee consumption and plasma biomarkers of metabolic and inflammatory pathways in US health professionals. Am. J. Clin. Nutr. 2019, 109, 635–647. [Google Scholar] [CrossRef]
  32. Komorita, Y.; Iwase, M.; Fujii, H.; Ohkuma, T.; Ide, H.; Jodai-Kitamura, T.; Yoshinari, M.; Oku, Y.; Higashi, T.; Nakamura, U.; et al. Additive effects of green tea and coffee on all-cause mortality in patients with type 2 diabetes mellitus: The Fukuoka Diabetes Registry. BMJ Open Diabetes Res. Care 2020, 8, e001252. [Google Scholar] [CrossRef]
  33. Wang, S.; Han, Y.; Zhao, H.; Han, X.; Yin, Y.; Wu, J.; Zhang, Y.; Zeng, X. Association between Coffee Consumption, Caffeine Intake, and Metabolic Syndrome Severity in Patients with Self-Reported Rheumatoid Arthritis: National Health and Nutrition Examination Survey 2003–2018. Nutrients 2022, 15, 107. [Google Scholar] [CrossRef]
  34. Kennedy, O.J.; Fallowfield, J.A.; Poole, R.; Hayes, P.C.; Parkes, J.; Roderick, P.J. All coffee types decrease the risk of adverse clinical outcomes in chronic liver disease: A UK Biobank study. BMC Public Health 2021, 21, 970. [Google Scholar] [CrossRef] [PubMed]
  35. Coelho, M.; Patarrão, R.S.; Sousa-Lima, I.; Ribeiro, R.T.; Meneses, M.J.; Andrade, R.; Mendes, V.M.; Manadas, B.; Raposo, J.F.; Macedo, M.P.; et al. Increased Intake of Both Caffeine and Non-Caffeine Coffee Components Is Associated with Reduced NAFLD Severity in Subjects with Type 2 Diabetes. Nutrients 2022, 15, 4. [Google Scholar] [CrossRef] [PubMed]
  36. Yuan, S.; Chen, J.; Li, X.; Fan, R.; Arsenault, B.; Gill, D.; Giovannucci, E.L.; Zheng, J.S.; Larsson, S.C. Lifestyle and metabolic factors for nonalcoholic fatty liver disease: Mendelian randomization study. Eur. J. Epidemiol. 2022, 37, 723–733. [Google Scholar] [CrossRef]
  37. Yang, J.; Tobias, D.K.; Li, S.; Bhupathiraju, S.N.; Ley, S.H.; Hinkle, S.N.; Qian, F.; Chen, Z.; Zhu, Y.; Bao, W.; et al. Habitual coffee consumption and subsequent risk of type 2 diabetes in individuals with a history of gestational diabetes—A prospective study. Am. J. Clin. Nutr. 2022, 116, 1693–1703. [Google Scholar] [CrossRef] [PubMed]
  38. Lee, H.J.; Park, J.I.; Kwon, S.O.; Hwang, D.D. Coffee consumption and diabetic retinopathy in adults with diabetes mellitus. Sci. Rep. 2022, 12, 3547. [Google Scholar] [CrossRef]
  39. Barré, T.; Fontaine, H.; Pol, S.; Ramier, C.; Di Beo, V.; Protopopescu, C.; Marcellin, F.; Bureau, M.; Bourlière, M.; Dorival, C.; et al. Metabolic Disorders in Patients with Chronic Hepatitis B Virus Infection: Coffee as a Panacea? (ANRS CO22 Hepather Cohort). Antioxidants 2022, 11, 379. [Google Scholar] [CrossRef]
  40. Ng, S.C.; Tang, W.; Leong, R.W.; Chen, M.; Ko, Y.; Studd, C.; Niewiadomski, O.; Bell, S.; Kamm, M.A.; de Silva, H.J.; et al. Environmental risk factors in inflammatory bowel disease: A population-based case-control study in Asia-Pacific. Gut 2015, 64, 1063–1071. [Google Scholar] [CrossRef] [Green Version]
  41. Cohen, A.B.; Lee, D.; Long, M.D.; Kappelman, M.D.; Martin, C.F.; Sandler, R.S.; Lewis, J.D. Dietary patterns and self-reported associations of diet with symptoms of inflammatory bowel disease. Dig. Dis. Sci. 2013, 58, 1322–1328. [Google Scholar] [CrossRef]
  42. Güngördük, K.; Özdemir, İ.A.; Güngördük, Ö.; Gülseren, V.; Gokçü, M.; Sancı, M. Effects of coffee consumption on gut recovery after surgery of gynecological cancer patients: A randomized controlled trial. Am. J. Obstet. Gynecol. 2017, 216, 145.e1–145.e7. [Google Scholar] [CrossRef] [Green Version]
  43. Jee, H.J.; Lee, S.G.; Bormate, K.J.; Jung, Y.S. Effect of Caffeine Consumption on the Risk for Neurological and Psychiatric Disorders: Sex Differences in Human. Nutrients 2020, 12, 3080. [Google Scholar] [CrossRef] [PubMed]
  44. Lu, M.Y.; Cheng, H.Y.; Lai, J.C.; Chen, S.J. The Relationship between Habitual Coffee Drinking and the Prevalence of Metabolic Syndrome in Taiwanese Adults: Evidence from the Taiwan Biobank Database. Nutrients 2022, 14, 1867. [Google Scholar] [CrossRef] [PubMed]
  45. Carter, P.; Yuan, S.; Kar, S.; Vithayathil, M.; Mason, A.M.; Burgess, S.; Larsson, S.C. Coffee consumption and cancer risk: A Mendelian randomization study. Clin. Nutr. 2022, 41, 2113–2123. [Google Scholar] [CrossRef]
  46. Kim, S.Y.; Yoo, D.M.; Min, C.; Choi, H.G. Association between coffee consumption/physical exercise and gastric, hepatic, colon, breast, uterine cervix, lung, thyroid, prostate, and bladder cancer. Nutrients 2021, 13, 3927. [Google Scholar] [CrossRef]
  47. Nehlig, A. Effects of Coffee on the Gastro-Intestinal Tract: A Narrative Review and Literature Update. Nutrients 2022, 14, 399. [Google Scholar] [CrossRef] [PubMed]
  48. Tran, K.T.; Coleman, H.G.; McMenamin, Ú.C.; Cardwell, C.R. Coffee consumption by type and risk of digestive cancer: A large prospective cohort study. Br. J. Cancer 2019, 120, 1059–1066. [Google Scholar] [CrossRef] [Green Version]
  49. Tanaka, K.; Tamakoshi, A.; Sugawara, Y.; Mizoue, T.; Inoue, M.; Sawada, N.; Matsuo, K.; Ito, H.; Naito, M.; Nagata, C.; et al. Coffee, green tea and liver cancer risk: An evaluation based on a systematic review of epidemiologic evidence among the Japanese population. Jpn. J. Clin. Oncol. 2019, 49, 972–984. [Google Scholar] [CrossRef]
  50. Bhurwal, A.; Rattan, P.; Yoshitake, S.; Pioppo, L.; Reja, D.; Dellatore, P.; Rustgi, V. Inverse association of coffee with liver cancer development an updated systematic review and meta-analysis. J. Gastrointest. Liver Dis. 2020, 29, 421–428. [Google Scholar] [CrossRef]
  51. Loftfield, E.; Rothwell, J.A.; Sinha, R.; Keski-Rahkonen, P.; Robinot, N.; Albanes, D.; Weinstein, S.J.; Derkach, A.; Sampson, J.; Scalbert, A.; et al. Prospective investigation of serum metabolites, coffee drinking, liver cancer incidence, and liver disease mortality. J. Natl. Cancer Inst. 2020, 112, 286–294. [Google Scholar] [CrossRef]
  52. Ugai, T.; Haruki, K.; Vayrynen, J.P.; Zhong, R.; Borowsky, J.; Fujiyoshi, K.; Lau, M.C.; Zhao, M.; Akimoto, N.; Chang, T.W.; et al. Coffee intake and colorectal cancer incidence according to T-cell response. JNCI Cancer Spectr. 2020, 4, pkaa068. [Google Scholar] [CrossRef]
  53. Rosato, V.; Guercio, V.; Bosetti, C.; Gracia-Lavedan, E.; Villanueva, C.M.; Polesel, J.; Toffoluti, F.; Moreno, V.; Martin, V.; Aragonés, N.; et al. Coffee consumption and colorectal cancer risk: A multicentre case-control study from Italy and Spain. Eur. J. Cancer Prev. Off. J. Eur. Cancer Prev. Organ. ECP 2021, 30, 204–210. [Google Scholar] [CrossRef]
  54. Martimianaki, G.; Bertuccio, P.; Alicandro, G.; Pelucchi, C.; Bravi, F.; Carioli, G.; Bonzi, R.; Rabkin, C.S.; Liao, L.M.; Sinha, R.; et al. Coffee consumption and gastric cancer: A pooled analysis from the Stomach cancer Pooling Project consortium. Eur. J. Cancer Prev. 2022, 31, 117–127. [Google Scholar] [CrossRef]
  55. Um, C.Y.; McCullough, M.L.; Guinter, M.A.; Campbell, P.T.; Jacobs, E.J.; Gapstur, S.M. Coffee consumption and risk of colorectal cancer in the Cancer Prevention Study-II Nutrition Cohort. Cancer Epidemiol. 2020, 67, 101730. [Google Scholar] [CrossRef] [PubMed]
  56. Deng, Y.; Huang, J.; Wong, M.C.S. Associations of alcohol and coffee with colorectal cancer risk in East Asian populations: A Mendelian randomization study. Eur. J. Nutr. 2022; ahead of print. [Google Scholar] [CrossRef]
  57. Lu, Y.T.; Gunathilake, M.; Lee, J.; Kim, Y.; Oh, J.H.; Chang, H.J.; Sohn, D.K.; Shin, A.; Kim, J. Coffee consumption and its interaction with the genetic variant AhR rs2066853 in colorectal cancer risk: A case-control study in Korea. Carcinogenesis 2022, 43, 203–216. [Google Scholar] [CrossRef]
  58. Bae, J.M.; Shim, S.R. Coffee consumption and pancreatic cancer risk: A meta-epidemiological study of population-based cohort studies. Asian Pac. J. Cancer Prev. 2020, 21, 2793–2798. [Google Scholar] [CrossRef] [PubMed]
  59. Zhou, C.D.; Kuan, A.S.; Reeves, G.K.; Green, J.; Floud, S.; Beral, V.; Yang, T.Y.O. Coffee and pancreatic cancer risk among never-smokers in the UK prospective Million Women Study. Int. J. Cancer 2019, 145, 1484–1492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Song, H.; Shen, X.; Chu, Q.; Zheng, X. Coffee consumption is not associated with the risk of gastric cancer: An updated systematic review and meta-analysis of prospective cohort studies. Nutr. Res. 2022, 102, 35–44. [Google Scholar] [CrossRef]
  61. Zeng, S.B.; Weng, H.; Zhou, M.; Duan, X.L.; Shen, X.F.; Zeng, X.T. Long-Term Coffee Consumption and Risk of Gastric Cancer: A PRISMA-Compliant Dose-Response Meta-Analysis of Prospective Cohort Studies. Medicine 2015, 94, e1640. [Google Scholar] [CrossRef]
  62. Zamora-Ros, R.; Luján-Barroso, L.; Bueno-De-Mesquita, H.B.; Dik, V.K.; Boeing, H.; Steffen, A.; Tjønneland, A.; Olsen, A.; Bech, B.H.; Overvad, K.; et al. Tea and coffee consumption and risk of esophageal cancer: The European prospective investigation into cancer and nutrition study. Int. J. Cancer 2014, 135, 1470–1479. [Google Scholar] [CrossRef]
  63. Zhang, J.; Zhou, B.; Hao, C. Coffee consumption and risk of esophageal cancer incidence: A meta-analysis of epidemiologic studies. Medicine 2018, 97, e0514. [Google Scholar] [CrossRef]
  64. Park, C.H.; Myung, S.K.; Kim, T.Y.; Seo, H.G.; Jeon, Y.J.; Kim, Y. Coffee consumption and risk of prostate cancer: A meta-analysis of epidemiological studies. BJU Int. 2010, 106, 762–769. [Google Scholar] [CrossRef] [PubMed]
  65. Lu, Y.; Zhai, L.; Zeng, J.; Peng, Q.; Wang, J.; Deng, Y.; Xie, L.; Mo, C.; Yang, S.; Li, S.; et al. Coffee consumption and prostate cancer risk: An updated meta-analysis. Cancer Causes Control 2014, 25, 591–604. [Google Scholar] [CrossRef] [PubMed]
  66. Xia, J.D.; Chen, J.; Xue, J.X.; Yang, J.; Wang, Z.J. An up-to-date meta-analysis of coffee consumption and risk of prostate cancer. Urol. J. 2017, 14, 4079–4088. [Google Scholar]
  67. Yu, X.; Bao, Z.; Zou, J.; Dong, J. Coffee consumption and risk of cancers: A meta-analysis of cohort studies. BMC Cancer 2011, 11, 96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Zhong, S.; Chen, W.; Yu, X.; Chen, Z.; Hu, Q.; Zhao, J. Coffee consumption and risk of prostate cancer: An up-to-date meta-analysis. Eur. J. Clin. Nutr. 2014, 68, 330–337. [Google Scholar] [CrossRef] [PubMed]
  69. Cao, S.; Liu, L.; Yin, X.; Wang, Y.; Liu, J.; Lu, Z. Coffee consumption and risk of prostate cancer: A meta-analysis of prospective cohort studies. Carcinogenesis 2014, 35, 256–261. [Google Scholar] [CrossRef] [Green Version]
  70. Liu, H.; Hu, G.H.; Wang, X.C.; Huang, T.B.; Xu, L.; Lai, P.; Guo, Z.F.; Xu, Y.F. Coffee consumption and prostate cancer risk: A meta-analysis of cohort studies. Nutr. Cancer 2015, 67, 392–400. [Google Scholar] [CrossRef]
  71. Chen, X.; Zhao, Y.; Tao, Z.; Wang, K. Coffee consumption and risk of prostate cancer: A systematic review and meta-analysis. BMJ Open 2021, 11, e038902. [Google Scholar] [CrossRef]
  72. Imatoh, T.; Sawada, N.; Yamaji, T.; Iwasaki, M.; Inoue, M.; Tsugane, S. Association between Coffee Consumption and Risk of Prostate Cancer in Japanese Men: A Population-Based Cohort Study in Japan. Cancer Epidemiol. Biomark. Prev. 2022, 31, 471–478. [Google Scholar] [CrossRef]
  73. Dai, Z.-W.; Cai, K.-D.; Li, F.-R.; Wu, X.-B.; Chen, G.-C. Association between coffee consumption and risk of bladder cancer in a meta-analysis of 16 prospective studies. Nutr. Metab. 2019, 16, 66. [Google Scholar] [CrossRef] [Green Version]
  74. Deng, Y.; Wu, T.; Luo, G.; Chen, L. Exploring the casual association between coffee intake and bladder cancer risk using Mendelian Randomization. Front. Genet. 2022, 13, 992599. [Google Scholar] [CrossRef] [PubMed]
  75. Li, B.H.; Yan, S.Y.; Li, X.H.; Huang, Q.; Luo, L.S.; Wang, Y.Y.; Huang, J.; Jin, Y.H.; Wang, Y.B. Coffee and caffeine consumption and risk of renal cell carcinoma: A Mendelian randomization study. Front. Nutr. 2022, 9, 898279. [Google Scholar] [CrossRef] [PubMed]
  76. Rhee, J.; Lim, R.K.; Purdue, M.P. Coffee consumption and risk of renal cancer: A meta-analysis of cohort evidence. Cancer Causes Control 2022, 33, 101–108. [Google Scholar] [CrossRef]
  77. Rhee, J.; Loftfield, E.; Freedman, N.D.; Liao, L.M.; Sinha, R.; Purdue, M.P. Coffee consumption and risk of renal cell carcinoma in the NIH-AARP Diet and Health Study. Int. J. Epidemiol. 2021, 50, 1473–1481. [Google Scholar] [CrossRef] [PubMed]
  78. Makino, T.; Izumi, K.; Hiratsuka, K.; Kano, H.; Shimada, T.; Nakano, T.; Kadomoto, S.; Naito, R.; Iwamoto, H.; Yaegashi, H.; et al. Anti-proliferative and anti-migratory properties of coffee diterpenes kahweol acetate and cafestol in human renal cancer cells. Sci. Rep. 2021, 11, 675. [Google Scholar] [CrossRef] [PubMed]
  79. Nehlig, A.; Reix, N.; Arbogast, P.; Mathelin, C. Coffee consumption and breast cancer risk: A narrative review in the general population and in different subtypes of breast cancer. Eur. J. Nutr. 2021, 60, 1197–1235. [Google Scholar] [CrossRef]
  80. Zheng, K.H.; Zhu, K.; Wactawski-Wende, J.; Freudenheim, J.L.; LaMonte, M.J.; Hovey, K.M.; Mu, L. Caffeine intake from coffee and tea and invasive breast cancer incidence among postmenopausal women in the Women’s Health Initiative. Int. J. Cancer 2021, 149, 2032–2044. [Google Scholar] [CrossRef]
  81. Yaghjyan, L.; McLaughlin, E.; Lehman, A.; Neuhouser, M.L.; Rohan, T.; Lane, D.S.; Snetselaar, L.; Paskett, E. Associations of coffee/caffeine consumption with postmenopausal breast cancer risk and their interactions with postmenopausal hormone use. Eur. J. Nutr. 2022, 61, 3449–3459. [Google Scholar] [CrossRef]
  82. Gapstur, S.M.; Gaudet, M.M.; Wang, Y.; Hodge, R.A.; Um, C.Y.; Hartman, T.J.; McCullough, M.L. Coffee Consumption and Invasive Breast Cancer Incidence among Postmenopausal Women in the Cancer Prevention Study-II Nutrition Cohort. Cancer Epidemiol. Biomark. Prev. 2020, 29, 2383–2386. [Google Scholar] [CrossRef]
  83. Li, Y.; Ma, L. The association between coffee intake and breast cancer risk: A meta-analysis and dose-response analysis using recent evidence. Ann. Palliat. Med. 2021, 10, 3804–3816. [Google Scholar] [CrossRef]
  84. Sánchez-Quesada, C.; Romanos-Nanclares, A.; Navarro, A.M.; Gea, A.; Cervantes, S.; Martínez-González, M.Á.; Toledo, E. Coffee consumption and breast cancer risk in the SUN project. Eur. J. Nutr. 2020, 59, 3461–3471. [Google Scholar] [CrossRef]
  85. Liu, M.; Song, S.S.; Park, S. Coffee Intake Interacted with the Bcl-2 rs1944420, rs7236090, and rs2849382 Haplotype to Influence Breast Cancer Risk in Middle-Aged Women. Nutr. Cancer 2022, 74, 131–140. [Google Scholar] [CrossRef] [PubMed]
  86. Gao, Y.; Zhai, P.; Jiang, F.; Zhou, F.; Wang, X. Association between coffee drinking and endometrial cancer risk: A meta-analysis. J. Obstet. Gynaecol. Res. 2022, 48, 774–795. [Google Scholar] [CrossRef] [PubMed]
  87. Crous-Bou, M.; Du, M.; Gunter, M.J.; Setiawan, V.W.; Schouten, L.J.; Shu, X.O.; Wentzensen, N.; Bertrand, K.A.; Cook, L.S.; Friedenreich, C.M.; et al. Coffee consumption and risk of endometrial cancer: A pooled analysis of individual participant data in the Epidemiology of Endometrial Cancer Consortium (E2C2). Am. J. Clin. Nutr. 2022, 116, 1219–1228. [Google Scholar] [CrossRef] [PubMed]
  88. Zhu, J.; Smith-Warner, S.A.; Yu, D.; Zhang, X.; Blot, W.J.; Xiang, Y.B.; Sinha, R.; Park, Y.; Tsugane, S.; White, E.; et al. Associations of coffee and tea consumption with lung cancer risk. Int. J. Cancer 2021, 148, 2457–2470. [Google Scholar] [CrossRef]
  89. Seow, W.J.; Koh, W.P.; Jin, A.; Wang, R.; Yuan, J.M. Associations between tea and coffee beverage consumption and the risk of lung cancer in the Singaporean Chinese population. Eur. J. Nutr. 2020, 59, 3083–3091. [Google Scholar] [CrossRef]
  90. Kudwongsa, W.; Promthet, S.; Suwanrungruang, K.; Phunmanee, A.; Vatanasapt, P. Coffee consumption and lung cancer risk: A prospective cohort study in Khon Kaen Thailand. Asian Pac. J. Cancer Prev. 2020, 21, 2367–2371. [Google Scholar] [CrossRef]
  91. Cote, D.J.; Bever, A.M.; Wilson, K.M.; Smith, T.R.; Smith-Warner, S.A.; Stampfer, M.J. A prospective study of tea and coffee intake and risk of glioma. Int. J. Cancer 2020, 146, 2442–2449. [Google Scholar] [CrossRef]
  92. Pranata, R.; Feraldho, A.; Lim, M.A.; Henrina, J.; Vania, R.; Golden, N.; July, J. Coffee and tea consumption and the risk of glioma: A systematic review and dose-response meta-analysis. Br. J. Nutr. 2022, 127, 78–86. [Google Scholar] [CrossRef]
  93. Creed, J.H.; Smith-Warner, S.A.; Gerke, T.A.; Egan, K.M. A prospective study of coffee and tea consumption and the risk of glioma in the UK Biobank. Eur. J. Cancer 2020, 129, 123–131. [Google Scholar] [CrossRef]
  94. Salaroli, L.B.; Ferreira, J.R.S.; Prado, C.B.D.; De Podestá, O.P.G.; Carvalho, A.L.; Mercante, A.M.D.C.; Toporcov, T.N. Cumulative Coffee Consumption as a Protective Factor for Head and Neck Cancer in Brazil. Nutr. Cancer 2022, 75, 228–235. [Google Scholar] [CrossRef] [PubMed]
  95. Karalexi, M.A.; Dessypris, N.; Clavel, J.; Metayer, C.; Erdmann, F.; Orsi, L.; Kang, A.Y.; Schüz, J.; Bonaventure, A.; Greenop, K.R.; et al. Coffee and tea consumption during pregnancy and risk of childhood acute myeloid leukemia: A Childhood Leukemia International Consortium (CLIC) study. Cancer Epidemiol. 2019, 62, 101581. [Google Scholar] [CrossRef] [PubMed]
  96. Madsen, C.N.; Henriksen, T.B.; Ramlau-Hansen, C.H.; Parner, E.T.; Olsen, J.; Bech, B.H. Coffee intake during pregnancy and childhood acute leukemia—A cohort study. Cancer Epidemiol. 2020, 67, 101747. [Google Scholar] [CrossRef] [PubMed]
  97. Soldato, D.; Havas, J.; Presti, D.; Lapidari, P.; Rassy, N.; Pistilli, B.; Martin, E.; del Mastro, L.; Martin, A.-L.; Jacquet, A.; et al. 1694P Coffee and tea consumption (CTC), patient-reported (PRO), and clinical outcomes in a longitudinal study of patients (pts) with breast cancer (BC). Ann. Oncol. 2021, 32, S1184. [Google Scholar] [CrossRef]
  98. Farvid, M.S.; Spence, N.D.; Rosner, B.A.; Willett, W.C.; Eliassen, A.H.; Holmes, M.D. Post-diagnostic coffee and tea consumption and breast cancer survival. Br. J. Cancer 2021, 124, 1873–1881. [Google Scholar] [CrossRef]
  99. Mackintosh, C.; Yuan, C.; Ou, F.S.; Zhang, S.; Niedzwiecki, D.; Chang, I.W.; O’Neil, B.H.; Mullen, B.C.; Lenz, H.J.; Blanke, C.D.; et al. Association of Coffee Intake with Survival in Patients with Advanced or Metastatic Colorectal Cancer. JAMA Oncol. 2020, 6, 1713. [Google Scholar] [CrossRef]
  100. Rubino, R.; Iuliucci, M.R.; Gatani, S.; Piscosquito, A.; D’Ambrosio, B.; Ingenito, C.; Scafuri, L.; Buonerba, C.; Di Lorenzo, G. Mediterranean Diet as a Supportive Intervention in Cancer Patients: Current Evidence and Future Directions. Curr. Oncol. 2022, 29, 7579–7582. [Google Scholar] [CrossRef]
  101. Mokhtari, E.; Jamshidi, S.; Farhadnejad, H.; Teymoori, F.; Rashidkhani, B.; Mirmiran, P.; Tehrani, F.R.; Heidari, Z. The relationship between Mediterranean-DASH diet intervention for the neurodegenerative delay (MIND) Diet and risk of breast Cancer: A case-control study among iranian adult women. BMC Nutr. 2022, 8, 123. [Google Scholar] [CrossRef]
  102. Kolb, H.; Kempf, K.; Martin, S. Health effects of coffee: Mechanism unraveled? Nutrients 2020, 12, 1842. [Google Scholar] [CrossRef]
  103. Sies, H.; Jones, D.P. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat. Rev. Mol. Cell Biol. 2020, 21, 363–383. [Google Scholar] [CrossRef]
  104. Gorrini, C.; Harris, I.S.; Mak, T.W. Modulation of oxidative stress as an anticancer strategy. Nat. Rev. Drug Discov. 2013, 12, 931–947. [Google Scholar] [CrossRef] [PubMed]
  105. Hassmann, U.; Haupt, L.M.; Smith, R.A.; Winkler, S.; Bytof, G.; Lantz, I.; Griffiths, L.R.; Marko, D. Potential antioxidant response to coffee—A matter of genotype? Meta Gene 2014, 2, 525–539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Miao, W.; Hu, L.; Scrivens, P.J.; Batist, G. Transcriptional Regulation of NF-E2 p45-related Factor (NRF2) Expression by the Aryl Hydrocarbon Receptor-Xenobiotic Response Element Signaling Pathway. J. Biol. Chem. 2005, 280, 20340–20348. [Google Scholar] [CrossRef] [Green Version]
  107. Hayes, J.D.; Dinkova-Kostova, A.T.; Mcmahon, M. Cross-talk between transcription factors AhR and Nrf2: Lessons for cancer chemoprevention from dioxin. Toxicol. Sci. 2009, 111, 199–201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Singh, R.; Chandrashekharappa, S.; Bodduluri, S.R.; Baby, B.V.; Hegde, B.; Kotla, N.G.; Hiwale, A.A.; Saiyed, T.; Patel, P.; Vijay-Kumar, M.; et al. Enhancement of the gut barrier integrity by a microbial metabolite through the Nrf2 pathway. Nat. Commun. 2019, 10, 89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Gupta, A.; Atanasov, A.G.; Li, Y.; Kumar, N.; Bishayee, A. Chlorogenic acid for cancer prevention and therapy: Current status on efficacy and mechanisms of action. Pharmacol. Res. 2022, 186, 106505. [Google Scholar] [CrossRef]
  110. Chen, X.Q.; Yang, J.H.; Cho, S.S.; Kim, J.H.; Xu, J.Q.; Seo, K.; Ki, S.H. 5-Caffeoylquinic acid ameliorates oxidative stress-mediated cell death via Nrf2 activation in hepatocytes. Pharm. Biol. 2020, 58, 999–1005. [Google Scholar] [CrossRef]
  111. Lonati, E.; Carrozzini, T.; Bruni, I.; Mena, P.; Botto, L.; Cazzaniga, E.; del Rio, D.; Labra, M.; Palestini, P.; Bulbarelli, A. Coffee-Derived Phenolic Compounds Activate Nrf2 Antioxidant Pathway in I/R Injury In Vitro Model: A Nutritional Approach Preventing Age Related-Damages. Molecules 2022, 27, 1049. [Google Scholar] [CrossRef]
  112. Sun, W.; Xie, W.; Huang, D.; Cui, Y.; Yue, J.; He, Q.; Jiang, L.; Xiong, J.; Sun, W.; Yi, Q. Caffeic acid phenethyl ester attenuates osteoarthritis progression by activating NRF2/HO 1 and inhibiting the NF κB signaling pathway. Int. J. Mol. Med. 2022, 50, 134. [Google Scholar] [CrossRef]
  113. Volz, N.; Boettler, U.; Winkler, S.; Teller, N.; Schwarz, C.; Bakuradze, T.; Eisenbrand, G.; Haupt, L.; Griffiths, L.R.; Stiebitz, H.; et al. Effect of coffee combining green coffee bean constituents with typical roasting products on the Nrf2/ARE pathway in vitro and in vivo. J. Agric. Food Chem. 2012, 60, 9631–9641. [Google Scholar] [CrossRef]
  114. Kweon, M.H.; In Park, Y.; Sung, H.C.; Mukhtar, H. The novel antioxidant 3-O-caffeoyl-1-methylquinic acid induces Nrf2-dependent phase II detoxifying genes and alters intracellular glutathione redox. Free Radic. Biol. Med. 2006, 40, 1349–1361. [Google Scholar] [CrossRef] [PubMed]
  115. Stähli, A.; Maheen, C.U.; Strauss, F.J.; Eick, S.; Sculean, A.; Gruber, R. Caffeic acid phenethyl ester protects against oxidative stress and dampens inflammation via heme oxygenase 1. Int. J. Oral Sci. 2019, 11, 6. [Google Scholar] [CrossRef] [PubMed]
  116. Amorim, R.; Cagide, F.; Tavares, L.C.; Simões, R.F.; Soares, P.; Benfeito, S.; Baldeiras, I.; Jones, J.G.; Borges, F.; Oliveira, P.J.; et al. Mitochondriotropic antioxidant based on caffeic acid AntiOxCIN4 activates Nrf2-dependent antioxidant defenses and quality control mechanisms to antagonize oxidative stress-induced cell damage. Free Radic. Biol. Med. 2022, 179, 119–132. [Google Scholar] [CrossRef] [PubMed]
  117. Yao, J.; Peng, S.; Xu, J.; Fang, J. Reversing ROS-mediated neurotoxicity by chlorogenic acid involves its direct antioxidant activity and activation of Nrf2-ARE signaling pathway. BioFactors 2019, 45, 616–626. [Google Scholar] [CrossRef]
  118. Balstad, T.R.; Carlsen, H.; Myhrstad, M.C.W.; Kolberg, M.; Reiersen, H.; Gilen, L.; Ebihara, K.; Paur, I.; Blomhoff, R. Coffee, broccoli and spices are strong inducers of electrophile response element-dependent transcription in vitro and in vivo—Studies in electrophile response element transgenic mice. Mol. Nutr. Food Res. 2011, 55, 185–197. [Google Scholar] [CrossRef] [PubMed]
  119. Higgins, L.G.; Cavin, C.; Itoh, K.; Yamamoto, M.; Hayes, J.D. Induction of cancer chemopreventive enzymes by coffee is mediated by transcription factor Nrf2. Evidence that the coffee-specific diterpenes cafestol and kahweol confer protection against acrolein. Toxicol. Appl. Pharmacol. 2008, 226, 328–337. [Google Scholar] [CrossRef] [PubMed]
  120. Toydemir, G.; Loonen, L.M.P.; Venkatasubramanian, P.B.; Mes, J.J.; Wells, J.M.; de Wit, N. Coffee induces AHR- and Nrf2-mediated transcription in intestinal epithelial cells. Food Chem. 2021, 341, 128261. [Google Scholar] [CrossRef] [PubMed]
  121. Yazheng, L.; Kitts, D.D. Activation of antioxidant response element (ARE)-dependent genes by roasted coffee extracts. Food Funct. 2012, 3, 950–954. [Google Scholar] [CrossRef]
  122. Ishikawa, T.; Takahashi, S.; Morita, K.; Okinaga, H.; Teramoto, T. Induction of AhR-mediated gene transcription by coffee. PLoS ONE 2014, 9, e102152. [Google Scholar] [CrossRef] [Green Version]
  123. Kalthoff, S.; Ehmer, U.; Freiberg, N.; Manns, M.P.; Strassburg, C.P. Coffee induces expression of glucuronosyl transferases by the aryl hydrocarbon receptor and Nrf2 in liver and stomach. Gastroenterology 2010, 139, 1699–1710.e2. [Google Scholar] [CrossRef]
  124. Nigra, A.D.; Teodoro, A.J.; Gil, G.A. A Decade of ReseArch. on Coffee as an Anticarcinogenic Beverage. Oxidative Med. Cell. Longev. 2021, 2021, 4420479. [Google Scholar] [CrossRef]
  125. Yang, J.S.; Liu, C.W.; Ma, Y.S.; Weng, S.W.; Tang, N.Y.; Wu, S.H.; Ji, B.C.; Ma, C.Y.; Ko, Y.C.; Funayama, S.; et al. Chlorogenic acid induces apoptotic cell death in U937 leukemia cells through caspase-and mitochondria-dependent pathways. In Vivo 2012, 26, 971–978. [Google Scholar] [PubMed]
  126. Rakshit, S.; Mandal, L.; Pal, B.C.; Bagchi, J.; Biswas, N.; Chaudhuri, J.; Chowdhury, A.A.; Manna, A.; Chaudhuri, U.; Konar, A.; et al. Involvement of ROS in chlorogenic acid-induced apoptosis of Bcr-Abl+ CML cells. Biochem. Pharmacol. 2010, 80, 1662–1675. [Google Scholar] [CrossRef] [PubMed]
  127. Zhang, X.; Huang, J.; Yu, C.; Xiang, L.; Li, L.; Shi, D.; Lin, F. Quercetin enhanced paclitaxel therapeutic effects towards PC-3 prostate cancer through ER stress induction and ros production. OncoTargets Ther. 2020, 13, 513–523. [Google Scholar] [CrossRef] [Green Version]
  128. Wu, Q.; Needs, P.W.; Lu, Y.; Kroon, P.A.; Ren, D.; Yang, X. Different antitumor effects of quercetin, quercetin-3′-sulfate and quercetin-3-glucuronide in human breast cancer MCF-7 cells. Food Funct. 2018, 9, 1736–1746. [Google Scholar] [CrossRef] [PubMed]
  129. Yi, L.; Zongyuan, Y.; Cheng, G.; Lingyun, Z.; Guilian, Y.; Wei, G. Quercetin enhances apoptotic effect of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in ovarian cancer cells through reactive oxygen species (ROS) mediated CCAAT enhancer-binding protein homologous protein (CHOP)-death receptor 5 pathway. Cancer Sci. 2014, 105, 520–527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Wang, I.K.; Lin-Shiau, S.Y.; Lin, J.K. Induction of apoptosis by apigenin and related flavonoids through cytochrome c release and activation of caspase-9 and caspase-3 in leukaemia HL-60 cells. Eur. J. Cancer 1999, 35, 1517–1525. [Google Scholar] [CrossRef]
  131. Safe, S.; Abbruzzese, J.; Abdelrahim, M.; Hedrick, E. Specificity Protein Transcription Factors and Cancer: Opportunities for Drug Development. Cancer Prev. Res. 2018, 11, 371–382. [Google Scholar] [CrossRef]
  132. Pouremamali, F.; Pouremamali, A.; Dadashpour, M.; Soozangar, N.; Jeddi, F. An update of Nrf2 activators and inhibitors in cancer prevention/promotion. Cell Commun. Signal. CCS 2022, 20, 100. [Google Scholar] [CrossRef]
  133. Jenkins, T.; Gouge, J. Nrf2 in Cancer, Detoxifying Enzymes and Cell Death Programs. Antioxidants 2021, 10, 1030. [Google Scholar] [CrossRef]
  134. Islam, S.S.; Qassem, K.; Islam, S.; Parag, R.R.; Rahman, M.Z.; Farhat, W.A.; Yeger, H.; Aboussekhra, A.; Karakas, B.; Noman, A.S.M. Genetic alterations of Keap1 confers chemotherapeutic resistance through functional activation of Nrf2 and Notch pathway in head and neck squamous cell carcinoma. Cell Death Dis. 2022, 13, 696. [Google Scholar] [CrossRef] [PubMed]
  135. Fragoulis, A.; Schenkel, J.; Schröder, N.; Brandt, E.F.; Weiand, M.; Neu, T.; Ramadori, P.; Caspers, T.; Kant, S.; Pufe, T.; et al. Nrf2 induces malignant transformation of hepatic progenitor cells by inducing β-catenin expression. Redox Biol. 2022, 57, 102453. [Google Scholar] [CrossRef] [PubMed]
  136. Yuan, G.; Hu, B.; Ma, J.; Zhang, C.; Xie, H.; Wei, T.; Yang, Y.; Ni, B. Histone lysine methyltransferase SETDB2 suppresses NRF2 to restrict tumor progression and modulates chemotherapy sensitivity in lung adenocarcinoma. Cancer Med. 2022; ahead of print. [Google Scholar] [CrossRef]
  137. Mancini, M.C.S.; Morelli, A.P.; Severino, M.B.; Pavan, I.C.B.; Zambalde, É.P.; Góis, M.M.; Silva, L.G.S.D.; Quintero-Ruiz, N.; Romeiro, C.F.; Dos Santos, D.F.G.; et al. Knockout of NRF2 triggers prostate cancer cells death through ROS modulation and sensitizes to cisplatin. J. Cell Biochem. 2022, 123, 2079–2092. [Google Scholar] [CrossRef] [PubMed]
  138. Wu, X.; Chen, S.; Huang, K.; Lin, G. Triptolide promotes ferroptosis by suppressing Nrf2 to overcome leukemia cell resistance to doxorubicin. Mol. Med. Rep. 2023, 27, 17. [Google Scholar] [CrossRef]
  139. Liu, B.; Wang, H. Oxaliplatin induces ferroptosis and oxidative stress in HT29 colorectal cancer cells by inhibiting the Nrf2 signaling pathway. Exp. Med. 2022, 23, 394. [Google Scholar] [CrossRef]
  140. Wu, S.; Liao, X.; Zhu, Z.; Huang, R.; Chen, M.; Huang, A.; Zhang, J.; Wu, Q.; Wang, J.; Ding, Y. Antioxidant and anti-inflammation effects of dietary phytochemicals: The Nrf2/NF-κB signalling pathway and upstream factors of Nrf2. Phytochemistry 2022, 204, 113429. [Google Scholar] [CrossRef]
  141. Liao, J.C.; Lee, K.T.; You, B.J.; Lee, C.L.; Chang, W.T.; Wu, Y.C.; Lee, H.Z. Raf/ERK/Nrf2 signaling pathway and MMP-7 expression involvement in the trigonelline-mediated inhibition of hepatocarcinoma cell migration. Food Nutr. Res. 2015, 59, 29884. [Google Scholar] [CrossRef] [Green Version]
  142. Pirpour Tazehkand, A.; Salehi, R.; Velaei, K.; Samadi, N. The potential impact of trigonelline loaded micelles on Nrf2 suppression to overcome oxaliplatin resistance in colon cancer cells. Mol. Biol. Rep. 2020, 47, 5817–5829. [Google Scholar] [CrossRef]
  143. Tanveer, M.A.; Rashid, H.; Nazir, L.A.; Archoo, S.; Shahid, N.H.; Ragni, G.; Umar, S.A.; Tasduq, S.A. Trigonelline, a plant derived alkaloid prevents ultraviolet-B-induced oxidative DNA damage in primary human dermal fibroblasts and BALB/c mice via modulation of phosphoinositide 3-kinase-Akt-Nrf2 signalling axis. Exp. Gerontol. 2022, 171, 112028. [Google Scholar] [CrossRef]
  144. Arlt, A.; Sebens, S.; Krebs, S.; Geismann, C.; Grossmann, M.; Kruse, M.L.; Schreiber, S.; Schäfer, H. Inhibition of the Nrf2 transcription factor by the alkaloid trigonelline renders pancreatic cancer cells more susceptible to apoptosis through decreased proteasomal gene expression and proteasome activity. Oncogene 2013, 32, 4825–4835. [Google Scholar] [CrossRef]
  145. Fouzder, C.; Mukhuty, A.; Mukherjee, S.; Malick, C.; Kundu, R. Trigonelline inhibits Nrf2 via EGFR signalling pathway and augments efficacy of Cisplatin and Etoposide in NSCLC cells. Toxicol. In Vitro 2021, 70, 105038. [Google Scholar] [CrossRef] [PubMed]
  146. Chapkin, R.S.; Davidson, L.A.; Park, H.; Jin, U.H.; Fan, Y.Y.; Cheng, Y.; Hensel, M.E.; Landrock, K.K.; Allred, C.; Menon, R.; et al. Role of the Aryl Hydrocarbon Receptor (AhR) in Mediating the Effects of Coffee in the Colon. Mol. Nutr. Food Res. 2021, 65, 2100539. [Google Scholar] [CrossRef]
  147. Serna, E.; Cespedes, C.; Vina, J. Anti-aging physiological roles of aryl hydrocarbon receptor and its dietary regulators. Int. J. Mol. Sci. 2021, 22, 374. [Google Scholar] [CrossRef] [PubMed]
  148. Salminen, A. Aryl hydrocarbon receptor (AhR) reveals evidence of antagonistic pleiotropy in the regulation of the aging process. Cell. Mol. Life Sci. CMLS 2022, 79, 489. [Google Scholar] [CrossRef] [PubMed]
  149. Ojo, E.S.; Tischkau, S.A. The role of ahr in the hallmarks of brain aging: Friend and foe. Cells 2021, 10, 2729. [Google Scholar] [CrossRef]
  150. Nacarino-Palma, A.; Rico-Leo, E.M.; Campisi, J.; Ramanathan, A.; González-Rico, F.J.; Rejano-Gordillo, C.M.; Ordiales-Talavero, A.; Merino, J.M.; Fernández-Salguero, P.M. Aryl hydrocarbon receptor blocks aging-induced senescence in the liver and fibroblast cells. Aging 2022, 14, 4281–4304. [Google Scholar] [CrossRef] [PubMed]
  151. Yang, C.E.; Wang, Y.N.; Hua, M.R.; Miao, H.; Zhao, Y.Y.; Cao, G. Aryl hydrocarbon receptor: From pathogenesis to therapeutic targets in aging-related tissue fibrosis. Ageing Res. Rev. 2022, 79, 101662. [Google Scholar] [CrossRef] [PubMed]
  152. Kawajiri, K.; Kobayashi, Y.; Ohtake, F.; Ikuta, T.; Matsushima, Y.; Mimura, J.; Pettersson, S.; Pollenz, R.S.; Sakaki, T.; Hirokawa, T.; et al. Aryl hydrocarbon receptor suppresses intestinal carcinogenesis in ApcMin/+ mice with natural ligands. Proc. Natl. Acad. Sci. USA 2009, 106, 13481–13486. [Google Scholar] [CrossRef] [Green Version]
  153. Díaz-Díaz, C.J.; Ronnekleiv-Kelly, S.M.; Nukaya, M.; Geiger, P.G.; Balbo, S.; Dator, R.; Megna, B.W.; Carney, P.R.; Bradfield, C.A.; Kennedy, G.D. The Aryl Hydrocarbon Receptor is a Repressor of Inflammation-associated Colorectal Tumorigenesis in Mouse. Ann. Surg. 2016, 264, 429–436. [Google Scholar] [CrossRef] [Green Version]
  154. Han, H.; Davidson, L.A.; Hensel, M.; Yoon, G.; Landrock, K.; Allred, C.; Jayaraman, A.; Ivanov, I.; Safe, S.H.; Chapkin, R.S. Loss of aryl hydrocarbon receptor promotes colon tumorigenesis in ApcS580/þ; KrasG12D/þ mice. Mol. Cancer Res. 2021, 19, 771–783. [Google Scholar] [CrossRef]
  155. Han, H.; Davidson, L.A.; Fan, Y.; Goldsby, J.S.; Yoon, G.; Jin, U.; Wright, G.A.; Landrock, K.K.; Weeks, B.R.; Wright, R.C.; et al. Loss of aryl hydrocarbon receptor potentiates FoxM1 signaling to enhance self-renewal of colonic stem and progenitor cells. EMBO J. 2020, 39, e104319. [Google Scholar] [CrossRef] [PubMed]
  156. Yang, Y.; Osorio, D.; Davidson, L.A.; Han, H.; Mullens, D.A.; Jayaraman, A.; Safe, S.; Ivanov, I.; Cai, J.J.; Chapkin, R.S. Single-cell RNA Sequencing Reveals How the Aryl Hydrocarbon Receptor Shapes Cellular Differentiation Potency in the Mouse Colon. Cancer Prev. Res. 2022, 15, 17–28. [Google Scholar] [CrossRef] [PubMed]
  157. Han, H.; Davidson, L.A.; Fan, Y.Y.; Landrock, K.K.; Jayaraman, A.; Safe, S.H.; Chapkin, R.S. Loss of aryl hydrocarbon receptor suppresses the response of colonic epithelial cells to IL22 signaling by upregulating SOCS3. Am. J. Physiol.-Gastrointest. Liver Physiol. 2022, 322, G93–G106. [Google Scholar] [CrossRef] [PubMed]
  158. Kolluri, S.K.; Jin, U.H.; Safe, S. Role of the aryl hydrocarbon receptor in carcinogenesis and potential as an anti-cancer drug target. Arch. Toxicol. 2017, 91, 2497–2513. [Google Scholar] [CrossRef]
  159. DiNatale, B.C.; Smith, K.; John, K.; Krishnegowda, G.; Amin, S.G.; Perdew, G.H. Ah receptor antagonism represses head and neck tumor cell aggressive phenotype. Mol. Cancer Res. 2012, 10, 1369–1379. [Google Scholar] [CrossRef] [Green Version]
  160. Pearen, M.A.; Muscat, G.E.O. Minireview: Nuclear hormone receptor 4A signaling: Implications for metabolic disease. Mol. Endocrinol. 2010, 24, 1891–1903. [Google Scholar] [CrossRef] [Green Version]
  161. Safe, S.; Jin, U.H.; Hedrick, E.; Reeder, A.; Lee, S.O. Minireview: Role of orphan nuclear receptors in cancer and potential as drug targets. Mol. Endocrinol. 2014, 28, 157–172. [Google Scholar] [CrossRef]
  162. Safe, S.; Karki, K. The paradoxical roles of orphan nuclear receptor 4A (NR4A) in cancer. Mol. Cancer Res. 2021, 19, 180–191. [Google Scholar] [CrossRef]
  163. Paillasse, M.R.; de Medina, P. The NR4A nuclear receptors as potential targets for anti-aging interventions. Med. Hypotheses 2015, 84, 135–140. [Google Scholar] [CrossRef]
  164. Asami, T.; Endo, K.; Matsui, R.; Sawa, T.; Tanaka, Y.; Saiki, T.; Tanba, N.; Haga, H.; Tanaka, S. Long-term caloric restriction ameliorates T cell immunosenescence in mice. Mech. Ageing Dev. 2022, 206, 111710. [Google Scholar] [CrossRef]
  165. Ma, G.; Chen, F.; Liu, Y.; Zheng, L.; Jiang, X.; Tian, H.; Wang, X.; Song, X.; Yu, Y.; Wang, D. Nur77 ameliorates age-related renal tubulointerstitial fibrosis by suppressing the TGF-β/Smads signaling pathway. FASEB J. 2022, 36, e22124. [Google Scholar] [CrossRef] [PubMed]
  166. Chatterjee, S.; Walsh, E.N.; Yan, A.L.; Giese, K.P.; Safe, S.; Abel, T. Pharmacological activation of Nr4a rescues age-associated memory decline. Neurobiol. Aging 2020, 85, 140–144. [Google Scholar] [CrossRef] [PubMed]
  167. Kwapis, J.L.; Alaghband, Y.; López, A.J.; Long, J.M.; Li, X.; Shu, G.; Bodinayake, K.K.; Matheos, D.P.; Rapp, P.R.; Wood, M.A. HDAC3-mediated repression of the Nr4a family contributes to age-related impairments in long-term memory. J. Neurosci. 2019, 39, 4999–5009. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Shrestha, R.; Mohankumar, K.; Martin, G.; Hailemariam, A.; Lee, S.O.; Jin, U.H.; Burghardt, R.; Safe, S. Flavonoids kaempferol and quercetin are nuclear receptor 4A1 (NR4A1, Nur77) ligands and inhibit rhabdomyosarcoma cell and tumor growth. J. Exp. Clin. Cancer Res. 2021, 40, 392. [Google Scholar] [CrossRef]
  169. Karabegović, I.; Portilla-Fernandez, E.; Li, Y.; Ma, J.; Maas, S.C.E.; Sun, D.; Hu, E.A.; Kühnel, B.; Zhang, Y.; Ambatipudi, S.; et al. Epigenome-wide association meta-analysis of DNA methylation with coffee and tea consumption. Nat. Commun. 2021, 12, 2830. [Google Scholar] [CrossRef] [PubMed]
  170. Thomas, S.; Izard, J.; Walsh, E.; Batich, K.; Chongsathidkiet, P.; Clarke, G.; Sela, D.A.; Muller, A.J.; Mullin, J.M.; Albert, K.; et al. The host microbiome regulates and maintains human health: A primer and perspective for non-microbiologists. Cancer Res. 2017, 77, 1783–1812. [Google Scholar] [CrossRef] [Green Version]
  171. Taylor, S.R.; Falcone, J.N.; Cantley, L.C.; Goncalves, M.D. Developing dietary interventions as therapy for cancer. Nat. Rev. Cancer 2022, 22, 452–466. [Google Scholar] [CrossRef]
  172. Chen, L.; Wang, D.; Garmaeva, S.; Kurilshikov, A.; Vich Vila, A.; Gacesa, R.; Sinha, T.; Segal, E.; Weersma, R.K.; Wijmenga, C.; et al. The long-term genetic stability and individual specificity of the human gut microbiome. Cell 2021, 184, 2302–2315.e12. [Google Scholar] [CrossRef]
  173. de Vos, W.M.; Tilg, H.; van Hul, M.; Cani, P.D. Gut microbiome and health: Mechanistic insights. Gut 2022, 71, 1020–1032. [Google Scholar] [CrossRef]
  174. Song, Z.; Liu, L.; Xu, Y.; Cao, R.; Lan, X.; Pan, C.; Zhang, S.; Zhao, H. Caffeine-Induced Sleep Restriction Alters the Gut Microbiome and Fecal Metabolic Profiles in Mice. Int. J. Mol. Sci. 2022, 23, 14837. [Google Scholar] [CrossRef] [PubMed]
  175. Gniechwitz, D.; Reichardt, N.; Blaut, M.; Steinhart, H.; Bunzel, M. Dietary fiber from coffee beverage: Degradation by human fecal microbiota. J. Agric. Food Chem. 2007, 55, 6989–6996. [Google Scholar] [CrossRef] [PubMed]
  176. Reichardt, N.; Gniechwitz, D.; Steinhart, H.; Bunzel, M.; Blaut, M. Characterization of high molecular weight coffee fractions and their fermentation by human intestinal microbiota. Mol. Nutr. Food Res. 2009, 53, 287–299. [Google Scholar] [CrossRef] [PubMed]
  177. González, S.; Salazar, N.; Ruiz-Saavedra, S.; Gómez-Martín, M.; de Los Reyes-Gavilán, C.G.; Gueimonde, M. Long-Term Coffee Consumption is Associated with Fecal Microbial Composition in Humans. Nutrients 2020, 12, 1287. [Google Scholar] [CrossRef] [PubMed]
  178. van der Beek, C.M.; Dejong, C.H.C.; Troost, F.J.; Masclee, A.A.M.; Lenaerts, K. Role of short-chain fatty acids in colonic inflammation, carcinogenesis, and mucosal protection and healing. Nutr. Rev. 2017, 75, 286–305. [Google Scholar] [CrossRef] [Green Version]
  179. Nowotny, B.; Zahiragic, L.; Bierwagen, A.; Kabisch, S.; Groener, J.B.; Nowotny, P.J.; Fleitmann, A.K.; Herder, C.; Pacini, G.; Erlund, I.; et al. Low-energy diets differing in fibre, red meat and coffee intake equally improve insulin sensitivity in type 2 diabetes: A randomised feasibility trial. Diabetologia 2015, 58, 255–264. [Google Scholar] [CrossRef] [Green Version]
  180. Iljazovic, A.; Roy, U.; Gálvez, E.J.C.; Lesker, T.R.; Zhao, B.; Gronow, A.; Amend, L.; Will, S.E.; Hofmann, J.D.; Pils, M.C.; et al. Perturbation of the gut microbiome by Prevotella spp. enhances host susceptibility to mucosal inflammation. Mucosal Immunol. 2021, 14, 113–124. [Google Scholar] [CrossRef]
  181. Jaquet, M.; Rochat, I.; Moulin, J.; Cavin, C.; Bibiloni, R. Impact of coffee consumption on the gut microbiota: A human volunteer study. Int. J. Food Microbiol. 2009, 130, 117–121. [Google Scholar] [CrossRef]
  182. Mills, C.E.; Tzounis, X.; Oruna-Concha, M.J.; Mottram, D.S.; Gibson, G.R.; Spencer, J.P.E. In vitro colonic metabolism of coffee and chlorogenic acid results in selective changes in human faecal microbiota growth. Br. J. Nutr. 2015, 113, 1220–1227. [Google Scholar] [CrossRef] [Green Version]
  183. Moco, S.; Martin, F.P.J.; Rezzi, S. Metabolomics view on gut microbiome modulation by polyphenol-rich foods. J. Proteome Res. 2012, 11, 4781–4790. [Google Scholar] [CrossRef]
  184. Qin, J.; Li, R.; Raes, J.; Arumugam, M.; Burgdorf, K.S.; Manichanh, C.; Nielsen, T.; Pons, N.; Levenez, F.; Yamada, T.; et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 2010, 464, 59–65. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Structures of individual compounds in roasted coffee after extraction with hot water.
Figure 1. Structures of individual compounds in roasted coffee after extraction with hot water.
Ijms 24 02706 g001
Figure 2. Development of intracellular oxidative stress is attenuated by coffee-induced stress. Coffee-induced stress results in the dissociation of NRF2 from KEAP and the subsequent translocation of NRF2 into the nucleus where the NRF2-sMAF complex binds cis-acting AREs to induce NRF2 regulated genes/pathways, including the antioxidant enzymes glutathione peroxidase (GPx), superoxide dismutase (SOD), quinone oxidoreductase 1 (NQO1), glutathione reductase (GR), harm oxygenase 1 (HO-1), and several thioreductases family members [104,105,106,107].
Figure 2. Development of intracellular oxidative stress is attenuated by coffee-induced stress. Coffee-induced stress results in the dissociation of NRF2 from KEAP and the subsequent translocation of NRF2 into the nucleus where the NRF2-sMAF complex binds cis-acting AREs to induce NRF2 regulated genes/pathways, including the antioxidant enzymes glutathione peroxidase (GPx), superoxide dismutase (SOD), quinone oxidoreductase 1 (NQO1), glutathione reductase (GR), harm oxygenase 1 (HO-1), and several thioreductases family members [104,105,106,107].
Ijms 24 02706 g002
Figure 3. Coffee extracts induce ROS. Treatment of cancer cells with ROS inducers, such as chlorogenic acids and quercetin, inhibits redox enzymes and decreases mitochondrial membrane potential (MMP) to induce ROS, which downregulates cMyc + cMyc-regulated microRNAs (miRs: miR 17-92/27a). This results in the induction of ZBTB genes (ZBTB4, ZBTB10) + inhibition of pro-oncogenic Sp1/Sp3/Sp4-regulated genes/pathways [127,128,129,130,131,132,133].
Figure 3. Coffee extracts induce ROS. Treatment of cancer cells with ROS inducers, such as chlorogenic acids and quercetin, inhibits redox enzymes and decreases mitochondrial membrane potential (MMP) to induce ROS, which downregulates cMyc + cMyc-regulated microRNAs (miRs: miR 17-92/27a). This results in the induction of ZBTB genes (ZBTB4, ZBTB10) + inhibition of pro-oncogenic Sp1/Sp3/Sp4-regulated genes/pathways [127,128,129,130,131,132,133].
Ijms 24 02706 g003
Figure 4. Coffee receptor-mediated responses: coffee extracts that bind AhR (A) or NR4A1 (B) may activate age- and cell-context-dependent chemopreventive and/or chemotherapeutic responses [156,157,158,159,160,161].
Figure 4. Coffee receptor-mediated responses: coffee extracts that bind AhR (A) or NR4A1 (B) may activate age- and cell-context-dependent chemopreventive and/or chemotherapeutic responses [156,157,158,159,160,161].
Ijms 24 02706 g004
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

Safe, S.; Kothari, J.; Hailemariam, A.; Upadhyay, S.; Davidson, L.A.; Chapkin, R.S. Health Benefits of Coffee Consumption for Cancer and Other Diseases and Mechanisms of Action. Int. J. Mol. Sci. 2023, 24, 2706. https://doi.org/10.3390/ijms24032706

AMA Style

Safe S, Kothari J, Hailemariam A, Upadhyay S, Davidson LA, Chapkin RS. Health Benefits of Coffee Consumption for Cancer and Other Diseases and Mechanisms of Action. International Journal of Molecular Sciences. 2023; 24(3):2706. https://doi.org/10.3390/ijms24032706

Chicago/Turabian Style

Safe, Stephen, Jainish Kothari, Amanuel Hailemariam, Srijana Upadhyay, Laurie A. Davidson, and Robert S. Chapkin. 2023. "Health Benefits of Coffee Consumption for Cancer and Other Diseases and Mechanisms of Action" International Journal of Molecular Sciences 24, no. 3: 2706. https://doi.org/10.3390/ijms24032706

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

Article Metrics

Back to TopTop