Skip to main content
Evidence-based Complementary and Alternative Medicine : eCAM logoLink to Evidence-based Complementary and Alternative Medicine : eCAM
. 2015 Feb 9;2015:593902. doi: 10.1155/2015/593902

Pharmacological Properties of Protocatechuic Acid and Its Potential Roles as Complementary Medicine

Yoswaris Semaming 1,2, Patchareewan Pannengpetch 3, Siriporn C Chattipakorn 2,4,5, Nipon Chattipakorn 2,5,6,*
PMCID: PMC4337037  PMID: 25737736

Abstract

This paper reviews the reported pharmacological properties of protocatechuic acid (PCA, 3,4-dihydroxy benzoic acid), a type of phenolic acid found in many food plants such as olives and white grapes. PCA is a major metabolite of anthocyanin. The pharmacological actions of PCA have been shown to include strong in vitro and in vivo antioxidant activity. In in vivo experiments using rats and mice, PCA has been shown to exert anti-inflammatory as well as antihyperglycemic and antiapoptotic activities. Furthermore, PCA has been shown to inhibit chemical carcinogenesis and exert proapoptotic and antiproliferative effects in different cancerous tissues. Moreover, in vitro studies have shown PCA to have antimicrobial activities and also to exert synergistic interaction with some antibiotics against resistant pathogens. This review aims to comprehensively summarize the pharmacological properties of PCA reported to date with an emphasis on its biological properties and mechanisms of action which could be therapeutically useful in a clinical setting.

1. Introduction

Protocatechuic acid (PCA, 3,4-dihydroxybenzoic acid) is a phenolic compound found in many food plants such as Olea europaea (olives), Hibiscus sabdariffa (roselle), Eucommia ulmoides (du-zhong), Citrus microcarpa Bunge (calamondin), and Vitis vinifera (white wine grapes) [13]. PCA content varies considerably depending on the type of food.

Recently, several investigations have shown that PCA is a major metabolite of complex polyphenols, especially anthocyanins [4, 5]. Anthocyanins have been shown to affect a variety of physiological activities which are of great benefit to health, including a reduced risk of cardiovascular diseases. This particular beneficial effect is partly due to the anti-inflammatory properties [68], antioxidant and free radical scavenging activities [912], peroxidation inhibition [13], and estrogenic/antiestrogenic activity [14] of PCA. PCA is of particular nutritional interest since it is a main anthocyanin metabolite that can reach tissues in amounts which can exert biological effects on health [15]. In vivo studies demonstrated that male balb/cA mice which were fed a standard diet supplemented with PCA for 12 weeks showed increased PCA levels in plasma and tissues such as brain, heart, liver, and kidney [16]. Moreover, PCA itself has been shown to possess antioxidant properties as well as having other potential health benefits such as anti-inflammatory effects.

The aim of this review is to comprehensively summarize the pharmacological properties of PCA reported to date including antioxidant, anti-inflammatory, antihyperglycemia, antiapoptosis/proapoptosis, and antimicrobial activities, with an emphasis on the biological properties and mechanisms of action which could be potentially useful in a clinical setting.

2. Antioxidant Activity of PCA

Oxidative stress plays a key role in the pathogenesis of degenerative diseases such as cardiovascular diseases, diabetes mellitus, neurodegenerative diseases, cancer, and aging [1721]. Mounting evidence from both in vitro and in vivo studies demonstrates that PCA exerts potent antioxidative effects. In in vitro studies, as summarized in Table 1, PCA was shown to have free radical scavenging and antioxidant activities by decreasing lipid peroxidation and increasing the scavenging of hydrogen peroxide (H2O2) and diphenylpicrylhydrazyl (DPPH) [22]. In J77A.1 macrophage, PCA decreased oxidized low-density lipoprotein levels (LDL), inhibited superoxide (O2 ) and H2O2 production, and also restored glutathione (GSH) related enzymes via c-Jun N-terminal kinase (JNK) mediated nuclear factor (erythroid-derived 2) like 2 (Nrf2) activation [23, 24]. PCA also reduced reactive oxygen species (ROS) induced apoptosis by improving mitochondrial function, inhibiting DNA fragmentation in H2O2-induced oxidative stress in human neuronal cells [25], preventing lactate dehydrogenase (LDH) release in H2O2-induced oxidative stress in PC12 cells [26], and inhibiting intracellular ROS level in BNLCL2 cells [27].

Table 1.

Summary of in vitro studies of antioxidant activities of PCA.

Model Method PCA concentration Major finding Interpretation Reference
Biochemical assay (i) TBAR assay
(ii) H2O2 assay
(iii) DPPH. assay
0.05 and 0.10 mg/mL (i) PCA increased % inhibition of lipid peroxidation
(ii) PCA increased % scavenging of H2O2
(iii) PCA increased % scavenging of DPPH.
PCA exerted antioxidant activity [22]

J774 A.1 macrophages J774 A.1 macrophages 3 and 25 mol/L (i) PCA decreased oxidation of LDL
(ii) PCA inhibited O2 and H2O2 production
(iii) PCA increased GSH content
(iv) PCA restored GR and GPx activities
(v) PCA restored the γ-GCS mRNA, GR, and GPx expression
PCA had an antioxidant activity via activation of mRNA transcription of GSH-related enzymes [23]

J774 A.1 macrophages Direct PCA application to cells 25 μM (i) PCA increased GSH, GPx, and GR expression
(ii) PCA increased Nrf2 expression and activation
(iii) PCA increased JNK mRNA level
PCA increased macrophage endogenous antioxidants via JNK-mediated Nrf2 activation [24]

Human neuronal cell line H2O2-induced oxidative stress 25, 50, and 100 μM (i) PCA inhibited ROS formation at cytosolic level
(ii) PCA inhibited apoptotic events
(iii) PCA improved mitochondrial function
(iv) PCA decreased DNA fragmentation
PCA reduced apoptosis via ROS reduction, improved mitochondrial function, and inhibited DNA fragmentation [25]

PC12 cells H2O2-induced oxidative damage 50, 100, 150, and 200 μM (i) PCA increased cell viability
(ii) PCA decreased % LDH release
PCA prevented H2O2-induced cell death [26]

BNLCL2 cells H2O2-induced oxidative damage 1, 5, 10, 20, and 100 μg/mL (i) PCA affected DPPH scavenging activity
(ii) PCA inhibited liposome peroxidation
(iii) PCA reduced intracellular ROS level
PCA had a radical scavenging activity and antioxidant property [27]

Consistent with in vitro reports, in vivo studies (as summarized in Table 2) also demonstrated that PCA treatment decreased oxidative stress by promoting endogenous antioxidant enzymes in aging rats and also reduced H2O2-induced oxidative damage in aging mice, thus indicating that PCA could prevent oxidative damage in aging animals [26, 28]. PCA also decreased advance glycation end products (AGEs) and ROS production in D-galactose-induced ROS and AGEs formation in mice [29]. In streptozotocin (STZ) induced diabetic rats, PCA was also found to decrease ROS formation in liver, heart, kidney, and brain by restoring endogenous antioxidant enzyme activities [3, 30]. All of these findings indicated that the PCA possess potential antioxidant activity, suggesting that it could be used as a complementary medication to prevent oxidative damage in various degenerative diseases.

Table 2.

Summary of in vivo studies of antioxidant activities of PCA.

Model Method PCA dose/route/duration Major finding Interpretation Reference
Sprague-Dawley rat STZ-induced T1DM (50 mg/kg, ip) 50, 100 mg/kg, po (i) PCA decreased plasma MDA
(ii) PCA decreased cardiac MDA
(iii) PCA decreased mitochondrial ROS production
PCA deceased oxidative stress in T1DM rats [30]

Sprague-Dawley rat H2O2-induced oxidative damage in young and age rats 5 mg/kg/day for 7 days (ip) (i) PCA improved scores during the passive avoidance testing
(ii) PCA decreased MDA in brain of aged rat
(iii) PCA increased GSH-PX activity
PCA promoted endogenous antioxidant enzymatic activities and inhibited ROS generation [26]

Mice D-galactose-induced ROS and AGEs 0.5%, 1%, or 2% in diet for 8 weeks (i) PCA decreased ROS and protein carbonyl content
(ii) PCA retained GSH content
(iii) PCA decreased CML, pentosidine, sorbitol, fructose, and methylglycoxal level in brain
PCA had antiglycative and antioxidant activity by retaining GSH  [29]

Mice Young and aged 5 and 10 mg/kg (ip) for 7 days In aged rats
  (i) PCA elevated splenic weight
  (ii) PCA increased the activities of GSH-PX
  (iii) PCA increased catalase (CAT) activity
  (iv) PCA decreased malondialdehyde (MDA) level
PCA was a potential antiageing agent by promoting endogenous antioxidant enzymatic activities [28]

Mice STZ-induced DM (50 mg/kg/iv) 1%, 2%, and 4% in diet for 8 weeks (i) PCA at all concentrations decreased cardiac and renal MDA level
(ii) PCA at 2% and 4% increased cardiac and renal GSH level
(iii) PCA at 2% and 4% decreased cardiac and renal GSSG formation
(iv) PCA at 2% and 4% increased GPX and catalase activity in cardiac and renal tissues
PCA had an antioxidative effect through the restoration of endogenous antioxidants [3]

3. Anti-Inflammatory Activity of PCA

The inflammatory process is regulated by coordinated activation of both pro- and anti-inflammatory mediators in tissue cells (such as fibroblasts, endothelial cells, tissue macrophages, and mast cells) and also by the recruitment of leucocytes [31, 32]. Prolonged activation of proinflammatory mediators causes tissue injury and organ dysfunction. As a consequence, chronic inflammation plays a critical role in the pathophysiology of major chronic diseases including obesity, cardiovascular disease, diabetes mellitus, Alzheimer's disease, and many types of cancer [33, 34]. The mediators, including nitric oxide (NO), lipid mediators, cytokines/chemokines, adhesion molecules, and matrix metalloproteinases (MMPs), are involved in the initiation, maintenance, and resolution of the inflammatory process [35, 36].

A summary of in vitro studies regarding the effects of PCA on the inflammatory process is shown in Table 3. PCA was shown to suppress tumor necrosis factor alpha (TNF-α), interleukin- (IL-) 1β, inducible nitric oxide synthase (iNOS), and cyclooxygenase 2 (COX-2) expression via the regulation of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB) and mitogen-activated protein kinase (MAPK) activation in lipopolysaccharide- (LPS-) induced RAW 264.7 cell damage [37]. Moreover, PCA also suppressed vascular cell adhesion protein 1 (VCAM-1) and intercellular adhesion molecule 1 (ICAM-1) mRNA expression in TNF-α-induced cellular damage [38] and inhibited monocyte infiltration [39].

Table 3.

Summary of in vitro studies of anti-inflammatory activities of PCA.

Model Method PCA concentration Major finding Interpretation Reference
RAW 264.7 cells Lipopolysaccharide- (LPS-) induced cellular damage 1, 2, 5, and 25 μM (i) PCA decreased TNF-α and IL-1β
(ii) PCA decreased NO and PGE2
(iii) PCA inhibited iNOS and COX-2 expression
(iv) PCA inhibited IkB-α degradation
(v) PCA inhibited NF-kB phosphorylation
(vi) PCA inhibited p38, ERK, and JNK
PCA had anti-inflammatory effects by regulating NF-kB and MAPK activation [37]

Mouse aortic endothelial cell (MAEC) TNF-α-induced cellular damage 0.05, 0.5, 5.0, 10, 20, and 40 μmol/L (i) PCA inhibited adhesion of HL-60 cells to MAECs
(ii) PCA suppressed VCAM-1 and ICAM-1 mRNA expression
(iii) PCA reduced NF-kB activation
PCA had an anti-inflammatory effect by inhibiting monocyte adhesion molecules [38]

Cell culture Isolated peripheral blood monocytes (PBMs) from ApoE-deficient mice 0.125, 0.25, and 0.5 μmol/L (i) PCA decreased CCR2 protein and mRNA expression
(ii) PCA inhibited mouse PBMs migration
PCA exerted antiatherogenic properties by inhibiting monocyte infiltration [39]

Consistent with in vitro reports, animal studies (Table 4) demonstrated that PCA strongly inhibited inflammation by inhibiting carrageenan-induced inflammation in mice by decreasing TNF-α, IL-1β, and prostaglandin E2 (PGE2) levels, suppressed iNOS and COX-2 expression in apolipoprotein E (ApoE) deficient mice [37], prevented LPS-induced sepsis in mice via decreased NO levels and suppressed IL-10 [40], reduced VCAM-1 and ICAM-1 [38], and inhibited monocyte/macrophage infiltration in mice [39]. Moreover, PCA also prevented coagulation and inflammation in STZ-induced diabetic rats by inhibiting the plasma levels of the plasminogen activator inhibitor 1 (PAI-1), antithrombin III (AT-III), protein C, C-reactive protein (CRP), and von Willebrand factor (vWF) and reduced IL-6, TNF-α, and monocyte chemoattractant protein-1 (MCP-1) levels in the heart and kidneys [3]. These findings suggest that the anti-inflammatory effects of PCA might be beneficial in various chronic degenerative diseases in which the inflammatory process plays an important part in the pathogenesis.

Table 4.

Summary of in vivo studies of anti-inflammatory activities of PCA.

Model Method PCA dose/route/duration Major finding Interpretation Reference
Mice Carrageenan-induced inflammation in BALB/c mice 5 and 25 mg/kg, po (24 h) (i) PCA reduced exudate
(ii) PCA decreased protein content
(iii) PCA reduced leukocyte number
(iv) PCA inhibited TNF-α, IL-1β, and PGE2 level
(v) PCA inhibited COX-2 and NF-kB expression
PCA exerted anti-inflammatory effects by inhibiting NF-kB activation. [37]

Mice ApoE-deficient mice 0.033% (w/w) of diet for 20 weeks (i) PCA reduced sinus plague area
(ii) PCA decreased cholesterol accumulation in aortas
(iii) PCA reduced VCAM-1 and ICAM-1 expression in aortas
(iv) PCA reduced NF-kB binding activity
(v) PCA reduced plasma-soluble VCAM-1 and ICAM-1 levels
PCA exerted antiatherosclerosis effects by inhibiting adhesion molecules and reducing NF-kB activation [38]

Mice Thioglycollate-induced peritonitis in ApoE-deficient mice 25 mg/kg (po) for 11 days (i) PCA decreased CCR2 protein and mRNA expression in PBMs of ApoE-deficient mice
(ii) PCA reduced macrophage infiltration into the abdominal cavity
PCA exerted antiatherogenic properties by inhibiting monocyte/macrophage infiltration [39]

Mice LPS-induced sepsis (20 mg/kg, ip) 50 mg/kg (ip) single dose (i) PCA reduced lethality
(ii) PCA suppressed TNF-α and IL-10
(iii) PCA decreased plasma ALT levels
(iv) PCA decreased plasma nitrite/nitrate levels
(v) PCA decreased hepatic MDA levels
PCA exerted sepsis prevention properties by inhibiting inflammatory cytokines and antioxidant activity [40]

Mice STZ-induced DM
(50 mg/kg/iv)
1%, 2%, and 4% in diet for 8 weeks (i) PCA lowered plasma PAI-1 levels
(ii) PCA increased plasma AP-III levels
(iii) PCA increased plasma protein C levels
(iv) PCA lowered plasma CRP levels
(v) PCA decreased plasma von Willebrand factor
(vi) PCA reduced IL-6, TNF-α, and MCP-1 levels in heart and kidney
PCA exerted anticoagulatory and anti-inflammatory effects by lowering inflammatory cytokines [3]

4. Antihyperglycemic Activity of PCA

Maintenance of glucose homeostasis by strict hormonal control is of the utmost importance to human physiology [41, 42]. Failure of the control of glucose levels, with defects in both insulin action and insulin secretion, can result in a metabolic syndrome which is a multisymptom disorder of energy homeostasis [43]. It has been demonstrated that peroxisome proliferator-activated receptor gamma (PPARγ) is one of several targets of insulin activity, which regulates the expression and activity of key players in the maintenance of glucose transport machinery efficiency, such as glucose transporter (GLUT) 4 and adiponectin [44, 45]. In in vitro studies, as summarized in Table 5, PCA has been shown to exert an insulin-like activity in oxidized LDL-induced insulin resistance in adipocytes via increased PPARγ activation [45]. Similarly, in vivo studies (Table 6) also demonstrated that PCA decreased blood glucose levels in STZ-induced diabetes via restored carbohydrate metabolic enzyme activity, increased plasma insulin level, and normalized the activity of pancreatic islets [3, 30, 46]. These findings suggest that PCA provides antihyperglycemic effects in addition to its reported antioxidant and anti-inflammatory effects.

Table 5.

Summary of in vitro study of antihyperglycemic activities of PCA.

Model Model/method PCA concentration Major finding Interpretation Reference
Human omental adipocytes and murine adipocyte 3T3-L1 cells oxLDL-induced insulin resistance 100 μM (i) PCA increased glucose uptake
(ii) PCA increased GLUT4 translocation
(iii) PCA increased PPARγ activity
(iv) PCA increased adiponectin
PCA exerted an insulin-like activity in adipocytes by increasing PPARγ activation [45]

Table 6.

Summary of in vivo studies of antihyperglycemic activities of PCA.

Model Model/method PCA dose/route/duration Major finding Interpretation Reference
Sprague-Dawley rat STZ-induced T1DM (50 mg/kg, ip) 50, 100 mg/kg (po) (i) PCA decreased FBG
(ii) PCA decreased HbA1c
PCA exerted hypoglycemic effects in T1DM [30]

Mice STZ-induced DM (50 mg/kg, iv) 1%, 2%, and 4% in diet for 8 weeks (i) PCA lowered plasma glucose levels
(ii) PCA increased insulin levels
(iii) PCA decreased TG and TC content in plasma, heart, and liver
PCA attenuated diabetic conditions by lowering plasma glucose, increasing insulin, and lowering triglyceride levels [3]

Sprague-Dawley rat STZ-induced DM (40 mg/kg, ip) 50, 100, 200 mg/
kg/day (po) for 45 days
(i) PCA decreased plasma glucose levels
(ii) PCA decreased HbA1c levels
(iii) PCA increased plasma insulin levels
(iv) PCA increased hexokinase activity and increased glycogen content in liver
(v) PCA decreased activity of glucose 6-phosphatase and fructose 1,6-bisphosphatase in liver
(vi) PCA reduced adipose tissue of DM pancreas and normalized pancreatic islets
PCA exerted antihyperglycemic effects by restoring carbohydrate metabolic enzyme activity and increasing plasma insulin levels [46]

Mice STZ-induced DM (50 mg/kg, iv) 2% and 4% in diet for 12 weeks (i) Content of PCA increased in plasma, brain, heart, liver, and kidney
(ii) PCA decreased water intake and food intake
(iii) PCA increased body weight
(iv) PCA decreased urine volume
(v) PCA reduced plasma glucose levels
(vi) PCA increased plasma insulin levels
(vii) PCA decreased plasma BUN level
(viii) PCA increased creatinine clearance rate
(ix) PCA decreased HbA1C level 
(x) PCA decreased urine glycated albumin
(xi) PCA reduced renal production of CML, pentosidine, sorbitol, and fructose
(xii) PCA decreased brain content of CML, pentosidine, fructose, and sorbitol
(xiii) PCA decreased urinary albumin
(xiv) PCA reduced level of fibronectin, type-IV collagen, and TGF-β in renal tissue 
(xv) PCA reduced renal activity and expression of AR and SDH
(xvi) PCA increased renal activity and expression of GLI
(xvii) PCA decreased renal activity and mRNA expression of PKC-α and PKC-β
(xviii) PCA decreased RAGE mRNA expression
PCA had an antihyperglycemic, antiglycative and renoprotective effects via increasing plasma insulin, reducing plasma glucose, reducing renal level of glycation end products, fibronectin, TGF-β, and repressing renal activity and expression of AR, SDH, GLI, PKC-α, PPAR-γ, restoring PPAR-γ, and suppressing RAGE [16]

5. Antiapoptosis versus Proapoptotic Activity of PCA

Polyphenols have been shown to improve cell survival and protect against cytotoxicity by inhibiting apoptosis [18]. However, they can also induce apoptosis and prevent tumor growth [47, 48]. These opposite effects are mainly due to its effects on the controlling of the cell redox state. Evidence from in vitro studies (Table 7) revealed that PCA has cell-protective effects via increased IkB degradation and subsequent NF-kB activation in TNF-α-induced cell death [49], attenuated changes of the mitochondrial membrane permeability, decreased oxidative stress damage and increased Bcl-2 levels in 1-methyl-4-phenylpyridinium- (MPP+-) induced apoptotic cell death [50], decreased caspase-3 activity in isolated neuronal stem cells (NSCs) [51], and reduced LDH leakage in H2O2-induced apoptosis [52]. In MPP+-induced cell death, PCA treatment resulted in a return to normal cellular morphology and normal mitochondria [53]. Moreover, PCA has been shown to have cell-protective effects via antioxidant and scavenging activities [54].

Table 7.

Summary of in vitro studies of antiapoptotic activities of PCA in noncancer cells.

Model Model/method PCA concentration Major finding Interpretation Reference
HUVECs and Jurkat cells TNF-α-induced cell death 50, 100 µM and 1 nM (i) PCA inhibited TNF-α-induced HUVECs and Jurkat cells death
(ii) PCA increased NF-kB activation
(iii) PCA increased degradation of IkB
PCA exerted cell-protective effects via increased IkB degradation and subsequent NF-kB activation [49]

PC12 cells MPP+-induced mitochondrial dysfunction and apoptotic cell death 0.3, 0.6, and 1.2 mM (i) PCA reduced the number of cell death
(ii) PCA at 0.6 and 1.2 mM decreased percentage of depolarized cell, reduced ROS formation, and increased GSH content
(iii) PCA at 0.6 and 1.2 mM decreased caspase-3 activity and increased Bcl-2 protein
PCA exerted antiapoptotic activities via attenuated changes of mitochondrial membrane permeability and decreased oxidative stress damage [50]

Isolated NSCs of embryonic rat Direct PCA application to cells 0.006, 0.03, 0.06, and 0.12 mM (i) PCA at 0.03, 0.06, and 0.12 mM increased cellular viability
(ii) PCA reduced nuclear fragmentation
(iii) PCA reduced the levels of apoptosis
(iv) PCA decreased endogenous ROS level
(v) PCA decreased caspase-3 activity
PCA inhibited cell apoptosis via suppression of the caspase cascade [51]

PC12 cells H2O2-induced apoptosis 0.006, 0.03, 0.06, and 0.12 mM (i) PCA (over 0.3 mM) increased cellular viability
(ii) PCA reduced LDH leakage
(iii) PCA reduced apoptotic sub-G1 population
PCA promoted cell viability and inhibited apoptotic cell death [52]

PC12 cells MPP+-induced apoptotic cell death 0.33, 0.65, and 1.30 mM (i) PCA reduced cell death in a dose-dependent manner
(ii) PCA treatment exhibited normal cellular morphology and normal mitochondria
(iii) PCA increased tyrosine hydroxylase expression
(iv) PCA reduced oligomeric α-synuclein
(v) PCA increased monomeric α-synuclein
PCA had neuroprotective effects via reducing cell death and inhibiting oligomerization of α-synuclein [53]

Rat primary hepatocytes t-BHP (1.5 mM) induced oxidative damage 0.02, 0.05, and 0.10 mg/mL (i) PCA 0.05 and 0.10 mg/mL decreased LDH, ALT, and MDA
(ii) PCA prevented mitochondrial depolarization
(iii) PCA increased scavenging activity on DPPH
PCA had a cell-protective effect via its antioxidant and scavenging activity [54]

Unlike the cells described in Table 7, evidence from cancer cell studies (Table 8) demonstrated that PCA can induce apoptosis and prevent the growth of tumor cells via causing reduced Bcl-2 protein, increased Bax protein expression in human leukemia (HL-60) cells [55], via activated JNK/p38 MAPK pathways and Fas/FasL pathways, increased translocation of Bax, and reduced Bcl-2 levels in human gastric adenocarcinoma cells [56] and via induced JNK and p38 MAPK pathways in HepG2 hepatocellular carcinoma cells [57]. Moreover, PCA also demonstrated anticancer properties by causing apoptosis or suppressing invasion and metastasis in human breast, lung, liver, cervix and prostate cancer cells [58]. Consistently, an in vivo study (Table 9) also demonstrated that PCA inhibited N-nitrosomethylbenzylamine (NMBA) induced esophageal tumorigenesis by its inhibitory effects on genes associated with inflammation in rats [59].

Table 8.

Summary of in vitro studies of proapoptotic activity of PCA in cancer cells.

Model Model/method PCA concentration Major finding Interpretation Reference
HL-60 leukemia cells Direct PCA application to cells 0.2, 0.5, 1, and 2 mM (i) PCA increased DNA fragmentation
(ii) PCA declined hyperphosphorylated RB level
(iii) PCA reduced Bcl-2 protein expression
(iv) PCA increased Bax protein expression
PCA had an antiproliferative effect via induced RB phosphorylation and degradation and Bcl-2 protein suppression in cancer cells [55]

Human gastric adenocarcinoma (AGS) cells Direct PCA application to cells 0.5, 1.0, 2.0, 4.0, 6.0, 8.0, and 9.0 mM (i) PCA increased apoptotic bodies formation
(ii) PCA increased p-JNK expression
(iii) PCA increased p-p53 expression
(iv) PCA increased phosphorylation of ATF-2 at Thr69/71 and c-Jun at Ser73
(v) PCA increased Fas expression
(vi) PCA increased FasL expression
(vii) PCA decreased Bcl-2 expression
(viii) PCA increased Bax expression
PCA induced apoptosis via JNK/p38 MAPK pathway, activated Fas/FasL pathway, increased translocation of Bax, and reduced Bcl-2 in cancer cells [56]

HepG2 hepatocellular carcinoma cells Direct PCA application to cells 0, 3, 10, 30, 100, and 300 μmol/L (i) PCA decreased viability of HepG2 hepatocellular carcinoma
(ii) PCA increased JNK and p53 expression
PCA induced cell death via activating JNK and p38 MAPK pathways in cancer cells [57]

Human breast, lung, liver, cervix, and prostate cancer cells Direct PCA application to cells 1, 2, 4, and 8 μmol/L (i) PCA decreased viability
(ii) PCA enhanced DNA fragmentation
(iii) PCA decreased MMP
(iv) PCA lowered Na+-K+-ATPase activity
(v) PCA increased caspase-3 activity
(vi) PCA increased caspase-8 activity
(vii) PCA decreased ICAM-1 level
(viii) PCA at 2, 4, and 8 μmol/L decreased VEGF level
(ix) PCA suppressed IL-6 and IL-8 levels
PCA had anticancer properties via increased apoptosis or suppressed invasion and metastasis cancer cells [58]

Table 9.

Summary of in vivo studies of proapoptotic activity of PCA.

Model Method PCA dose/route/duration Major finding Interpretation Reference
Rat NMBA-induced esophageal cancer in rats PCA 0.05% in diet for 15, 25, and 35 weeks (i) PCA reduced area of hyperplasia began at week 25
(ii) PCA reduced COX-2, iNOS, soluble epoxide hydrolase (she), and pentraxin-3 (PTX3) mRNA expression levels
PCA prevented esophageal tumorigenesis, by inhibitory effects on genes associated with inflammation [59]

6. Antimicrobial Activity of PCA

In vitro studies (Table 10) demonstrated that PCA has an antimicrobial effect against gram positive and negative bacteria and fungi [60, 61]. PCA also prevented contamination of meat by Campylobacter and aerobes, by decreasing lipid oxidation [62]. PCA exerted its antibacterial effects due to its ability to inhibit bacterial growth and increase the synergistic effects of antibiotics hence reducing the possibility of resistance to drugs [63]. These antimicrobial activities of PCA have been proposed as promising applications in both health protection and food preservation in order to avoid food-borne illnesses [62, 64].

Table 10.

Summary of in vitro studies of antimicrobial activity of PCA.

Model Method PCA concentration Major finding Interpretation Reference
Campylobacter spp Antimicrobial activity testing (i) 10 mg/mL
(ii) 5, 10 mg/100 g beef
(i) PCA inhibited growth and susceptible and antibiotic-resistant Campylobacter species
(ii) PCA inhibited growth of aerobes in beef samples
(iii) PCA decreased lipid oxidation levels in ground beef
(i) PCA could preserve foods to prevent contamination by Campylobacter and aerobes, via decreased lipid oxidation [62]

Pseudomonas aeruginosa Antimicrobial susceptibility testing 2,000 μg/mL (i) PCA inhibited growth of Pseudomonas  aeruginosa
(ii) PCA plus sulfamethoxazole increased synergistic mode of inhibition of P. aeruginosa
(i) PCA had an antibacterial effect by inhibiting bacterial growth and increasing the synergistic effects on antibiotics to reduced drug resistance [63]

Bacteria and fungi Antimicrobial activity testing 1.22–625 μg/mL (i) PCA prevented 80% of the growth of organisms (i) PCA had an antimicrobial effect against gram positive and negative bacteria and against fungi [60]

Helicobacter pylori Antimicrobial susceptibility testing 8–64 mg/L (i) PCA inhibited growth of H. pylori
(ii) PCA reduced drug-resistant H. pylori
(iii) PCA at (32–40 mg/L) reduced urease activity of H. pylori to 40%
(i) PCA had growth prevention effects on H. pylori [61]

7. Conclusion

Growing evidence suggests the significant biological potential of PCA through the modulation of cellular signals involved in the control of oxidative stress and inflammation. Moreover, its antiapoptotic effects in normal cells and proapoptotic effects in cancer cells suggest definite benefits as a potential chemotherapeutic agent. However, much evidence of such properties has been collected from cellular and animal studies, while clinical studies are still lacking. Future clinical studies are needed to warrant the clinical usefulness of the PCA.

Acknowledgments

This work was supported by the Thailand Research Fund RTA5580006 (NC) and BRG5780016 (SC), Chiang Mai University Center of Excellence Award (NC), Udon Thani Rajabhat University Fund (YS), and a NSTDA Research Chair Grant from the National Science and Technology Development Agency (NC).

Conflict of Interests

The authors declare that there is no conflict of interests regarding the publication of this paper.

References

  • 1.Lin W.-L., Hsieh Y.-J., Chou F.-P., Wang C.-J., Cheng M.-T., Tseng T.-H. Hibiscus protocatechuic acid inhibits lipopolysaccharide-induced rat hepatic damage. Archives of Toxicology. 2003;77(1):42–47. doi: 10.1007/s00204-002-0404-0. [DOI] [PubMed] [Google Scholar]
  • 2.Pacheco-Palencia L. A., Mertens-Talcott S., Talcott S. T. Chemical composition, antioxidant properties, and thermal stability of a phytochemical enriched oil from Açai (Euterpe oleracea Mart.) Journal of Agricultural and Food Chemistry. 2008;56(12):4631–4636. doi: 10.1021/jf800161u. [DOI] [PubMed] [Google Scholar]
  • 3.Li P., Wang X. Q., Wang H. Z., Wu Y. N. High performance liquid chromatographic determination of phenolic acids in fruits and vegetables. Biomedical and Environmental Sciences. 1993;6(4):389–398. [PubMed] [Google Scholar]
  • 4.Vitaglione P., Donnarumma G., Napolitano A., et al. Protocatechuic acid is the major human metabolite of cyanidin-glucosides. Journal of Nutrition. 2007;137(9):2043–2048. doi: 10.1093/jn/137.9.2043. [DOI] [PubMed] [Google Scholar]
  • 5.Pimpao R. C., Dew T., Figueira M. E., et al. Urinary metabolite profiling identifies novel colonic metabolites and conjugates of phenolics in healthy volunteers. Molecular Nutrition & Food Research. 2014;58(7):1414–1425. doi: 10.1002/mnfr.201300822. [DOI] [PubMed] [Google Scholar]
  • 6.González-Gallego J., García-Mediavilla M. V., Sánchez-Campos S., Tuñó M. J. Fruit polyphenols, immunity and inflammation. British Journal of Nutrition. 2010;104(supplement 3):S15–S27. doi: 10.1017/s0007114510003910. [DOI] [PubMed] [Google Scholar]
  • 7.Landberg R., Sun Q., Rimm E. B., et al. Selected dietary flavonoids are associated with markers of inflammation and endothelial dysfunction in U.S. women. The Journal of Nutrition. 2011;141(4):618–625. doi: 10.3945/jn.110.133843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Rotelli A. E., Guardia T., Juárez A. O., de la Rocha N. E., Pelzer L. E. Comparative study of flavonoids in experimental models of inflammation. Pharmacological Research. 2003;48(6):601–606. doi: 10.1016/s1043-6618(03)00225-1. [DOI] [PubMed] [Google Scholar]
  • 9.Garcia-Alonso M., Minihane A.-M., Rimbach G., Rivas-Gonzalo J. C., de Pascual-Teresa S. Red wine anthocyanins are rapidly absorbed in humans and affect monocyte chemoattractant protein 1 levels and antioxidant capacity of plasma. The Journal of Nutritional Biochemistry. 2009;20(7):521–529. doi: 10.1016/j.jnutbio.2008.05.011. [DOI] [PubMed] [Google Scholar]
  • 10.Wang H., Nair M. G., Strasburg G. M., et al. Antioxidant and antiinflammatory activities of anthocyanins and their aglycon, cyanidin, from tart cherries. Journal of Natural Products. 1999;62(2):294–296. doi: 10.1021/np980501m. [DOI] [PubMed] [Google Scholar]
  • 11.Kähkönen M. P., Heinonen M. Antioxidant activity of anthocyanins and their aglycons. Journal of Agricultural and Food Chemistry. 2003;51(3):628–633. doi: 10.1021/jf025551i. [DOI] [PubMed] [Google Scholar]
  • 12.Matsumoto H., Nakamura Y., Hirayama M., Yoshiki Y., Okubo K. Antioxidant activity of black currant anthocyanin aglycons and their glycosides measured by chemiluminescence in a neutral pH region and in human plasma. Journal of Agricultural and Food Chemistry. 2002;50(18):5034–5037. doi: 10.1021/jf020292i. [DOI] [PubMed] [Google Scholar]
  • 13.Tsuda T., Shiga K., Ohshima K., Kawakishi S., Osawa T. Inhibition of lipid peroxidation and the active oxygen radical scavenging effect of anthocyanin pigments isolated from Phaseolus vulgaris L. Biochemical Pharmacology. 1996;52(7):1033–1039. doi: 10.1016/0006-2952(96)00421-2. [DOI] [PubMed] [Google Scholar]
  • 14.Cassidy A., de Pascual Teresa S., Rimbach G. Molecular mechanisms by which dietary isoflavones potentially prevent atherosclerosis. Expert Reviews in Molecular Medicine. 2003;5(24):1–15. doi: 10.1017/S1462399403006732. [DOI] [PubMed] [Google Scholar]
  • 15.Kay C. D., Kroon P. A., Cassidy A. The bioactivity of dietary anthocyanins is likely to be mediated by their degradation products. Molecular Nutrition and Food Research. 2009;53(supplement 1):S92–S101. doi: 10.1002/mnfr.200800461. [DOI] [PubMed] [Google Scholar]
  • 16.Lin C.-Y., Tsai S.-J., Huang C.-S., Yin M.-C. Antiglycative effects of protocatechuic acid in the kidneys of diabetic mice. Journal of Agricultural and Food Chemistry. 2011;59(9):5117–5124. doi: 10.1021/jf200103f. [DOI] [PubMed] [Google Scholar]
  • 17.Moura M. B., dos Santos L. S., van Houten B. Mitochondrial dysfunction in neurodegenerative diseases and cancer. Environmental and Molecular Mutagenesis. 2010;51(5):391–405. doi: 10.1002/em.20575. [DOI] [PubMed] [Google Scholar]
  • 18.Queen B. L., Tollefsbol T. O. Polyphenols and aging. Current Aging Science. 2010;3(1):34–42. doi: 10.2174/1874609811003010034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Patten D. A., Germain M., Kelly M. A., Slack R. S. Reactive oxygen species: stuck in the middle of neurodegeneration. Journal of Alzheimer's Disease. 2010;20(2):S357–S367. doi: 10.3233/jad-2010-100498. [DOI] [PubMed] [Google Scholar]
  • 20.Kaneto H., Katakami N., Matsuhisa M., Matsuoka T.-A. Role of reactive oxygen species in the progression of type 2 diabetes and atherosclerosis. Mediators of Inflammation. 2010;2010:11. doi: 10.1155/2010/453892.453892 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Patten D. A., Germain M., Kelly M. A., Slack R. S. Reactive oxygen species: stuck in the middle of neurodegeneration. Journal of Alzheimer's Disease. 2010;20(supplement 2):S357–S367. doi: 10.3233/jad-2010-100498. [DOI] [PubMed] [Google Scholar]
  • 22.Sroka Z., Cisowski W. Hydrogen peroxide scavenging, antioxidant and anti-radical activity of some phenolic acids. Food and Chemical Toxicology. 2003;41(6):753–758. doi: 10.1016/s0278-6915(02)00329-0. [DOI] [PubMed] [Google Scholar]
  • 23.Masella R., Varì R., D'Archivio M., et al. Extra virgin olive oil biophenols inhibit cell-mediated oxidation of LDL by increasing the mRNA transcription of glutathione-related enzymes. Journal of Nutrition. 2004;134(4):785–791. doi: 10.1093/jn/134.4.785. [DOI] [PubMed] [Google Scholar]
  • 24.Varì R., D'Archivio M., Filesi C., et al. Protocatechuic acid induces antioxidant/detoxifying enzyme expression through JNK-mediated Nrf2 activation in murine macrophages. Journal of Nutritional Biochemistry. 2011;22(5):409–417. doi: 10.1016/j.jnutbio.2010.03.008. [DOI] [PubMed] [Google Scholar]
  • 25.Tarozzi A., Morroni F., Hrelia S., et al. Neuroprotective effects of anthocyanins and their in vivo metabolites in SH-SY5Y cells. Neuroscience Letters. 2007;424(1):36–40. doi: 10.1016/j.neulet.2007.07.017. [DOI] [PubMed] [Google Scholar]
  • 26.Shi G.-F., An L.-J., Jiang B., Guan S., Bao Y.-M. Alpinia protocatechuic acid protects against oxidative damage in vitro and reduces oxidative stress in vivo. Neuroscience Letters. 2006;403(3):206–210. doi: 10.1016/j.neulet.2006.02.057. [DOI] [PubMed] [Google Scholar]
  • 27.Chou T.-H., Ding H.-Y., Lin R.-J., Liang J.-Y., Liang C.-H. Inhibition of melanogenesis and oxidation by protocatechuic acid from Origanum vulgare (Oregano) Journal of Natural Products. 2010;73(11):1767–1774. doi: 10.1021/np100281g. [DOI] [PubMed] [Google Scholar]
  • 28.Zhang X., Shi G.-F., Liu X.-Z., An L.-J., Guan S. Anti-ageing effects of protocatechuic acid from Alpinia on spleen and liver antioxidative system of senescent mice. Cell Biochemistry and Function. 2011;29(4):342–347. doi: 10.1002/cbf.1757. [DOI] [PubMed] [Google Scholar]
  • 29.Tsai S.-J., Yin M.-C. Anti-glycative and anti-inflammatory effects of protocatechuic acid in brain of mice treated by d-galactose. Food and Chemical Toxicology. 2012;50(9):3198–3205. doi: 10.1016/j.fct.2012.05.056. [DOI] [PubMed] [Google Scholar]
  • 30.Semaming Y., Kumfu S., Pannangpetch P., Chattipakorn S. C., Chattipakorn N. Protocatechuic acid exerts a cardioprotective effect in type 1 diabetic rats. Journal of Endocrinology. 2014;223(1):13–23. doi: 10.1530/joe-14-0273. [DOI] [PubMed] [Google Scholar]
  • 31.Feghali C. A., Wright T. M. Cytokines in acute and chronic inflammation. Frontiers in Bioscience. 1997;2:d12–d26. doi: 10.2741/a171. [DOI] [PubMed] [Google Scholar]
  • 32.Lawrence T., Gilroy D. W. Chronic inflammation: a failure of resolution? International Journal of Experimental Pathology. 2007;88(2):85–94. doi: 10.1111/j.1365-2613.2006.00507.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Fonseca M. I., Chu S.-H., Berci A. M., et al. Contribution of complement activation pathways to neuropathology differs among mouse models of Alzheimer's disease. Journal of Neuroinflammation. 2011;8(1, article 4) doi: 10.1186/1742-2094-8-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Medzhitov R. Inflammation 2010: new adventures of an old flame. Cell. 2010;140(6):771–776. doi: 10.1016/j.cell.2010.03.006. [DOI] [PubMed] [Google Scholar]
  • 35.Manicone A. M., McGuire J. K. Matrix metalloproteinases as modulators of inflammation. Seminars in Cell and Developmental Biology. 2008;19(1):34–41. doi: 10.1016/j.semcdb.2007.07.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Lawrence T., Fong C. The resolution of inflammation: anti-inflammatory roles for NF-κB. The International Journal of Biochemistry & Cell Biology. 2010;42(4):519–523. doi: 10.1016/j.biocel.2009.12.016. [DOI] [PubMed] [Google Scholar]
  • 37.Min S.-W., Ryu S.-N., Kim D.-H. Anti-inflammatory effects of black rice, cyanidin-3-O-β-d-glycoside, and its metabolites, cyanidin and protocatechuic acid. International Immunopharmacology. 2010;10(8):959–966. doi: 10.1016/j.intimp.2010.05.009. [DOI] [PubMed] [Google Scholar]
  • 38.Wang D., Wei X., Yan X., Jin T., Ling W. Protocatechuic acid, a metabolite of anthocyanins, inhibits monocyte adhesion and reduces atherosclerosis in apolipoprotein E-deficient mice. Journal of Agricultural and Food Chemistry. 2010;58(24):12722–12728. doi: 10.1021/jf103427j. [DOI] [PubMed] [Google Scholar]
  • 39.Wang D., Zou T., Yang Y., Yan X., Ling W. Cyanidin-3-O-β-glucoside with the aid of its metabolite protocatechuic acid, reduces monocyte infiltration in apolipoprotein E-deficient mice. Biochemical Pharmacology. 2011;82(7):713–719. doi: 10.1016/j.bcp.2011.04.007. [DOI] [PubMed] [Google Scholar]
  • 40.Yan J.-J., Jung J.-S., Hong Y.-J., et al. Protective effect of protocatechuic acid isopropyl ester against murine models of sepsis: inhibition of TNF-alpha and nitric oxide production and augmentation of IL-10. Biological and Pharmaceutical Bulletin. 2004;27(12):2024–2027. doi: 10.1248/bpb.27.2024. [DOI] [PubMed] [Google Scholar]
  • 41.Rosen E. D., Spiegelman B. M. Adipocytes as regulators of energy balance and glucose homeostasis. Nature. 2006;444(7121):847–853. doi: 10.1038/nature05483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Selvin E., Steffes M. W., Zhu H., et al. Glycated hemoglobin, diabetes, and cardiovascular risk in nondiabetic adults. The New England Journal of Medicine. 2010;362(9):800–811. doi: 10.1056/nejmoa0908359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Nathan D. M., Cleary P. A., Backlund J.-Y. C., et al. Intensive diabetes treatment and cardiovascular disease in patients with type 1 diabetes. The New England Journal of Medicine. 2005;353(25):2643–2653. doi: 10.1056/nejmoa052187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Jones J. R., Barrick C., Kim K.-A., et al. Deletion of PPARγ in adipose tissues of mice protects against high fat diet-induced obesity and insulin resistance. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(17):6207–6212. doi: 10.1073/pnas.0306743102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Scazzocchio B., Varì R., Filesi C., et al. Cyanidin-3-O-β-glucoside and protocatechuic acid exert insulin-like effects by upregulating PPARγ activity in human omental adipocytes. Diabetes. 2011;60(9):2234–2244. doi: 10.2337/db10-1461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Harini R., Pugalendi K. V. Antihyperglycemic effect of protocatechuic acid on streptozotocin-diabetic rats. Journal of Basic and Clinical Physiology and Pharmacology. 2010;21(1):79–91. doi: 10.1515/jbcpp.2010.21.1.79. [DOI] [PubMed] [Google Scholar]
  • 47.D'Archivio M., Santangelo C., Scazzocchio B., et al. Modulatory effects of polyphenols on apoptosis induction: relevance for cancer prevention. International Journal of Molecular Sciences. 2008;9(3):213–228. doi: 10.3390/ijms9030213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Giovannini C., Scazzocchio B., Varì R., Santangelo C., D'Archivio M., Masella R. Apoptosis in cancer and atherosclerosis: polyphenol activities. Annali dell'Istituto Superiore di Sanita. 2007;43(4):406–416. [PubMed] [Google Scholar]
  • 49.Zhou-Stache J., Buettner R., Artmann G., Mittermayer C., Bosserhoff A. K. Inhibition of TNF-α induced cell death in human umbilical vein endothelial cells and Jurkat cells by protocatechuic acid. Medical and Biological Engineering and Computing. 2002;40(6):698–703. doi: 10.1007/bf02345308. [DOI] [PubMed] [Google Scholar]
  • 50.Guan S., Jiang B., Bao Y. M., An L. J. Protocatechuic acid suppresses MPP+-induced mitochondrial dysfunction and apoptotic cell death in PC12 cells. Food and Chemical Toxicology. 2006;44(10):1659–1666. doi: 10.1016/j.fct.2006.05.004. [DOI] [PubMed] [Google Scholar]
  • 51.Guan S., Ge D., Liu T.-Q., Ma X.-H., Cui Z.-F. Protocatechuic acid promotes cell proliferation and reduces basal apoptosis in cultured neural stem cells. Toxicology in Vitro. 2009;23(2):201–208. doi: 10.1016/j.tiv.2008.11.008. [DOI] [PubMed] [Google Scholar]
  • 52.Guan S., Bao Y.-M., Jiang B., An L.-J. Protective effect of protocatechuic acid from Alpinia oxyphylla on hydrogen peroxide-induced oxidative PC12 cell death. European Journal of Pharmacology. 2006;538(1–3):73–79. doi: 10.1016/j.ejphar.2006.03.065. [DOI] [PubMed] [Google Scholar]
  • 53.Zhang H.-N., An C.-N., Xu M., Guo D.-A., Li M., Pu X.-P. Protocatechuic acid inhibits rat pheochromocytoma cell damage induced by a dopaminergic neurotoxin. Biological and Pharmaceutical Bulletin. 2009;32(11):1866–1869. doi: 10.1248/bpb.32.1866. [DOI] [PubMed] [Google Scholar]
  • 54.Tseng T.-H., Wang C.-J., Kao E.-S., Chu H.-Y. Hibiscus protocatechuic acid protects against oxidative damage induced by tert-butylhydroperoxide in rat primary hepatocytes. Chemico-Biological Interactions. 1996;101(2):137–148. doi: 10.1016/0009-2797(96)03721-0. [DOI] [PubMed] [Google Scholar]
  • 55.Tseng T.-H., Kao T.-W., Chu C.-Y., Chou F.-P., Lin W.-L., Wang C.-J. Induction of apoptosis by Hibiscus protocatechuic acid in human leukemia cells via reduction of retinoblastoma (RB) phosphorylation and Bcl-2 expression. Biochemical Pharmacology. 2000;60(3):307–315. doi: 10.1016/s0006-2952(00)00322-1. [DOI] [PubMed] [Google Scholar]
  • 56.Lin H.-H., Chen J.-H., Huang C.-C., Wang C.-J. Apoptotic effect of 3,4-dihydroxybenzoic acid on human gastric carcinoma cells involving JNK/p38 MAPK signaling activation. International Journal of Cancer. 2007;120(11):2306–2316. doi: 10.1002/ijc.22571. [DOI] [PubMed] [Google Scholar]
  • 57.Yip E. C. H., Chan A. S. L., Pang H., Tam Y. K., Wong Y. H. Protocatechuic acid induces cell death in HepG2 hepatocellular carcinoma cells through a c-Jun N-terminal kinase-dependent mechanism. Cell Biology and Toxicology. 2006;22(4):293–302. doi: 10.1007/s10565-006-0082-4. [DOI] [PubMed] [Google Scholar]
  • 58.Yin M.-C., Lin C.-C., Wu H.-C., Tsao S.-M., Hsu C.-K. Apoptotic effects of protocatechuic acid in human breast, lung, liver, cervix, and prostate cancer cells: potential mechanisms of action. Journal of Agricultural and Food Chemistry. 2009;57(14):6468–6473. doi: 10.1021/jf9004466. [DOI] [PubMed] [Google Scholar]
  • 59.Peiffer D. S., Zimmerman N. P., Wang L.-S., et al. Chemoprevention of esophageal cancer with black raspberries, their component anthocyanins, and a major anthocyanin metabolite, protocatechuic acid. Cancer Prevention Research. 2014;7(6):574–584. doi: 10.1158/1940-6207.capr-14-0003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Kuete V., Nana F., Ngameni B., Mbaveng A. T., Keumedjio F., Ngadjui B. T. Antimicrobial activity of the crude extract, fractions and compounds from stem bark of Ficus ovata (Moraceae) Journal of Ethnopharmacology. 2009;124(3):556–561. doi: 10.1016/j.jep.2009.05.003. [DOI] [PubMed] [Google Scholar]
  • 61.Liu W. H., Hsu C. C., Yin M. C. In vitro anti-Helicobacter pylori activity of diallyl sulphides and protocatechuic acid. Phytotherapy Research. 2008;22(1):53–57. doi: 10.1002/ptr.2259. [DOI] [PubMed] [Google Scholar]
  • 62.Yin M.-C., Chao C.-Y. Anti-Campylobacter, anti-aerobic, and anti-oxidative effects of roselle calyx extract and protocatechuic acid in ground beef. International Journal of Food Microbiology. 2008;127(1-2):73–77. doi: 10.1016/j.ijfoodmicro.2008.06.002. [DOI] [PubMed] [Google Scholar]
  • 63.Jayaraman P., Sakharkar M. K., Lim C. S., Tang T. H., Sakharkar K. R. Activity and interactions of antibiotic and phytochemical combinations against pseudomonas aeruginosa in vitro. International Journal of Biological Sciences. 2010;6(6):556–568. doi: 10.7150/ijbs.6.556. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Chao C.-Y., Yin M.-C. Antibacterial effects of roselle calyx extracts and protocatechuic acid in ground beef and apple juice. Foodborne Pathogens and Disease. 2009;6(2):201–206. doi: 10.1089/fpd.2008.0187. [DOI] [PubMed] [Google Scholar]

Articles from Evidence-based Complementary and Alternative Medicine : eCAM are provided here courtesy of Wiley

RESOURCES