Next Article in Journal
Who Adopts Agroforestry in a Subsistence Economy?—Lessons from the Terai of Nepal
Next Article in Special Issue
Evaluation of Wild Foods for Responsible Human Consumption and Sustainable Use of Natural Resources
Previous Article in Journal
Quantifying Ecosystem Service Trade-Offs to Inform Spatial Identification of Forest Restoration
Previous Article in Special Issue
Antioxidant Properties of Green Coffee Extract
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Perspective

Hawthorn (Crataegus spp.): An Updated Overview on Its Beneficial Properties

by
Amirhossein Nazhand
1,
Massimo Lucarini
2,*,
Alessandra Durazzo
2,
Massimo Zaccardelli
3,
Santo Cristarella
4,
Selma B. Souto
5,
Amélia M. Silva
6,7,
Patrícia Severino
8,9,10,
Eliana B. Souto
11,12 and
Antonello Santini
13,*
1
Department of Biotechnology, Sari Agricultural Science and Natural Resource University, 9th km of Farah Abad Road, Sari 48181 68984, Mazandaran, Iran
2
CREA-Research Centre for Food and Nutrition, Via Ardeatina 546, 00178 Roma, Italy
3
CREA-Research Centre for Vegetable and Ornamental Crops, Via Cavalleggeri 25, 84098 Pontecagnano (Salerno), Italy
4
Department of Veterinary Sciences, University of Messina, Polo Universitario dell’Annunziata, 98168 Messina, Italy
5
Department of Endocrinology of Braga Hospital, Sete Fontes, São Victor, 4710-243 Braga, Portugal
6
School of Biology and Environment, University of Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, P-5001-801 Vila Real, Portugal
7
Centre for Research and Technology of Agro-Environmental and Biological Sciences (CITAB), University of Trás-os-Montes e Alto Douro (UTAD), Quinta de Prados, 5001-801 Vila Real, Portugal
8
Industrial Biotechnology Program, University of Tiradentes (UNIT), Av. Murilo Dantas 300, Aracaju 49032-490, Brazil
9
Tiradentes Institute, 150 Mt Vernon St., Dorchester, MA 02125, USA
10
Laboratory of Nanotechnology and Nanomedicine (LNMED), Institute of Technology and Research (ITP), Av. Murilo Dantas, 300, Aracaju 49010-390, Brazil
11
Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
12
CEB-Centre of Biological Engineering, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
13
Department of Pharmacy, University of Napoli Federico II, Via D. Montesano 49, 80131 Napoli, Italy
*
Authors to whom correspondence should be addressed.
Forests 2020, 11(5), 564; https://doi.org/10.3390/f11050564
Submission received: 20 April 2020 / Revised: 5 May 2020 / Accepted: 12 May 2020 / Published: 18 May 2020
(This article belongs to the Special Issue Forest, Foods and Nutrition)

Abstract

:
Medicinal plants, many of which are wild, have recently been under the spotlight worldwide due to growing requests for natural and sustainable eco-compatible remedies for pathological conditions with beneficial health effects that are able to support/supplement a daily diet or to support and/or replace conventional pharmacological therapy. The main requests for these products are: safety, minimum adverse unwanted effects, better efficacy, greater bioavailability, and lower cost when compared with synthetic medications available on the market. One of these popular herbs is hawthorn (Crataegus spp.), belonging to the Rosaceae family, with about 280 species present in Europe, North Africa, West Asia, and North America. Various parts of this herb, including the berries, flowers, and leaves, are rich in nutrients and beneficial bioactive compounds. Its chemical composition has been reported to have many health benefits, including medicinal and nutraceutical properties. Accordingly, the present review gives a snapshot of the in vitro and in vivo therapeutic potential of this herb on human health.

1. Introduction

Medicinal wild plants and herbs have recently received increased interest worldwide since they are rich sources of bioactive compounds and for their potential beneficial health properties, which have often been well known for centuries [1,2,3,4,5,6,7,8,9,10,11,12,13,14,15,16,17,18]. The World Health Organization (WHO) reported that about 80% of the world’s population uses traditional drugs, including herbal medicine, for the treatment of diseases before considering conventional drugs when available [19]. One of these interesting popular medicinal plants is hawthorn (Crataegus spp.), a deciduous branched shrub/small tree that is twisted and thorny, belonging to the Rosaceae family and Maloideae sub-family. Hawthorn is present worldwide with about 280 species, among which the most common are: C. monogyna, C. laevigata, C. mexicana and C. douglasii, grown in Europe, North Africa, West Asia, and North America. The scientific name of hawthorn comes from the Greek word “kràtaigos” which means “strength and robustness” due to its hard and durable wood. Natural habitats of hawthorn are wooded and sunny areas on predominantly limestone soils up to 1500 m above sea level. This species is very rustic and is not very water demanding. C. monogyna has leaves that are 20–60 mm long with a rhomboidal shape that are deeply engraved and have notched lobes; the flowers are white/pink and form blooms of 5–35 units; the fruits are red berries of 10 mm when ripened, and contain one seed. Flowering takes place between April and May, and fruit ripening between September and October. Various parts of this plant—in particular, the berries, flowers, and leaves—are rich in nutrients, and have been traditionally associated with many health, medicinal or nutraceutical beneficial health effects [20], e.g., anti-microbial, anti-inflammatory, antioxidant, anti-cancer, and anticoagulant properties. Some of the most relevant properties associated to this plant are reported in Figure 1. According to its traditional use, and since it is generally recognized as safe (GRAS), the Committee for Herbal Medicinal Products of the European Medicines Agency classified hawthorn as a “traditional herbal medicinal product” [21,22]. This wild plant has been used as a traditional medicine, herbal drug, and food supplement for centuries [23,24]. According to the holistic and traditional approach, hawthorn leaves and flowers are used to prepare infusions that can be used to control palpitations, tachycardia, and nervousness. Away from meals, hawthorn has been used against hypertension and, before sleeping, for its relaxing and sedative actions. The berries promote cardiovascular health, protecting from angina, hypertension, heart failure, cardiac arrhythmias, myocarditis, arteriosclerosis, insomnia, and anxiety. Moreover, the berries are astringents and diuretics, and can act against diarrhea, urinary retention, and intestinal cramps. Indigenous peoples from Latin America use the berries for the preparation of a highly energetic drink called “Pennican”, and, in many parts of the world, the berries are used to prepare jams and as flavoring for dishes like white meats. Hawthorn, however, can also have a few collateral effects and contraindications; in particular, it is not recommended when blood pressure is low. Considering the multiple health properties of this medicinal wild herb, this review describes the potential use of hawthorn in therapy and as a support of some human health conditions.

2. Phytochemical Composition of Hawthorn

Chemical analysis has allowed for the identification of more than 150 bioactive molecules in hawthorn, including phenolic acids (ferulic, gallic, p-coumaric, syringic, chlorogenic, caffeic), quercetin, pyrocatechin, phlorodizin, terpenoids, lignans, steroids, organic acids (fumaric, tartaric, succinic, citric, malic), and sugars (maltose, sucrose, glucose, fructose). These are represented in Figure 2 [25,26].
Polyphenol compounds from C. oxyacantha extracts, including epicatechin, epicatechin gallate (ECG), rutin, caffeic, and caftaric acids, were identified using HPLC-DAD and LC-MS/MS techniques [27]. In a study, UV/MS analysis coupled with 1D/2D nuclear magnetic resonance (NMR) spectroscopy was used to detect the compounds extracted from the ethyl acetate extract of C. oxyacantha, which included naringenin, epicatechin, quercetin-3-O-β-glucoside, and quercetin [28]. The presence of rutin and quercetin obtained from C. oxyacantha extracts using HPLC was also reported [29]. The work of Nabavi et al. focused on the polyphenolic composition of C. monogyna Jacq., ranging from its chemistry and composition to its medical applications [30]. The recent work of Cao et al. [31] gives an updated snapshot of the water-based extraction of the bioactive principles of hawthorn, describing the current experimental laboratory research and further valuable information. In this study, attention has been addressed to the quantitative and qualitative aspects of the extraction, as well as to the kinetics of the extraction according to the part of the plant (flowers or leaves), their state (fresh or dried), and the granulometry of the dry plant, also taking into account parameters like stirring speed, temperature, extraction time, volume of the container (cup, mug or bowl) and the use of infusion bags. In agreement with green technologies [32,33], it is worth mentioning the work of Hu et al. [34], which proposed an eco-friendly microwave-assisted extraction of bioactive compounds from hawthorn leaf combined with ultra-high-performance liquid chromatography coupled with an ultraviolet detector for the identification and quantification of compounds. In a recent study, mannose, glucose and fructose were extracted from hawthorn fruits by acid hydrolysis using 2 M trifluoroacetic acid, and then identified and characterized by gas chromatography/mass spectrometry [35]. Zhao et al. [36] used headspace/solid phase microextraction (HS/SPME) coupled with gas chromatography/mass spectrometry (GC/MS) to determine the chemical composition of hawthorn fruits, reporting that alcohols and esters are the main compounds present. Salmanian et al. detected the phenolic acids contained in the hawthorn pulp and seed extract using RP-HPLC and reported that chlorogenic acid is the main one [37]. Liu et al. [38] applied HPLC-UV/ESI-MS to determine the phenolic constituents of hawthorn, which was found to contain C-glycosyl flavones, hyperoside, procyanidins B2/C1, and epicatechin. Lund et al. [39], by using nuclear magnetic resonance (NMR) spectrometry, identified chlorogenic acid and flavonoids of Crataegus species, including vitexin-2″-O-rhamnoside, rutin, hyperoside, and naringenin. In their study, HPLC-DAD analysis was also used to confirm the obtained results. The hawthorn seed extract distillation at the optimum temperature (in the range of 211 to 230 °C) was analyzed by gas chromatography coupled with a mass spectrometer (GC-MS) to determine the chemical composition, with the aim of proposing this method as a cost-effective technique to obtain hawthorn products on an industrial scale [40]. The chemical compounds present in Crataegus species, mainly quercetin, hyperoside, rutin, and vitexin, have been also studied using HPLC-UV and UV-Vis spectrophotometry [41]. The hawthorn fruit examined by spectrophotometry at a wavelength of 285 ± 2 nm revealed the presence of hyperoside flavonoid in an amount up to 0.112–0.183% (w/w) [42]. Table 1 reports the main compounds found in hawthorn and the methodological and analytical approach used in their characterization.

3. In Vitro and In Vivo Therapeutic Potentials of Hawthorn: An Updated Snapshot

The evaluation of phytochemical composition can be considered as the first step for the determination of the beneficial health properties of a plant [53,54]. Figure 1 summarizes the health properties as reported in the literature from in vitro and in vivo studies.
As indicated above, many beneficial properties have been attributed to hawthorn, including anticancer [55], anti-HIV, anti-diabetic [56], and anticoagulant activity [57], cardioprotective effects [58,59,60,61,62,63,64,65], hepatoprotective effects, antihyperglycemic and antihyperlipidemic activities, wound healing effects [66], antimicrobial effects, gastroprotective effects, treatment of metabolic syndrome [67], regulation of cholesterol homeostasis [68], anti-atherosclerosis effects [69,70,71,72], anti-aging effects [73], ischemia protective effects [74], treatment of cognitive disorders, neuroprotective effects, regulating gastrointestinal motility [75], anti-inflammatory activities [76,77], regulation of the gut–brain axis [78], treatment of hypertension [79], antioxidant activity [80,81,82,83,84,85], anti-hypoxic activities [86], antidepressant effects [87], anti-Alzheimer’s effects, and treatment of intestinal microbial disorder [88].
In the following sections, an updated snapshot of the various potential therapeutic effects of hawthorn in vitro and in vivo are described, as well as its beneficial properties for human health.

3.1. Health-Promoting Activities of Hawthorn In Vitro

Many in vitro studies reported different health-promoting effects for hawthorn extracts [89,90,91,92]. The administration of homogeneous polysaccharide (HPS) extracted from hawthorn at a concentration of 125–1000 µg/mL showed anticancer activity against a human colon cancer cell line HCT116, after 12 h by arresting the cell cycle and inducing cell apoptosis through extrinsic and intrinsic mechanisms using P38 mitogen-activated protein kinase and the phosphatidylinositol-3-kinase/AKT/mammalian target of rapamycin signaling pathway [93]. Hawthorn fruit peel extract exhibited antioxidant activity (2,2,1-diphenyl-1-picrylhydrazyl (DPPH) IC50 value of 6.72 μg/mL), acetylcholinesterase inhibitory effects (IC50 value of 11.72 μg/mL), and cytotoxic effects against the human tumor cells SKOV-3 and MCF-7 (IC50 values of 80.11 μg/mL and 2.76 μg/mL, respectively) [94]. A recent study concluded that hawthorn extract-Selenium nano particles caused mitochondrial dysfunction and intracellular oxidative stress to start the apoptosis of HepG2 cells via the mitochondrial pathway [95]. Table 2 reports the results of the main in vitro studies.
The use of hawthorn induced anti-inflammatory properties through the modulation of lipopolysaccharide-induced pro-inflammatory (Interleukin-6 and Tumor necrosis factor-α) and anti-inflammatory (Interleukin-10) cytokines [96]. The flavonoids extracted from hawthorn could treat inflammatory bowel disease via the prevention of the nuclear factor kappa-light-chain-enhancer of activated B cells and extra cellular signal-regulated kinase 1/2 activity, the suppression of myosin light chain kinase and phosphorylatedmyosin light chain upregulation, the suppression of the production of inflammatory cytokines in Caco-2 cells, and the alleviation of inflammatory cytokine-induced intestinal barrier deficit [97].
The administration of C. orientalis berries and leaves at the concentration of 0.4 mg/mL displayed a DPPH radical scavenging effect and anti-inflammatory activity via the inhibition of 12- lipoxygenase (12-LOX) and cyclooxygenase-1 (COX-1), thereby impeding the generation of thromboxane B2 (up to 55.2%) and 12-Hydroxyheptadecatrienoic acid (up to 68.9%) [98]. In a study by Wyspianska et al., the procyanidins obtained from hawthorn bark extract revealed anti-inflammatory and antioxidant properties [99]. Furthermore, neolignans obtained from the ethanolic extract of hawthorn seeds exhibited anti-inflammatory and antioxidant properties, most likely due to the prevention of tumor necrosis factor-α) via the compounds 7′,8′-threo,7S,8R-1-[4- [(2-hydroxy-2-(4-hydroxy-3-methoxyphenyl)-1-(hydroxymethyl)ethoxy]-3-methoxyphenyl]-1,2,3-propanetriol and 7′,8′-threo,7R, 8R-1-[4-[(2-hydroxy-2-(4-hydroxyl-3-methoxyphenyl)-1-(hydro-xymethyl)ethoxy]-3-methoxyphenyl]-1,2,3-propanetriol, and the inhibition of NO production via leptolepisol D [100].
The antioxidant and anti-inflammatory bioassay-guided fractionation of the seed extract of mountain hawthorn, C. pinnatifida, led to the isolation of eight new lignans called hawthornnins, which showed different promising activities by scavenging free radicals and inhibiting TNF-α and NO production [101]. Zhao et al. observed α-glucosidase inhibitory and antioxidant activity for C. pinnatifida fruit [102]. In another study, 8-O-4′ neolignans extracted from C. pinnatifida seeds blocked the activity of tyrosinase by 66.67%, in addition to exhibiting antioxidant activity [103]. Among the triterpenoids extracted from hawthorn berries, the compounds 3β,6β,18β,23-tetrahydroxy-olean-12-en-28-oic acid, 2α,3β,6β,18β-tetrahydroxy-olean-12-en-28-oic acid, and 2α,3β,6β,18β,23-pentahydroxy-olean-12-en-28-oic acid had antioxidant functions and could inhibit the proliferation of MCF-7 and HepG2 cells (EC50 = <5 µM) [104]. In a study by Chai et al. the proanthocyanidin compounds extracted from Chinese hawthorn fruits were characterized by HPLC-ESI-MS and MALDI-TOF-MS and examined for their bioactivities. The results showed anti-tyrosinase properties by preventing tyrosinases such as diphenolase and monophenolase and antioxidant activity [105].
Hawthorn seed extract at a concentration of 50 μM protected SH-SY5Y cells from damage through cell apoptosis prevention due to the presence of a sesquineolignan compound, 7″,8″-erythro;7R,8R,7′R,8′S)-3,7,3′,5′,3″-pentamethoxy-4-hydroxy-4′,8-oxy-4″,7′-epoxy-8′,5″sesquineolignan-9,9′,7″,8″,9″-pentanol, which was found to have a neuroprotective effect [106].
The extractions of C. pinnatifida fructus and Rhodiolae kirliowii radix and rhizome showed antiviral potential towards infection by the human polyomaviruses BK (BKPyV) and JC (JCPyV) by reducing the expression of viral proteins in the infected cells [107]. The growth of pathogenic S. aureus and E. coli was inhibited by gold and silver chloride nanoparticles functionalized by fruit extract of C. pinnatifida, which also scavenged DPPH free radicals and showed anti-inflammatory function via a reduction in the levels of inflammatory cytokines such as prostaglandin E2 (PGE2) and NO [108].

3.2. Health-Promoting Activities of Hawthorn in Animals

Many in vivo investigations have reported different beneficial functions for hawthorn [109,110,111,112,113,114,115,116,117,118]. The administration of hawthorn extract could attenuate atherosclerosis through the prevention of factors related to apoptosis and inflammation signaling pathways, by an apoptosis and inflammation resistance effect, vascular smooth muscle cells calcium deposition, lipidosis, preventing proliferation, lipid regulation, reducing interleukin-1β, hypersensitive C-reactive protein, monocyte chemoattractant protein-1, Bax mRNA expression and protein levels, as well as the enhancement of adiponectin level in serum and Bcl-2 (mRNA and protein expression) in the aorta [119]. In another study, the administration of hawthorn leaf flavonoids (20 mg/kg) to apo-lipoprotein E (apoE) knock-out mice for 16 weeks showed an improvement in atherosclerosis via the in vivo promotion of reverse cholesterol transport, the inhibition of foam cell synthesis, and the induction of antioxidant-related gene expression [120]. In a recent study, ethanolic hawthorn fruit extract in hypocholesterolemic rats exposed vascular protective activities due to the phenolic compounds with reactive oxygen species scavenging and cholesterol-lowering activities, resulting in high cholesterol intake and bile acid production via the upregulation of hepatic CYP7A1 mRNA expression [121]. The co-administration of resveratrol with hawthorn flavonoids following coronary artery bypass graft could decrease thrombotic restenosis and endothelial cell injury [122]. The cardioprotective role of hawthorn leaf extract in rats was attributed to some functions, including the enhancement of the antioxidant defense system, the improvement of heart antioxidant biomarkers, the elevation of inflammatory cytokine biomarkers, and the enhancement of serum parameters related to heart function [123]. Anti-inflammation and anti-oxidative stress effects for hawthorn leaf flavonoids through the suppression of PKC-α activation in rats with diabetes-induced cardiomyopathy has also been reported [124]. Alp et al. reported that C. oxyacantha alcoholic extract (40 µg/kg/min of digoxin) showed antiarrhythmic activity in rats [125]. The alcoholic extract of C. oxyacantha berries was given to rats with isoproterenol-induced myocardial infarction, and anti-apoptotic and anti-inflammatory functions were found as a result of reducing nitritive stress, lipid peroxidation and apoptotic processes [126]. Table 3 reports the main studies in animals.
A study reported anti-melanogenesis, antioxidant and antitumor roles for hawthorn extract. The treatment of tumor-implanted mice with total oligomer flavonoids from hawthorn extract (150 mg/kg body weight) for 21 days reduced the tumor weight and volume, prevented intracellular free radical scavenging activity, decreased the melanin production and blocked the tyrosinase in melanoma cells [127]. Yonekubo et al. observed that the use of different concentrations of C. oxyacantha fruit extracts for a week in mice induced genotoxicity activity [128].
The co-treatment of type I diabetes-induced rats by hawthorn extract (100 mg/kg per day), plus resistance training for five days/week for 10 consecutive weeks, improved memory and learning by decreasing lipid peroxidation and increasing total antioxidant capacity [129]. In another study, the administration of C. oxyacantha leaves (200 mg/kg and 400 mg/kg) improved memory and learning in rats with scopolamine-induced amnesia through the inhibition of dementia and oxidative damage [130]. Lee et al. observed that the administration of ethanol extract of C. pinnatifida fruits could treat Alzheimer’s disease by inhibiting amyloid β accumulation [131].
The treatment of high-fat-diet-fed rats with L. plantarum grade A pasteurized milk ordinance -fermented hawthorn juice for 28 days showed hypolipidemic activity through the regulation of adipose tissues and liver morphology, the restoration of liver tissue and the reduction in low-density lipoprotein cholesterol, serum total cholesterol, lipid vacuolization and lipid metabolism levels [132]. The administration of C. pinnatifida with high-fat-diet-induced obese mice modulated the gut microbiota activity by reducing serum triglyceride, decreasing fat and body weight, inhibiting adipogenesis and inflammation, and altering gut microbial abundance and diversity [133]. In a recent study, the use of different concentrations of HT048 (obtained from the extractions of Citrus unshiu peel plus C. pinnatifida leaves) in rats resulted in an anti-obesity effect after 12 weeks by dose-dependently suppressing the differentiation of adipocytes and the release of stimulated glycerol, reducing peroxisome proliferator-activated receptor-gamma and CCAAT/enhancer binding protein-alpha mRNA expression, decreasing body weight, lowering the serum lipid content, reducing hepatic lipogenesis-related gene expression and increasing β-oxidation-related gene expression, thereby indicating positive effects of HT048 to prevent obesity by blocking adipogenesis and lipogenesis [134].
Diabetic nephropathy was improved in rats treated with hawthorn leaf flavonoids through the improvement of renal function and the reduction of renal damage via a decrease in oxidative stress injury and the regulation of the p38/MAPK signaling pathway [135]. In another study, the methanolic extract of C. oxyacantha (100 mg/kg BW) in rats for 12 weeks treated hyperglycemia and dyslipidemia [136]. Aierken et al. treated rats with streptozotocin-induced type II diabetes mellitus with different concentrations of hawthorn extracts and reported hypoglycemic activity in the treatment animals via the elevation of pancreatic-released plasma insulin and by the reduction of total cholesterol, triglyceride and glucose levels in the blood [137].
Hawthorn showed hepatoprotective effects in rats with alcoholic liver damage via the reduction of LDL and total cholesterol levels, the regulation of serum lipids as triglycerides, the reduction of sinusoidal distension, congestion, necrosis, steatosis and fibrosis, as well as the reduction of cell damage markers (acid phosphatase, γ-glutamyltranspeptidase, alanine aminotransferase and aspartate aminotransferase). Furthermore, hawthorn exhibited antioxidant activity via the elimination of bilirubin, the regulation of glycogen levels in liver tissue, the elevation of serum total antioxidant capacity levels and the reduction of lipid peroxidation [138]. Li et al. [139] reported that the daily administration of flavonoids extracted from hawthorn leaf (50 mg/kg/day and 100 mg/kg/day) for three months reduced hepatic steatosis in rats with non-alcoholic fatty liver disease induced by a high fat diet due to the activation of the adiponectin/AMPK pathway. The use of hawthorn pectin pentaglaracturonide (150 mg/kg/day and 300 mg/kg/day) for 10 weeks in high-fat-diet-fed mice inhibited hepatic lipid accumulation and prevented hepatic fatty acid synthesis by reducing the gene expression of high-fat-diet-induced sterol regulatory element binding factor-1c, pyruvate kinase, acetyl-CoA carboxylase and fatty acid synthase [140].
In a study by Mustafa et al., the antioxidant activity and the immunomodulatory potential were seen for the hyperoside and ethyl acetate extractions of C. azarolus leaves on macrophages, cytotoxic T lymphocytes and natural killer cells [141]. Elango et al. [142] reported an immunomodulatory role for the ethanolic extract of hawthorn (100 mg/kg) in stroke rats over 15 days due to diminished brain apoptosis during reperfusion through the expression of Bcl-xL, the phosphorylation of signal transducer and activator of transcription 3, the elevation of the regulatory T cell (Treg) population and the prevention of activated inflammatory cells via increased levels of Foxp3-positive Tregs and IL-10, and reduced pro-inflammatory immune responses to ischemia and reperfusion-induced damage.
The daily use of hawthorn extract (100 mg/kg/day) for 11 days prevented alveolar bone loss in rats with periodontal disease via the regulation of oxidative stress, total oxidant and serum total antioxidant levels [143]. Others observed that the methanol extract of C. dahurica fruit caused an acceleration of the gastrointestinal tract and activation of the antioxidant system [144].
The polyphenol extract of hawthorn controlled the skin damage induced by UVB radiation via the suppression of p53, the reduction of DNA damage, the elimination of excess ROS, the downregulation of pro-apoptotic BAX and the upregulation of anti-apoptotic BCL-2, thereby preventing apoptosis and suppressing caspase-3/9 activation [145]. In another study, mice experienced the promotion of hair growth by taking C. pinnatifida extract through the induction of anagen phase, by mediating cellular signaling activation resulting in high proliferation and survival rate of human dermal papilla cells, as well as by increasing the Bcl-2/Bax ratio, resulting in protection from cell death [146]. Rats with dehydroepiandrosterone-induced polycystic ovary syndrome experienced protective effects due to the consumption of hawthorn leaf flavonoids [147].

3.3. Health-Promoting Activities of Hawthorn Reported in Clinical Trials

Many clinical trials have reported different health-promoting activities for hawthorn [148,149,150,151,152,153,154]. In a study on 2681 patients suffering from congestive heart failure, the administration of hawthorn extract (900 mg/day) for 620 days reduced the odds ratio of sudden cardiac death in patients with lower left ventricular function [155]. Following the administration of hawthorn (450 mg, twice per day) for six months, 120 ambulatory patients suffering from symptomatic chronic heart showed no positive clinical effects in inflammation, oxidative stress, neurohormones, functional capacity and quality of life measures, but modest change in left ventricular ejection fraction was found [156]. Moeini et al. showed that 5 mL of hawthorn fruit extract after each meal in male and female patients with gastroesophageal reflux disease controlled the main symptoms over four weeks, as well as causing a 94.2% and 93.5% alleviation in regurgitation and heartburn, respectively [157]. According to the findings of Trexler et al. [158], 160 mg of hawthorn supplementation in adult subjects for a week could not influence electrocardiographic indices. In another study, adolescent subjects experienced hypertension following the supplementation of ethanolic extract of fresh Crataegus berries and natural D-camphor (Korodin®) [159]. Similarly, in a study by Erfurt et al. [160], sphygmomanometric blood pressure measurements before and after intervention confirmed the hypertension. In a recent clinical trial, a greater reduction was observed in the diastolic blood pressure in patients with type 2 diabetes over 16 weeks following daily consumption of 1200 mg of hawthorn extract [161]. Mildly hypertensive patients taking hawthorn extract (500–600 mg/day) over 10 weeks caused a decrease in both diastolic and systolic blood pressure [162]. The short-term use of camphor from Crataegus berry extract in women enhanced mental performance and blood pressure [163]. In Table 4, we list the reported examples of studies in humans involving hawthorn.

4. Conclusions and Future Remarks

Medicinal herbs, including hawthorn, are rich sources of high market impact medicines around the world due to the presence of significant amounts of naturally occurring bioactive chemical compounds with therapeutic properties. However, further in vivo and in vitro research and clinical trials are needed to evaluate the link between the chemical compositions of such plants, particularly hawthorn, and their mechanisms of action in the treatment of various diseases. An emerging direction is suggested by the possible use of nanonutraceuticals, assuring their nutraceutical value at a nano level as well as safety and efficacy [164,165,166,167,168]. Nutraceutical science represents a great challenge for the future [169,170,171,172].

Author Contributions

A.N., M.L. and A.S. conceived and designed the work. A.N., M.L., A.D., M.Z., E.B.S. and A.S. wrote the work. A.N., A.D., S.C., S.B.S., A.M.S. and P.S. validated and elaborated data information and figures. A.N., M.L., A.D., M.Z., S.C., S.B.S., A.M.S., P.S., E.B.S., and A.S. have made a substantial contribution to the revision of work and approved it for publication. All authors have read and agreed to the published version of the manuscript.

Funding

The authors acknowledge the support of the research project: Nutraceutica come supporto nutrizionale nel paziente oncologico, CUP: B83D18000140007. E. B. Souto acknowledges the sponsorship of the projects M-ERA-NET-0004/2015-PAIRED and UIDB/04469/2020 (strategic fund), receiving support from the Portuguese Science and Technology Foundation, Ministry of Science and Education (FCT/MEC) through national funds, and co-financed by FEDER, under the Partnership Agreement PT2020.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Santini, A.; Novellino, E. Nutraceuticals: Beyond the diet before the drugs. Curr. Bioact. Compd. 2014, 10, 1–12. [Google Scholar] [CrossRef]
  2. Durazzo, A. Extractable and Non-extractable polyphenols: An overview. In Non-Extractable Polyphenols and Carotenoids: Importance in Human Nutrition and Health; Saura-Calixto, F., Pérez-Jiménez, J., Eds.; Royal Society of Chemistry: London, UK, 2018; pp. 37–45. [Google Scholar]
  3. Durazzo, A.; Lucarini, M.; Kiefer, J.; Mahesar, S.A. State-of-the-Art Infrared Applications in Drugs, Dietary Supplements, and Nutraceuticals. J. Spectrosc. 2020, 2020, 1397275. [Google Scholar] [CrossRef]
  4. Durazzo, A.; Lucarini, M. The State of Science and Innovation of Bioactive Research and Applications, Health and Diseases. Front. Nutr. 2019, 6, 178. [Google Scholar] [CrossRef] [PubMed]
  5. Santini, A.; Novellino, E.; Armini, V.; Ritieni, A. State of the art of Ready-to-Use Therapeutic Food: A tool for nutraceuticals addition to foodstuff. Food Chem. 2013, 140, 843–849. [Google Scholar] [CrossRef]
  6. Durazzo, A.; Lucarini, M.; Novellino, E.; Souto, E.B.; Daliu, P.; Santini, A. Abelmoschus esculentus (L.): Bioactive Components’ Beneficial Properties—Focused on Antidiabetic Role—For Sustainable Health Applications. Molecules 2019, 24, 38. [Google Scholar] [CrossRef] [Green Version]
  7. Lucarini, M.; Durazzo, A.; Kiefer, J.; Santini, A.; Lombardi-Boccia, G.; Souto, E.B.; Romani, A.; Lampe, A.; Ferrari Nicoli, S.; Gabrielli, P. Grape Seeds: Chromatographic Profile of Fatty Acids and Phenolic Compounds and Qualitative Analysis by FTIR-ATR Spectroscopy. Foods 2020, 9, 10. [Google Scholar] [CrossRef] [Green Version]
  8. Salehi, B.; Venditti, A.; Sharifi-Rad, M.; Kręgiel, D.; Sharifi-Rad, J.; Durazzo, A.; Lucarini, M.; Santini, A.; Souto, E.B.; Novellino, E. The therapeutic potential of apigenin. Int. J. Mol. Sci. 2019, 20, 1305. [Google Scholar] [CrossRef] [Green Version]
  9. Durazzo, A.; Lucarini, M.; Souto, E.B.; Cicala, C.; Caiazzo, E.; Izzo, A.A.; Novellino, E.; Santini, A. Polyphenols: A concise overview on the chemistry, occurrence, and human health. Phytother. Res. 2019, 33, 2221–2243. [Google Scholar] [CrossRef] [Green Version]
  10. Abenavoli, L.; Izzo, A.A.; Milić, N.; Cicala, C.; Santini, A.; Capasso, R. Milk thistle (Silybum marianum): A concise overview on its chemistry, pharmacological, and nutraceutical uses in liver diseases. Phytother. Res. 2018, 32, 2202–2213. [Google Scholar] [CrossRef]
  11. Santini, A.; Tenore, G.C.; Novellino, E. Nutraceuticals: A paradigm of proactive medicine. Eur. J. Pharm. Sci. 2017, 96, 53–61. [Google Scholar] [CrossRef]
  12. Daliu, P.; Santini, A.; Novellino, E. A decade of nutraceutical patents: Where are we now in 2018? Expert Opin. Ther. Pat. 2018, 28, 875–882. [Google Scholar] [CrossRef] [PubMed]
  13. Santini, A.; Novellino, E. Nutraceuticals-shedding light on the grey area between pharmaceuticals and food. Expert Rev. Clin. Pharmacol. 2018, 11, 545–547. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Bircher, J.; Hahn, E.G. Understanding the nature of health: New perspectives for medicine and public health. Improved wellbeing at lower costs: New Perspectives for Medicine and Public Health: Improved Wellbeing at lower Cost. F1000Res. 2016, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Santini, A.; Cammarata, S.M.; Capone, G.; Ianaro, A.; Tenore, G.C.; Pani, L.; Novellino, E. Nutraceuticals: Opening the debate for a regulatory framework. Br. J. Clin. Pharmacol. 2018, 84, 659–672. [Google Scholar] [CrossRef] [Green Version]
  16. Daliu, P.; Santini, A.; Novellino, E. From pharmaceuticals to nutraceuticals: Bridging disease prevention and management. Expert Rev. Clin. Pharmacol. 2019, 12, 1–7. [Google Scholar] [CrossRef]
  17. Durazzo, A.; D’Addezio, L.; Camilli, E.; Piccinelli, R.; Turrini, A.; Marletta, L.; Marconi, S.; Lucarini, M.; Lisciani, S.; Gabrielli, P. From plant compounds to botanicals and back: A current snapshot. Molecules 2018, 23, 1844. [Google Scholar] [CrossRef] [Green Version]
  18. Durazzo, A.; Camilli, E.; D’Addezio, L.; Piccinelli, R.; Mantur-Vierendeel, A.; Marletta, L.; Finglas, P.; Turrini, A.; Sette, S. Development of Dietary Supplement Label Database in Italy: Focus of FoodEx2 Coding. Nutr. 2020, 12, 89. [Google Scholar] [CrossRef] [Green Version]
  19. WHO (World Health Organization). 2013. Available online: http://www.who.int/traditional-complementary-integrative-medicine/publications/trm_strategy14_23/en/ (accessed on 5 May 2020).
  20. Attard, E.; Attard, H. Chapter 3.25-Hawthorn: Crataegus oxyacantha, Crataegus monogyna and related species. In Nonvitamin and Nonmineral Nutritional Supplements; Nabavi, S.M., Silva, A.S., Eds.; Academic Press: Cambridge, MA, USA, 2019. [Google Scholar]
  21. European Medicines Agency. 2016. Available online: http://www.ema.europa.eu/ema/index.jspcurl=pages/medicines/herbal/medicines/herbal_med_000061.jsp&mid=WC0b01ac058001fa1d (accessed on 5 May 2020).
  22. European Pharmacopoeia. Europäisches Arzneibuch; Deutscher Apotheker Verlag: Stuttgart, Germany, 2017; Volume 9.0, pp. 2359–2360. [Google Scholar]
  23. Venskutonis, P. Phytochemical composition and bioactivities of hawthorn (Crataegus spp.): Review of recent research advances. J. Food Bioact. 2018, 4. [Google Scholar] [CrossRef] [Green Version]
  24. Wang, C. Crataegus pinnatifida Bge. 山楂 (Shanzha, Hawthorn Fruit). In Dietary Chinese Herbs, 1st ed.; Liu, Y., Wang, Z., Zhang, J., Eds.; Springer: Berlin/Heidelberg, Germany, 2015; pp. 355–361. [Google Scholar]
  25. Wu, J.; Peng, W.; Qin, R.; Zhou, H. Crataegus pinnatifida: Chemical constituents, pharmacology, and potential applications. Molecules 2014, 19, 1685–1712. [Google Scholar] [CrossRef]
  26. Orhan, I.E. Phytochemical and Pharmacological Activity Profile of Crataegus oxyacantha L. (Hawthorn)-A Cardiotonic Herb. Curr. Med. Chem. 2018, 25, 4854–4865. [Google Scholar] [CrossRef]
  27. Benabderrahmane, W.; Lores, M.; Lamas, J.P.; Benayache, S. Matrix solid-phase dispersion as a tool for phytochemical and bioactivities characterisation: Crataegus oxyacantha L. A case study. Nat. Prod. Res. 2018, 32, 1220–1223. [Google Scholar] [CrossRef]
  28. Benabderrahmane, W.; Lores, M.; Benaissa, O.; Lamas, J.P.; de Miguel, T.; Amrani, A.; Benayache, F.; Benayache, S. Polyphenolic content and bioactivities of Crataegus oxyacantha L. (Rosaceae). Nat. Prod. Res. 2019, 1–6. [Google Scholar] [CrossRef]
  29. Cuevas-Durán, R.E.; Medrano-Rodríguez, J.C.; Sánchez-Aguilar, M.; Soria-Castro, E.; Rubio-Ruíz, M.E.; Valle-Mondragón, D.; Sánchez-Mendoza, A.; Torres-Narvaéz, J.C.; Pastelín-Hernández, G.; Ibarra-Lara, L. Extracts of Crataegus oxyacantha and Rosmarinus officinalis attenuate ischemic myocardial damage by decreasing oxidative stress and regulating the production of cardiac vasoactive agents. Int. J. Mol. Sci. 2017, 18, 2412. [Google Scholar] [CrossRef] [Green Version]
  30. Alirezalu, A.; Ahmadi, N.; Salehi, P.; Sonboli, A.; Alirezalu, K.; Mousavi Khaneghah, A.; Barba, F.J.; Munekata, P.E.; Lorenzo, J.M. Physicochemical Characterization, Antioxidant Activity, and Phenolic Compounds of Hawthorn (Crataegus spp.) Fruits Species for Potential Use in Food Applications. Foods 2020, 9, 436. [Google Scholar] [CrossRef] [Green Version]
  31. Ngoc, P.C.; Leclercq, L.; Rossi, J.C.; Desvignes, I.; Hertzog, J.; Fabiano-Tixier, A.S.; Chemat, F.; Schmitt-Kopplin, P.; Cottet, H. Optimizing Water-Based Extraction of Bioactive Principles of Hawthorn: From Experimental Laboratory Research to Homemade Preparations. Molecules 2019, 24, 4420. [Google Scholar] [CrossRef] [Green Version]
  32. Lin, C.; Luque, R. Renewable Resources for Biorefineries; Royal Society of Chemistry: London, UK, 2014; pp. 1–216. [Google Scholar]
  33. Zuin, V.G.; Ramin, L.Z. Green and sustainable separation of natural products from agro-industrial waste: Challenges, potentialities, and perspectives on emerging approaches. In Chemistry and Chemical Technologies in Waste Valorization; Springer: Berlin/Heidelberg, Germany, 2018; pp. 229–282. [Google Scholar]
  34. Hu, Y.H.; Peng, L.Q.; Wang, Q.Y.; Yang, J.; Dong, X.; Wang, S.L.; Cao, J.; Liu, F.M. Ecofriendly microwave-assisted reaction and extraction of bioactive compounds from hawthorn leaf. Phytochem. Anal. 2019, 30, 710–719. [Google Scholar] [CrossRef]
  35. Sydora, N.V.; Kovalyova, A.M.; Iakovenko, V.K. The study of the carbohydrate composition of hawthorn fruits. News Pharm. 2018, 3, 14–18. [Google Scholar] [CrossRef] [Green Version]
  36. Zhao, Y.; Wang, Y.; Wang, J.; Wu, Z.; Sun, Z.; Tian, T.; Niu, H.; Jing, L.; Fang, Z.; Yang, J. Characterization of volatile constituents of Chinese hawthorn (Crataegus spp.) Fruit Juices. In Advances in Applied Biotechnology; Springer: Berlin/Heidelberg, Germany, 2015; pp. 533–545. [Google Scholar]
  37. Salmanian, S.; Sadeghi, M.A.; Alami, M.; Ghorbani, M. Phenolic content, antiradical, antioxidant, and antibacterial properties of hawthorn (Crataegus elbursensis) seed and pulp extract. J. Agric. Sci. Technol. 2014, 16, 343–354. [Google Scholar]
  38. Liu, P.; Kallio, H.; Yang, B. Phenolic compounds in hawthorn (Crataegus grayana) fruits and leaves and changes during fruit ripening. J. Agric. Food Chem. 2011, 59, 11141–11149. [Google Scholar] [CrossRef]
  39. Lund, J.A.; Brown, P.N.; Shipley, P.R. Quantification of North American and European Crataegus flavonoids by nuclear magnetic resonance spectrometry. Fitoterapia 2020, 143, 104537. [Google Scholar] [CrossRef]
  40. Rao, H.; Li, P.; Wu, H.; Liu, C.; Peng, W.; Su, W. Simultaneous Determination of Six Compounds in Destructive Distillation Extracts of Hawthorn Seed by GC-MS and Evaluation of Their Antimicrobial Activity. Molecules 2019, 24, 4328. [Google Scholar] [CrossRef] [Green Version]
  41. Sagaradze, V.A.; Babaeva, E.Y.; Ufimov, R.A.; Trusov, N.A.; Kalenikova, E.I. Study of the variability of rutin, vitexin, hyperoside, quercetin in “Crataegi folium cum flore” of hawthorn (Crataegus L.) species from Russian flora. J. Appl. Res. Med. Aromat. Plants 2019, 15, 100217. [Google Scholar] [CrossRef]
  42. Kurkina, A. Determination of total flavonoids in siberian hawthorn fruit. Pharm. Chem. J. 2015, 48, 800–803. [Google Scholar] [CrossRef]
  43. Zhao, P.; Guo, R.; Zhang, Y.-Y.; Zhang, H.; Yao, G.-D.; Lin, B.; Wang, X.-B.; Huang, X.-X.; Song, S.-J. Phenylpropanoid and dibenzofuran derivatives from Crataegus pinnatifida with antiproliferative activities on hepatoma cells. Bioorg. Chem. 2019, 93, 103354. [Google Scholar] [CrossRef]
  44. Mraihi, F.; Fadhil, H.; Trabelsi-Ayadi, M.; Chérif, J.K. Chemical characterization by HPLC-DAD-ESI/MS of flavonoids from hawthorn fruits and their inhibition of human tumor growth. J. New Sci. 2015, JS-INAT, 840–846. [Google Scholar]
  45. Huang, X.X.; Zhou, C.C.; Li, L.Z.; Li, F.F.; Lou, L.L.; Li, D.M.; Ikejima, T.; Peng, Y.; Song, S.J. The cytotoxicity of 8-O-4’ neolignans from the seeds of Crataegus pinnatifida. Bioorg. Med. Chem. Lett. 2013, 23, 5599–5604. [Google Scholar] [CrossRef]
  46. Gao, P.Y.; Li, L.Z.; Liu, K.C.; Sun, C.; Sun, X.; Wu, Y.N.; Song, S.J. Natural terpenoid glycosides with in vitro/vivo antithrombotic profiles from the leaves of Crataegus pinnatifida. RSC Adv. 2017, 7, 48466–48474. [Google Scholar] [CrossRef] [Green Version]
  47. Abu-Gharbieh, E.; Shehab, N.G. Therapeutic potentials of Crataegus azarolus var. eu-azarolus Maire leaves and its isolated compounds. BMC Complement. Altern. Med. 2017, 17, 218. [Google Scholar] [CrossRef]
  48. Guo, R.; Lv, T.M.; Han, F.Y.; Lin, B.; Yao, G.D.; Wang, X.B.; Huang, X.X.; Song, S.J. Chiral resolution and neuroprotective activities of enantiomeric dihydrobenzofuran neolignans from the fruit of Crataegus pinnatifida. Bioorg. Chem. 2019, 85, 469–474. [Google Scholar] [CrossRef]
  49. Zhao, P.; Zhang, H.; Han, F.Y.; Guo, R.; Huang, S.W.; Lin, B.; Huang, X.X.; Song, S.J. Chiral resolution and neuroprotective activities of enantiomeric 8-O-4′ neolignans from the fruits of Crataegus pinnatifida Bge. Fitoterapia 2019, 136, 104164. [Google Scholar] [CrossRef]
  50. González Jiménez, F.E.; Salazar Montoya, J.A.; Calva-Calva, G.; Ramos-Ramírez, E. Phytochemical Characterization, In Vitro Antioxidant Activity, and Quantitative Analysis by Micellar Electrokinetic Chromatography of Hawthorn (Crataegus pubescens) Fruit. J. Food Qual. 2018, 2018, 1–11. [Google Scholar] [CrossRef] [Green Version]
  51. Luo, M.; Hu, J.Y.; Song, Z.Y.; Jiao, J.; Mu, F.S.; Ruan, X.; Gai, Q.Y.; Qiao, Q.; Zu, Y.G.; Fu, Y.J. Optimization of ultrasound-assisted extraction (UAE) of phenolic compounds from Crataegus pinnatifida leaves and evaluation of antioxidant activities of extracts. RSC Adv. 2015, 5. [Google Scholar] [CrossRef]
  52. Huang, X.X.; Xu, Y.; Bai, M.; Zhou, L.; Song, S.J.; Wang, X.B. Lignans from the seeds of Chinese hawthorn (Crataegus pinnatifida var major N.E.Br.) against β-amyloid aggregation. Nat. Prod. Res. 2018, 32, 1706–1713. [Google Scholar] [CrossRef]
  53. Durazzo, A.; Lucarini, M. A current shot and re-thinking of antioxidant research strategy. Braz. J. Anal. Chem. 2018, 5, 9–11. [Google Scholar] [CrossRef]
  54. Durazzo, A.; Lucarini, M. Extractable and non-extractable antioxidants. Molecules 2019, 24, 1933. [Google Scholar] [CrossRef] [Green Version]
  55. Ganie, S.A.; Ali Dar, T.; Zargar, S.; Bhat, A.H.; Dar, K.B.; Masood, A.; Zargar, M.A. Crataegus songarica methanolic extract accelerates enzymatic status in kidney and heart tissue damage in albino rats and its in vitro cytotoxic activity. Pharm. Biol. 2016, 54, 1246–1254. [Google Scholar] [CrossRef] [Green Version]
  56. Gao, Z.; Xie, M.; Wang, N.; Chen, L.; Huang, X. Effects of combination treatment of metformin and hawthorn in patients with prediabetes complicated by nonalcoholic fatty liver disease. Int. J. Clin. Exp. Med. 2019, 12, 1979–1984. [Google Scholar]
  57. Pawlaczyk-Graja, I. Polyphenolic-polysaccharide conjugates from flowers and fruits of single-seeded hawthorn (Crataegus monogyna Jacq.): Chemical profiles and mechanisms of anticoagulant activity. Int. J. Biol. Macromol. 2018, 116, 869–879. [Google Scholar] [CrossRef]
  58. Cloud, A.M.E.; Vilcins, D.; McEwen, B.J. The effect of hawthorn (Crataegus spp.) on blood pressure: A systematic review. Adv. Integr. Med 2019. [Google Scholar] [CrossRef]
  59. Halver, J.; Wenzel, K.; Sendker, J.; Carrillo García, C.; Erdelmeier, C.A.J.; Willems, E.; Mercola, M.; Symma, N.; Könemann, S.; Koch, E.; et al. Crataegus Extract WS®1442 Stimulates Cardiomyogenesis and Angiogenesis From Stem Cells: A Possible New Pharmacology for Hawthorn? Front. Pharmacol. 2019, 10. [Google Scholar] [CrossRef]
  60. Ranjbar, K.; Zarrinkalam, E.; Salehi, I.; Komaki, A.; Fayazi, B. Cardioprotective effect of resistance training and Crataegus oxyacantha extract on ischemia reperfusion-induced oxidative stress in diabetic rats. Biomed. Pharmacother. 2018, 100, 455–460. [Google Scholar] [CrossRef] [PubMed]
  61. Pahlavan, S.; Tousi, M.S.; Ayyari, M.; Alirezalu, A.; Ansari, H.; Saric, T.; Baharvand, H. Effects of hawthorn (Crataegus pentagyna) leaf extract on electrophysiologic properties of cardiomyocytes derived from human cardiac arrhythmia-specific induced pluripotent stem cells. FASEB J. 2018, 32, 1440–1451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Fuchs, S.; Bischoff, I.; Willer, E.; Bräutigam, J.; Bubik, M.; Erdelmeier, C.; Koch, E.; Faleschini, M.; Mieri, M.; Bauhart, M.; et al. The Dual Edema-Preventing Molecular Mechanism of the Crataegus Extract WS 1442 Can Be Assigned to Distinct Phytochemical Fractions. Planta Med. 2016, 83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Yoo, J.H.; Liu, Y.; Kim, H.S. Hawthorn Fruit Extract Elevates Expression of Nrf2/HO-1 and Improves Lipid Profiles in Ovariectomized Rats. Nutrients 2016, 8, 283. [Google Scholar] [CrossRef] [Green Version]
  64. Diane, A.; Borthwick, F.; Wu, S.; Lee, J.; Brown, P.N.; Dickinson, T.A.; Croft, K.D.; Vine, D.F.; Proctor, S.D. Hypolipidemic and cardioprotective benefits of a novel fireberry hawthorn fruit extract in the JCR:LA-cp rodent model of dyslipidemia and cardiac dysfunction. Food Funct. 2016, 7, 3943–3952. [Google Scholar] [CrossRef]
  65. Hu, H.J.; Luo, X.G.; Dong, Q.Q.; Mu, A.; Shi, G.L.; Wang, Q.T.; Chen, X.Y.; Zhou, H.; Zhang, T.C.; Pan, L.W. Ethanol extract of Zhongtian hawthorn lowers serum cholesterol in mice by inhibiting transcription of 3-hydroxy-3-methylglutaryl-CoA reductase via nuclear factor-kappa B signal pathway. Exp. Biol. Med. 2016, 241, 667–674. [Google Scholar] [CrossRef] [Green Version]
  66. Kalantari, H.; Hemmati, A.A.; Foruozandeh, H.; Kalantar, M.; Aghel, N.; Aslani, M.; Ehsan, T. Healing Effect of Hawthorn (Crataegus pontica C. Koch) Leaf Extract in Dermal Toxicity Induced by T-2 Toxin in Rabbit. Jundishapur J. Nat. Pharm. Prod. 2016, 11, e35688. [Google Scholar] [CrossRef] [Green Version]
  67. Dehghani, S.; Mehri, S.; Hosseinzadeh, H. The effects of Crataegus pinnatifida (Chinese hawthorn) on metabolic syndrome: A review. Iran. J. Basic Med. Sci. 2019, 22, 460–468. [Google Scholar] [CrossRef]
  68. Zhu, R.G.; Sun, Y.D.; Hou, Y.T.; Fan, J.G.; Chen, G.; Li, T.P. Pectin penta-oligogalacturonide reduces cholesterol accumulation by promoting bile acid biosynthesis and excretion in high-cholesterol-fed mice. Chem. Biol. Interact. 2017, 272, 153–159. [Google Scholar] [CrossRef]
  69. Wu, M.; Liu, L.; Xing, Y.; Yang, S.; Li, H.; Cao, Y. Roles and Mechanisms of Hawthorn and Its Extracts on Atherosclerosis: A Review. Front. Pharmacol. 2020, 11. [Google Scholar] [CrossRef]
  70. Shatoor, A.S.; Al Humayed, S. The Protective Effect of Crataegus aronia Against High-Fat Diet-Induced Vascular Inflammation in Rats Entails Inhibition of the NLRP-3 Inflammasome Pathway. Cardiovasc. Toxicol. 2020, 20, 82–99. [Google Scholar] [CrossRef] [PubMed]
  71. Pashaie, B.; Hobbenaghi, R.; Malekinejad, H. Anti-atherosclerotic effect of Cynodon dactylon extract on experimentally induced hypercholesterolemia in rats. Vet. Res. Forum 2017, 8, 185–193. [Google Scholar] [PubMed]
  72. Zhu, R.; Li, T.; Dong, Y.; Liu, Y.; Li, S.; Chen, G.; Zhao, Z.; Jia, Y. Pectin pentasaccharide from hawthorn (Crataegus pinnatifida Bunge. Var major) ameliorates disorders of cholesterol metabolism in high-fat diet fed mice. Food Res. Int. 2013, 54, 262–268. [Google Scholar] [CrossRef]
  73. Hwang, E.; Park, S.Y.; Yin, C.S.; Kim, H.T.; Kim, Y.M.; Yi, T.H. Antiaging effects of the mixture of Panax ginseng and Crataegus pinnatifida in human dermal fibroblasts and healthy human skin. J. Gins. Res. 2017, 41, 69–77. [Google Scholar] [CrossRef] [Green Version]
  74. Ao, N.; Qu, Y.; Zheng, Y.; Cai, Q.; Deng, Y.; Suo, T. Chemical basis of hawthorn processed with honey on myocardial ischaemia protective effect. Food Funct. 2020, 11, 3134–3143. [Google Scholar] [CrossRef]
  75. Niu, Z.; Yan, M.; Zhao, X.; Jin, H.; Gong, Y. Effect of hawthorn seed extract on the gastrointestinal function of rats with diabetic gastroparesis. S. Afr. J. Bot. 2020, 130, 448–455. [Google Scholar] [CrossRef]
  76. Tadić, V.M.; Dobrić, S.; Marković, G.M.; Ðorđević, S.M.; Arsić, I.A.; Menković, N.R.; Stević, T. Anti-inflammatory, Gastroprotective, Free-Radical-Scavenging, and Antimicrobial Activities of Hawthorn Berries Ethanol Extract. J. Agric. Food Chem. 2008, 56, 7700–7709. [Google Scholar] [CrossRef]
  77. Strugała, P.; Gładkowski, W.; Kucharska, A.Z.; Sokół-Łętowska, A.; Gabrielska, J. Antioxidant activity and anti-inflammatory effect of fruit extracts from blackcurrant, chokeberry, hawthorn, and rosehip, and their mixture with linseed oil on a model lipid membrane. Eur. J. Lipid Sci. Technol. 2016, 118, 461–474. [Google Scholar] [CrossRef]
  78. Wang, Y.; Lv, M.; Wang, T.; Sun, J.; Wang, Y.; Xia, M.; Jiang, Y.; Zhou, X.; Wan, J. Research on mechanism of charred hawthorn on digestive through modulating “brain-gut” axis and gut flora. J. Ethnopharmacol. 2019, 245, 112166. [Google Scholar] [CrossRef]
  79. Zheng, X.; Li, X.; Chen, M.; Yang, P.; Zhao, X.; Zeng, L.; OuYang, Y.; Yang, Z.; Tian, Z. The protective role of hawthorn fruit extract against high salt-induced hypertension in Dahl salt-sensitive rats: Impact on oxidative stress and metabolic patterns. Food Funct. 2019, 10, 849–858. [Google Scholar] [CrossRef]
  80. Liu, H.; Liu, J.; Lv, Z.; Yang, W.; Zhang, C.; Chen, D.; Jiao, Z. Effect of dehydration techniques on bioactive compounds in hawthorn slices and their correlations with antioxidant properties. J. Food Sci. Technol. 2019, 56, 2446–2457. [Google Scholar] [CrossRef] [PubMed]
  81. Lou, X.; Yuan, B.; Wang, L.; Xu, H.; Hanna, M.; Yuan, L. Evaluation of physicochemical characteristics, nutritional composition and antioxidant capacity of Chinese organic hawthorn berry (Crataegus pinnatifida). Int. J. Food Sci. Technol. 2019, 55, 1679–1688. [Google Scholar] [CrossRef]
  82. Alirezalu, A.; Salehi, P.; Ahmadi, N.; Sonboli, A.; Aceto, S.; Hatami Maleki, H.; Ayyari, M. Flavonoids profile and antioxidant activity in flowers and leaves of hawthorn species (Crataegus spp.) from different regions of Iran. Int. J. Food Prop. 2018, 21, 452–470. [Google Scholar] [CrossRef] [Green Version]
  83. Wen, L.; Guo, X.; Liu, R.H.; You, L.; Abbasi, A.M.; Fu, X. Phenolic contents and cellular antioxidant activity of Chinese hawthorn “Crataegus pinnatifida”. Food Chem. 2015, 186, 54–62. [Google Scholar] [CrossRef]
  84. Mraihi, F.; Hidalgo, M.; de Pascual-Teresa, S.; Trabelsi-Ayadi, M.; Chérif, J.-K. Wild grown red and yellow hawthorn fruits from Tunisia as source of antioxidants. Arab. J. Chem. 2015, 8, 570–578. [Google Scholar] [CrossRef]
  85. Li, T.; Li, S.; Dong, Y.; Zhu, R.; Liu, Y. Antioxidant activity of penta-oligogalacturonide, isolated from haw pectin, suppresses triglyceride synthesis in mice fed with a high-fat diet. Food Chem. 2014, 145, 335–341. [Google Scholar] [CrossRef]
  86. Ebrahimzadeh, M.; Khalili, M.; Zareh, G.; Farzin, D.; Amin, G. Antihypoxic activities of Crataegus pentaegyn and Crataegus microphylla fruits-an in vivo assay. Braz. J. Pharm. Sci. 2018, 54. [Google Scholar] [CrossRef]
  87. Lim, D.W.; Han, T.; Jung, J.; Song, Y.; Um, M.Y.; Yoon, M.; Kim, Y.T.; Cho, S.; Kim, I.H.; Han, D.; et al. Chlorogenic Acid from Hawthorn Berry (Crataegus pinnatifida Fruit) Prevents Stress Hormone-Induced Depressive Behavior, through Monoamine Oxidase B-Reactive Oxygen Species Signaling in Hippocampal Astrocytes of Mice. Mol. Nutr. Food Res. 2018, e1800029. [Google Scholar] [CrossRef]
  88. Zhang, S.; Zhang, C.; Li, M.; Chen, X.; Ding, K. Structural elucidation of a glucan from Crataegus pinnatifida and its bioactivity on intestinal bacteria strains. Int. J. Biol. Macromol. 2019, 128, 435–443. [Google Scholar] [CrossRef]
  89. Bisignano, C.; Furneri, P.M.; Mandalari, G. In Vitro Efficacy of Crataegus oxycantha L. (Hawthorn) and Its Major Components against ATCC and Clinical Strains of Ureaplasma urealyticum. Adv. Microbiol. 2016, 6, 909–916. [Google Scholar] [CrossRef] [Green Version]
  90. Tsai, S.J.; Yin, M.C. Antioxidative and anti-inflammatory protection of oleanolic acid and ursolic acid in PC12 cells. J. Food Sci. 2008, 73, H174–H178. [Google Scholar] [CrossRef] [PubMed]
  91. Keser, S.; Celik, S.; Turkoglu, S.; Yilmaz, O.; Turkoglu, I. Hydrogen peroxide radical scavenging and total antioxidant activity of hawthorn. Chem. J. 2012, 2, 9–12. [Google Scholar]
  92. Li, C.; Wang, M.H. Anti-inflammatory effect of the water fraction from hawthorn fruit on LPS-stimulated RAW 264.7 cells. Nutr. Res. Pract. 2011, 5, 101–106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Ma, L.; Xu, G.B.; Tang, X.; Zhang, C.; Zhao, W.; Wang, J.; Chen, H. Anti-cancer potential of polysaccharide extracted from hawthorn (Crataegus) on human colon cancer cell line HCT116 via cell cycle arrest and apoptosis. J. Funct. Foods 2020, 64, 103677. [Google Scholar] [CrossRef]
  94. Wu, P.; Li, F.; Zhang, J.; Yang, B.; Ji, Z.; Chen, W. Phytochemical compositions of extract from peel of hawthorn fruit, and its antioxidant capacity, cell growth inhibition, and acetylcholinesterase inhibitory activity. BMC Complement. Altern. Med. 2017, 17, 151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Cui, D.; Liang, T.; Sun, L.; Meng, L.; Yang, C.; Wang, L.; Liang, T.; Li, Q. Green synthesis of selenium nanoparticles with extract of hawthorn fruit induced HepG2 cells apoptosis. Pharm. Biol. 2018, 56, 528–534. [Google Scholar] [CrossRef] [Green Version]
  96. Kmail, A.; Lyoussi, B.; Zaid, H.; Imtara, H.; Saad, B. In vitro evaluation of anti-inflammatory and antioxidant effects of Asparagus aphyllus L., Crataegus azarolus L., and Ephedra alata Decne.in monocultures and co-cultures of HepG2 and THP-1-derived macrophages. Pharmacogn. Commun. 2017, 7, 24–33. [Google Scholar] [CrossRef] [Green Version]
  97. Liu, F.; Zhang, X.; Ji, Y. Total Flavonoid Extract from Hawthorn (Crataegus pinnatifida) Improves Inflammatory Cytokines-Evoked Epithelial Barrier Deficit. Med. Sci. Monit. 2020, 26, e920170. [Google Scholar] [CrossRef]
  98. Savikin, K.P.; Krstic-Milosevic, D.B.; Menkovic, N.R.; Beara, I.N.; Mrkonjic, Z.O.; Pijevijakusic, D.S. Crataegus orientalis Leaves and Berries: Phenolic Profiles, Antioxidant and Anti-inflammatory Activity. Nat. Prod. Commun. 2017, 12, 159–162. [Google Scholar] [CrossRef] [Green Version]
  99. Wyspianska, D.; Kucharska, A.Z.; Sokol-Letowska, A.; Kolniak-Ostek, J. Physico-chemical, antioxidant, and anti-inflammatory properties and stability of hawthorn (Crataegus monogyna Jacq.) procyanidins microcapsules with inulin and maltodextrin. J. Sci. Food Agric. 2017, 97, 669–678. [Google Scholar] [CrossRef]
  100. Peng, Y.; Lou, L.L.; Liu, S.F.; Zhou, L.; Huang, X.X.; Song, S.J. Antioxidant and anti-inflammatory neolignans from the seeds of hawthorn. Bioorg. Med. Chem. Lett. 2016, 26, 5501–5506. [Google Scholar] [CrossRef] [PubMed]
  101. Huang, X.X.; Bai, M.; Zhou, L.; Lou, L.L.; Liu, Q.B.; Zhang, Y.; Li, L.Z.; Song, S.J. Food Byproducts as a New and Cheap Source of Bioactive Compounds: Lignans with Antioxidant and Anti-inflammatory Properties from Crataegus pinnatifida Seeds. J. Agric. Food Chem. 2015, 63, 7252–7260. [Google Scholar] [CrossRef] [PubMed]
  102. Zhao, C.; Miao, J.; Li, X.; Chen, X.; Mao, X.; Wang, Y.; Hua, X.; Gao, W. Impact of in vitro simulated digestion on the chemical composition and potential health benefits of Chaenomeles speciosa and Crataegus pinnatifida. Food Biosci. 2020, 35, 100511. [Google Scholar] [CrossRef]
  103. Huang, X.-X.; Liu, Q.B.; Zhou, L.; Liu, S.; Cheng, Z.-Y.; Sun, Q.; Li, L.-Z.; Song, S.-J. The Antioxidant and Tyrosinase-inhibiting Activities of 8-O-4’Neolignans from Crataegus pinnatifida Seeds. Rec. Nat. Prod. 2015, 9, 305. [Google Scholar]
  104. Qiao, A.; Wang, Y.; Xiang, L.; Zhang, Z.; He, X. Novel triterpenoids isolated from hawthorn berries functioned as antioxidant and antiproliferative activities. J. Funct. Foods 2015, 13, 308–313. [Google Scholar] [CrossRef]
  105. Chai, W.M.; Chen, C.M.; Gao, Y.S.; Feng, H.L.; Ding, Y.M.; Shi, Y.; Zhou, H.T.; Chen, Q.X. Structural analysis of proanthocyanidins isolated from fruit stone of Chinese hawthorn with potent antityrosinase and antioxidant activity. J. Agric. Food Chem. 2014, 62, 123–129. [Google Scholar] [CrossRef]
  106. Huang, X.X.; Ren, Q.; Song, X.Y.; Zhou, L.; Yao, G.D.; Wang, X.B.; Song, S.J. Seven new sesquineolignans isolated from the seeds of hawthorn and their neuroprotective activities. Fitoterapia 2018, 125, 6–12. [Google Scholar] [CrossRef]
  107. Chen, S.Y.; Teng, R.H.; Wang, M.; Chen, P.L.; Lin, M.C.; Shen, C.H.; Chao, C.N.; Chiang, M.K.; Fang, C.Y.; Chang, D. Rhodiolae Kirliowii Radix et Rhizoma and Crataegus pinnatifida Fructus Extracts Effectively Inhibit BK Virus and JC Virus Infection of Host Cells. Evid. Based Complement. Altern. Med. 2017, 2017, 5620867. [Google Scholar] [CrossRef] [Green Version]
  108. Kang, J.P.; Kim, Y.J.; Singh, P.; Huo, Y.; Soshnikova, V.; Markus, J.; Ahn, S.; Chokkalingam, M.; Lee, H.A.; Yang, D.C. Biosynthesis of gold and silver chloride nanoparticles mediated by Crataegus pinnatifida fruit extract: In vitro study of anti-inflammatory activities. Artif. Cells Nanomed. Biotechnol. 2018, 46, 1530–1540. [Google Scholar] [CrossRef]
  109. Wang, T.; Zhang, P.; Zhao, C.; Zhang, Y.; Liu, H.; Hu, L.; Gao, X.; Zhang, D. Prevention effect in selenite-induced cataract in vivo and antioxidative effects in vitro of Crataegus pinnatifida leaves. Biol. Trace Elem. Res. 2011, 142, 106–116. [Google Scholar] [CrossRef]
  110. Niu, C.S.; Chen, C.T.; Chen, L.J.; Cheng, K.C.; Yeh, C.H.; Cheng, J.T. Decrease of blood lipids induced by Shan-Zha (fruit of Crataegus pinnatifida) is mainly related to an increase of PPARα in liver of mice fed high-fat diet. Horm. Metab. Res. 2011, 43, 625–630. [Google Scholar] [CrossRef] [PubMed]
  111. Mohana, T.; Navin, A.V.; Jamuna, S.; Sadullah, M.S.S.; Devaraj, S.N. Inhibition of differentiation of monocyte to macrophages in atherosclerosis by oligomeric proanthocyanidins–In-vivo and in-vitro study. Food Chem. Toxicol. 2015, 82, 96–105. [Google Scholar] [CrossRef]
  112. Qin, R.; Xiao, K.; Li, B.; Jiang, W.; Peng, W.; Zheng, J.; Zhou, H. The combination of catechin and epicatechin gallate from Fructus crataegi potentiates β-lactam antibiotics against methicillin-resistant Staphylococcus aureus (MRSA) in vitro and in vivo. Int. J. Mol. Sci. 2013, 14, 1802–1821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Kao, E.-S.; Wang, C.-J.; Lin, W.-L.; Yin, Y.-F.; Wang, C.-P.; Tseng, T.-H. Anti-inflammatory potential of flavonoid contents from dried fruit of Crataegus pinnatifida in vitro and in vivo. J. Agric. Food Chem. 2005, 53, 430–436. [Google Scholar] [CrossRef] [PubMed]
  114. Hosseinimehr, S.J.; Azadbakht, M.; Mousavi, S.M.; Mahmoudzadeh, A.; Akhlaghpoor, S. Radioprotective effects of hawthorn fruit extract against gamma irradiation in mouse bone marrow cells. J. Radiat. Res. 2006, 48, 63–68. [Google Scholar] [CrossRef]
  115. Zhang, J.; Liang, R.; Wang, L.; Yan, R.; Hou, R.; Gao, S.; Yang, B. Effects of an aqueous extract of Crataegus pinnatifida Bge. var. major NE Br. fruit on experimental atherosclerosis in rats. J. Ethnopharmacol. 2013, 148, 563–569. [Google Scholar]
  116. Koçyõldõz, Z.Ç.; Birman, H.; Olgaç, V.; Akgün-Dar, K.; Melikoğlu, G.; Meriçli, A. Crataegus tanacetifolia leaf extract prevents L-NAME-induced hypertension in rats: A morphological study. Phytother. Res. 2006, 20, 66–70. [Google Scholar] [CrossRef]
  117. Jayalakshmi, R.; Thirupurasundari, C.; Devaraj, S.N. Pretreatment with alcoholic extract of shape Crataegus oxycantha (AEC) activates mitochondrial protection during isoproterenol–induced myocardial infarction in rats. Mol. Cell. Biochem. 2006, 292, 59–67. [Google Scholar] [CrossRef]
  118. Can, Ö.D.; Özkay, Ü.D.; Öztürk, N.; Öztürk, Y. Effects of hawthorn seed and pulp extracts on the central nervous system. Pharm. Biol. 2010, 48, 924–931. [Google Scholar] [CrossRef] [Green Version]
  119. Wang, S.Z.; Wu, M.; Chen, K.J.; Liu, Y.; Sun, J.; Sun, Z.; Ma, H.; Liu, L.T. Hawthorn Extract Alleviates Atherosclerosis through Regulating Inflammation and Apoptosis Related Factors: An Experimental Study. Chin. J. Integr. Med. 2019, 25, 108–115. [Google Scholar] [CrossRef]
  120. Dong, P.; Pan, L.; Zhang, X.; Zhang, W.; Wang, X.; Jiang, M.; Chen, Y.; Duan, Y.; Wu, H.; Xu, Y.; et al. Hawthorn (Crataegus pinnatifida Bunge) leave flavonoids attenuate atherosclerosis development in apoE knock-out mice. J. Ethnopharmacol. 2017, 198, 479–488. [Google Scholar] [CrossRef] [PubMed]
  121. Kwok, C.Y.; Li, C.; Cheng, H.-L.; Ng, Y.F.; Chan, T.Y.; Kwan, Y.W.; Leung, G.P.H.; Lee, S.M.Y.; Mok, D.K.W.; Yu, P.H.F.; et al. Cholesterol lowering and vascular protective effects of ethanolic extract of dried fruit of Crataegus pinnatifida, hawthorn (Shan Zha), in diet-induced hypercholesterolaemic rat model. J. Funct. Foods 2013, 5, 1326–1335. [Google Scholar] [CrossRef]
  122. Zhu, Y.; Feng, B.; He, S.; Su, Z.; Zheng, G. Resveratrol combined with total flavones of hawthorn alleviate the endothelial cells injury after coronary bypass graft surgery. Phytomedicine 2018, 40, 20–26. [Google Scholar] [CrossRef] [PubMed]
  123. Turkistani, A.M. Hawthorn leaves extract suppress the cardiotoxicity-induced by doxorubicin in rats: Mechanistic study. Entomol. Appl. Sci. Lett. 2019, 5, 106–113. [Google Scholar]
  124. Min, Q.; Bai, Y.; Zhang, Y.; Yu, W.; Zhang, M.; Liu, D.; Diao, T.; Lv, W. Hawthorn Leaf Flavonoids Protect against Diabetes-Induced Cardiomyopathy in Rats via PKC-alpha Signaling Pathway. Evid. Based Complement. Altern. Med. 2017, 2017, 2071952. [Google Scholar] [CrossRef] [Green Version]
  125. Alp, H.; Soner, B.C.; Baysal, T.; Sahin, A.S. Protective effects of Hawthorn (Crataegus oxyacantha) extract against digoxin-induced arrhythmias in rats. Anatol. J. Cardiol. 2015, 15, 970–975. [Google Scholar] [CrossRef]
  126. Vijayan, N.A.; Thiruchenduran, M.; Devaraj, S.N. Anti-inflammatory and anti-apoptotic effects of Crataegus oxyacantha on isoproterenol-induced myocardial damage. Mol. Cell. Biochem. 2012, 367, 1–8. [Google Scholar] [CrossRef]
  127. Mustapha, N.; Mokdad-Bzeouich, I.; Maatouk, M.; Ghedira, K.; Hennebelle, T.; Chekir-Ghedira, L. Antitumoral, antioxidant, and antimelanogenesis potencies of Hawthorn, a potential natural agent in the treatment of melanoma. Melanoma Res. 2016, 26, 211–222. [Google Scholar] [CrossRef]
  128. Yonekubo, B.T.; Alves, H.D.M.C.; de Souza Marques, E.; Perazzo, F.F.; Rosa, P.C.P.; Gaivão, I.O.N.D.M.; Maistro, E.L. The genotoxic effects of fruit extract of Crataegus oxyacantha (hawthorn) in mice. J. Toxicol. Environ. Health 2018, 81, 974–982. [Google Scholar] [CrossRef]
  129. Zarrinkalam, E.; Ranjbar, K.; Salehi, I.; Kheiripour, N.; Komaki, A. Resistance training and hawthorn extract ameliorate cognitive deficits in streptozotocin-induced diabetic rats. Biomed. Pharmacother. 2018, 97, 503–510. [Google Scholar] [CrossRef]
  130. Paul, S.; Sharma, S.; Paliwal, S.K.; Kasture, S. Role of Crataegus oxyacantha (Hawthorn) on scopolamine induced memory deficit and monoamine mediated behaviour in rats. Orient. Pharm. Exp. Med. 2017, 17, 315–324. [Google Scholar] [CrossRef]
  131. Lee, J.; Cho, E.; Kwon, H.; Jeon, J.; Jung, C.J.; Moon, M.; Jun, M.; Lee, Y.C.; Kim, D.H.; Jung, J.W. The fruit of Crataegus pinnatifida ameliorates memory deficits in β-amyloid protein-induced Alzheimer’s disease mouse model. J. Ethnopharmacol. 2019, 243, 112107. [Google Scholar] [CrossRef] [PubMed]
  132. Gan, Y. Synergistic Hypolipidemic Effects of Lactobacillus Plantarum PMO Fermented Hawthorn Juice on High-Fat Diet Rats. Revista Cientifica Facultad de Ciencias Veterinarias 2019, 29, 1143–1150. [Google Scholar]
  133. Kim, M.-J.; Choi, Y.; Shin, N.; Lee, M.-J.; Kim, H. Anti-obesity Effect of Crataegus pinnatifida through Gut Microbiota Modulation in High-fat-diet Induced Obese Mice. J. Korean Med. Rehabil. 2019, 29, 15–27. [Google Scholar] [CrossRef]
  134. Lee, Y.H.; Kim, Y.-S.; Song, M.; Lee, M.; Park, J.; Kim, H. A herbal formula HT048, Citrus unshiu and Crataegus pinnatifida, prevents obesity by inhibiting adipogenesis and lipogenesis in 3T3-L1 preadipocytes and HFD-induced obese rats. Molecules 2015, 20, 9656–9670. [Google Scholar] [CrossRef] [Green Version]
  135. Qin, C.; Xia, T.; Li, G.; Zou, Y.; Cheng, Z.; Wang, Q. Hawthorne leaf flavonoids prevent oxidative stress injury of renal tissues in rats with diabetic kidney disease by regulating the p38 MAPK signaling pathway. Int. J. Clin. Exp. Pathol. 2019, 12, 3440–3446. [Google Scholar]
  136. Kanyonga, M.; Faouzi, M.; Zellou, A.; Essassi, M.; Cherrah, Y. Effects of methanolic extract of Crataegus oxyacantha on blood homeostasis in rat. J. Chem. Pharm. Res. 2011, 3, 713–717. [Google Scholar]
  137. Aierken, A.; Buchholz, T.; Chen, C.; Zhang, X.; Melzig, M.F. Hypoglycemic effect of hawthorn in type II diabetes mellitus rat model. J. Sci. Food Agric. 2017, 97, 4557–4561. [Google Scholar] [CrossRef]
  138. Martínez-Rodríguez, J.; Reyes-Estrada, C.; Hernández, R.; López, J. Antioxidant, hypolipidemic and preventive effect of hawthorn (Crataegus oxyacantha) on alcoholic liver damage in rats. J. Pharmacogn. Phytother. 2016, 8, 193–202. [Google Scholar] [CrossRef] [Green Version]
  139. Li, Z.; Xu, J.; Zheng, P.; Xing, L.; Shen, H.; Yang, L.; Zhang, L.; Ji, G. Hawthorn leaf flavonoids alleviate nonalcoholic fatty liver disease by enhancing the adiponectin/AMPK pathway. Int. J. Clin. Exp. Med. 2015, 8, 17295. [Google Scholar]
  140. Li, S.; Huang, Z.; Dong, Y.; Zhu, R.; Li, T. Haw pectin pentaglaracturonide inhibits fatty acid synthesis and improves insulin sensitivity in high-fat-fed mice. J. Funct. Foods 2017, 34, 440–446. [Google Scholar] [CrossRef]
  141. Mustapha, N.; Mokdad-Bzeouich, I.; Sassi, A.; Abed, B.; Ghedira, K.; Hennebelle, T.; Chekir-Ghedira, L. Immunomodulatory potencies of isolated compounds from Crataegus azarolus through their antioxidant activities. Tumour Biol. 2016, 37, 7967–7980. [Google Scholar] [CrossRef] [PubMed]
  142. Elango, C.; Devaraj, S.N. Immunomodulatory effect of Hawthorn extract in an experimental stroke model. J. Neuroinflamm. 2010, 7, 97. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Hatipoğlu, M.; Sağlam, M.; Köseoğlu, S.; Köksal, E.; Keleş, A.; Esen, H.H. The effectiveness of Crataegus orientalis M. Bieber (Hawthorn) extract administration in preventing alveolar bone loss in rats with experimental periodontitis. PLoS ONE 2015, 10, e0128134. [Google Scholar] [CrossRef] [Green Version]
  144. Wang, X.; Zhang, C.; Peng, Y.; Zhang, H.; Wang, Z.; Gao, Y.; Liu, Y.; Zhang, H. Chemical constituents, antioxidant and gastrointestinal transit accelerating activities of dried fruit of Crataegus dahurica. Food Chem. 2018, 246, 41–47. [Google Scholar] [CrossRef]
  145. Liu, S.; Sui, Q.; Zou, J.; Zhao, Y.; Chang, X. Protective effects of hawthorn (Crataegus pinnatifida) polyphenol extract against UVB-induced skin damage by modulating the p53 mitochondrial pathway in vitro and in vivo. J. Food Biochem. 2019, 43, e12708. [Google Scholar] [CrossRef]
  146. Shin, H.S.; Lee, J.M.; Park, S.Y.; Yang, J.E.; Kim, J.H.; Yi, T.H. Hair growth activity of Crataegus pinnatifida on C57BL/6 mouse model. Phytother. Res. 2013, 27, 1352–1357. [Google Scholar] [CrossRef]
  147. Shi, Y.; Kong, X.; Yin, H.; Zhang, W.; Wang, W. Effect of Hawthorn Leaf Flavonoids in Dehydroepiandrosterone-Induced Polycystic Ovary Syndrome in Rats. Pathobiology 2019, 86, 102–110. [Google Scholar] [CrossRef]
  148. Song, J.; Shin, S.M.; Kim, H. Efficacy and safety of HT048 and HT077 for body fat and weight loss in overweight adults: A study protocol for a double-blind, randomized, placebo-controlled trial. Medicine 2019, 98, e17922. [Google Scholar] [CrossRef]
  149. Kadas, Z.; Evrendilek, G.A.; Heper, G. The metabolic effects of hawthorn vinegar in patients with high cardiovascular risk group. J. Food Nutr. Res. 2014, 2, 539–545. [Google Scholar] [CrossRef]
  150. Asher, G.N.; Viera, A.J.; Weaver, M.A.; Dominik, R.; Caughey, M.; Hinderliter, A.L. Effect of hawthorn standardized extract on flow mediated dilation in prehypertensive and mildly hypertensive adults: A randomized, controlled cross-over trial. BMC Complement. Altern. Med. 2012, 12, 26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Al-Gareeb, A.I.A. Effect of hawthorn extract on blood pressure and lipid profile in patients with stage I hypertension: A placebo-controlled, double-blind randomized trial. Mustansiriya Med. J. 2012, 11, 52–57. [Google Scholar]
  152. Asgary, S.; Naderi, G.H.; Sadeghi, M.; Kelishadi, R.; Amiri, M. Antihypertensive effect of Iranian Crataegus curvisepala Lind.: A randomized, double-blind study. Drugs Exp. Clin. Res. 2004, 30, 221–225. [Google Scholar]
  153. Degenring, F.; Suter, A.; Weber, M.; Saller, R. A randomised double blind placebo controlled clinical trial of a standardised extract of fresh Crataegus berries (Crataegisan®) in the treatment of patients with congestive heart failure NYHA II. Phytomedicine 2003, 10, 363–369. [Google Scholar] [CrossRef] [Green Version]
  154. Zapfe, G. Clinical efficacy of Crataegus extract WS® 1442 in congestive heart failure NYHA class II. Phytomedicine 2001, 8, 262–266. [Google Scholar] [CrossRef] [PubMed]
  155. Holubarsch, C.J.; Colucci, W.S.; Meinertz, T.; Gaus, W.; Tendera, M. The efficacy and safety of Crataegus extract WS 1442 in patients with heart failure: The SPICE trial. Eur. J. Heart Fail. 2008, 10, 1255–1263. [Google Scholar] [CrossRef]
  156. Zick, S.M.; Vautaw, B.M.; Gillespie, B.; Aaronson, K.D. Hawthorn Extract Randomized Blinded Chronic Heart Failure (HERB CHF) trial. Eur. J. Heart Fail. 2009, 11, 990–999. [Google Scholar] [CrossRef] [Green Version]
  157. Moeini, F.; Jafarian, A.; Aletaha, N.; Kamalinejad, M.; Naderi, N.; Babaeian, M. The Effect of Common Hawthorn (Crataegus monogyna Jacq.) Syrup on Gastroesophageal Reflux Disease Symptoms. Iran. J. Pharm. Sci. 2016, 12, 69–76. [Google Scholar]
  158. Trexler, S.E.; Nguyen, E.; Gromek, S.M.; Balunas, M.J.; Baker, W.L. Electrocardiographic effects of hawthorn (Crataegus oxyacantha) in healthy volunteers: A randomized controlled trial. Phytother. Res. 2018, 32, 1642–1646. [Google Scholar] [CrossRef]
  159. Schandry, R.; Lindauer, D.; Mauz, M. Blood pressure and cognitive performance after a single administration of a camphor-crataegus combination in adolescents with low blood pressure. Planta Med. 2018, 84, 1249–1254. [Google Scholar] [CrossRef] [Green Version]
  160. Erfurt, L.; Schandry, R.; Rubenbauer, S.; Braun, U. The effects of repeated administration of camphor-crataegus berry extract combination on blood pressure and on attentional performance–A randomized, placebo-controlled, double-blind study. Phytomedicine 2014, 21, 1349–1355. [Google Scholar] [CrossRef] [PubMed]
  161. Walker, A.F.; Marakis, G.; Simpson, E.; Hope, J.L.; Robinson, P.A.; Hassanein, M.; Simpson, H.C. Hypotensive effects of hawthorn for patients with diabetes taking prescription drugs: A randomised controlled trial. Br. J. Gen. Pract. 2006, 56, 437–443. [Google Scholar] [PubMed]
  162. Walker, A.F.; Marakis, G.; Morris, A.P.; Robinson, P.A. Promising hypotensive effect of hawthorn extract: A randomized double-blind pilot study of mild, essential hypertension. Phytother. Res. 2002, 16, 48–54. [Google Scholar] [CrossRef] [PubMed]
  163. Werner, N.S.; Duschek, S.; Schandry, R. D-camphor-crataegus berry extract combination increases blood pressure and cognitive functioning in the elderly–A randomized, placebo controlled double blind study. Phytomedicine 2009, 16, 1077–1082. [Google Scholar] [CrossRef]
  164. Rigon, R.B.; Fachinetti, N.; Severino, P.; Durazzo, A.; Lucarini, M.; Atanasov, A.G.; El Mamouni, S.; Chorilli, M.; Santini, A.; Souto, E.B. Quantification of Trans-Resveratrol-Loaded Solid Lipid Nanoparticles by a Validated Reverse-Phase HPLC Photodiode Array. Appl. Sci. 2019, 9, 4961. [Google Scholar] [CrossRef] [Green Version]
  165. Souto, E.B.; Fernandes, A.R.; Martins-Gomes, C.; Coutinho, T.E.; Durazzo, A.; Lucarini, M.; Souto, S.B.; Silva, A.M.; Santini, A. Nanomaterials for Skin Delivery of Cosmeceuticals and Pharmaceuticals. Appl. Sci. 2020, 10, 1594. [Google Scholar] [CrossRef] [Green Version]
  166. Sánchez-López, E.; Gomes, D.; Esteruelas, G.; Bonilla, L.; Lopez-Machado, A.L.; Galindo, R.; Cano, A.; Espina, M.; Ettcheto, M.; Camins, A. Metal-Based Nanoparticles as Antimicrobial Agents: An Overview. Nanomaterials 2020, 10, 292. [Google Scholar] [CrossRef] [Green Version]
  167. Vieira, R.; Severino, P.; Nalone, L.A.; Souto, S.B.; Silva, A.M.; Lucarini, M.; Durazzo, A.; Santini, A.; Souto, E.B. Sucupira Oil-Loaded Nanostructured Lipid Carriers (NLC): Lipid Screening, Factorial Design, Release Profile, and Cytotoxicity. Molecules 2020, 25, 685. [Google Scholar] [CrossRef] [Green Version]
  168. Souto, E.B.; Ribeiro, A.F.; Ferreira, M.I.; Teixeira, M.C.; Shimojo, A.A.; Soriano, J.L.; Naveros, B.C.; Durazzo, A.; Lucarini, M.; Souto, S.B. New Nanotechnologies for the Treatment and Repair of Skin Burns Infections. Int. J. Mol. Sci. 2020, 21, 393. [Google Scholar] [CrossRef] [Green Version]
  169. Souto, E.B.; Silva, G.F.; Dias-Ferreira, J.; Zielinska, A.; Ventura, F.; Durazzo, A.; Lucarini, M.; Novellino, E.; Santini, A. Nanopharmaceutics: Part I—Clinical trials legislation and good manufacturing practices (GMP) of nanotherapeutics in the EU. Pharmaceutics 2020, 12, 146. [Google Scholar] [CrossRef] [Green Version]
  170. Souto, E.B.; Silva, G.F.; Dias-Ferreira, J.; Zielinska, A.; Ventura, F.; Durazzo, A.; Lucarini, M.; Novellino, E.; Santini, A. Nanopharmaceutics: Part II—Production scales and clinically compliant production methods. Nanomaterials 2020, 10, 455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  171. Pimentel-Moral, S.; Teixeira, M.; Fernandes, A.; Arraez-Roman, D.; Martinez-Ferez, A.; Segura-Carretero, A.; Souto, E. Lipid nanocarriers for the loading of polyphenols–A comprehensive review. Adv. Colloid Interface Sci. 2018, 260, 85–94. [Google Scholar] [CrossRef] [PubMed]
  172. Singh, B. (Ed.) NanoNutraceuticals; CRC Press: Boca Raton, FL, USA, 2018; p. 326. [Google Scholar] [CrossRef]
Figure 1. Scheme of the hawthorn therapeutic properties.
Figure 1. Scheme of the hawthorn therapeutic properties.
Forests 11 00564 g001
Figure 2. Overview of the main compounds found in hawthorn.
Figure 2. Overview of the main compounds found in hawthorn.
Forests 11 00564 g002
Table 1. Identified compounds from hawthorn.
Table 1. Identified compounds from hawthorn.
SpeciesCompound IdentifiedMethodological and Analytical ApproachReference
Crataegus oxyacanthaEpicatechin, epicatechin gallate (ECG), rutin, cafeic and caftaric acidsHPLC-DAD and LC-MS/MS[27]
Crataegus oxyacanthaNaringenin, epicatechin, quercetin-3-O-β-glucoside, and quercetinNuclear magnetic resonance (NMR) spectroscopy[28]
Crataegus oxyacanthaRutin and QuercetinHPLC[29]
Crataegus pinnatifidaCrataequinone ANuclear magnetic resonance (NMR) spectroscopy and electronic circular dichroism (ECD)[43]
Crataegus songaricaQuercitin 3-O-galactoside and kaempherol-3-O-glucosideHPLC-DAD-ESI/MS[44]
Crataegus pinnatifidaPinnatifidanin BVINuclear magnetic resonance (NMR) spectroscopy[45]
Crataegus pinnatifidaPinnatifidanoside FNuclear magnetic resonance (NMR) spectroscopy[46]
Crataegus azarolus varQuercetin 3-O-methyl ether, 3-β-O acetyl ursolic acidReversed phase HPLC (RP-HPLC)[47]
Crataegus pinnatifida(+)-(7S,8R)-crataegusin A and (−)-(7R,8S)-crataegusin AElectronic circular dichroism (ECD)[48]
Crataegus pinnatifida Bge(−)-7S,8R-4,7,9,9′-tetrahydroxy-3,5,3′,5′-tetramethoxy-8-O-4′-neolignanElectronic circular dichroism (ECD) and HPLC[49]
Crataegus pubescens(+)-catechin and (−)-epicatechinMicellar electrokinetic chromatography (MEKC) and HPLC/UV[50]
Crataegus pinnatifidaChlorogenic acid (CA), vitexin-400-o-glucoside (VG), vitexin-200-o-rhamnoside (VR), orientoside (ORT), rutin (RT), vitexin (VIT) and hyperoside (HYP)HPLC[51]
Crataegus pinnatifida var. major N.E.Br.(7′S, 8′R, 8R)-isolariciresinol-9′-β-D-glucopyranoside and lyonisideNuclear magnetic resonance (NMR) spectroscopy and LC-MS[52]
Table 2. In vitro reported activities for hawthorn.
Table 2. In vitro reported activities for hawthorn.
Experimental Conditions: In vitro
ActivityEffectReference
AntimicrobialApigenin-7-O-glucoside and luteolin 3,7-diglucoside extracted from hawthorn were the most potent chemicals to eliminate Ureaplasma urealyticum with minimum inhibitory concentration value ranges of 0.48–3.9 μg/mL and 0.48–1.95 μg/mL, respectively.[89]
Antioxidant and anti-inflammatoryUrsolic acid and oleanolic acid extracted from hawthorn showed anti-inflammatory and antioxidative effects in PC12 cells by decreasing the cell death induced by 1-methyl-4-phenylpyridinium ions (MPP+) and hydrogen peroxide (H2O2) as well as reducing lactate dehydrogenase leakage.[90]
AnticancerCrataequinone A exhibited cytotoxic effects on Hep3B and HepG2 cell lines with IC50 values of 24.90 μM and 12.24 μM, respectively.[43]
AnticancerQuercitin 3-O-galactoside and kaempherol-3-O-glucoside inhibited the culture of MCF-7 human breast cancer cells.[44]
AnticancerPinnatifidanin BVI extracted from hawthorn had a preventive effect against Mrc5 human lung cells. [45]
AntioxidantNaturally occurring compounds from ethanolic and aqueous extracts of C. monogyna showed antioxidant and hydrogen peroxide scavenging properties.[91]
Anti-inflammatoryAqueous hawthorn fruit extract inhibited the expression of ILInterleukin-6, Interleukin-1β, Tumor necrosis factor-α and cyclooxygenase-2 genes, and prevented NO formation in RAW 264.7 cells.[92]
Table 3. The main studies in animals involving hawthorn.
Table 3. The main studies in animals involving hawthorn.
Experimental Conditions: In Animal Model
ActivityEffectReference
Anticataract potentialC. pinnatifida leaf extracts used three times a day reduced the level of malondialdehyde and increased serum levels of catalase and superoxide dismutase in rats with selenite-induced cataracts.[109]
Dyslipidemia therapy effectC. pinnatifi fruit extract (250 mg/kg) for 7 days in high-fat-diet-fed mice with hyperlipidemia reduced blood lipid and lipid degradation by enhancing the hepatic expression of peroxisome proliferator-activated receptor α.[110]
Anti- atherosclerosis effectOligomeric proanthocyanidins extracted from C. oxyacantha in Wistar rats decreased the differentiation of monocytes to macrophages via the downregulation of inflammation and the reduction of monocyte chemoattractant protein -1 and vascular cell adhesion molecule-1 levels.[111]
Antibacterial effectHawthorn fruit extract (including monomers of (+)-catechin, (−)-epicatechin gallate and (−)-epigallocatechin) could control methicillin-resistant Staphylococcus aureus (MRSA) in septic mice by enhancing the accumulation of daunomycin inside MRSA cells and by downregulating the expression of norA, norC and abcA mRNAs (the main efflux pumps of MRSA).[112]
Anti-inflammatory effectThe administration of C. pinnatifida dried fruit extract reduced the expression of hepatic cyclooxygenase-2 and nitric oxide synthase.[113]
Radioprotective effectThe treatment of mouse bone marrow cells with phenolic compounds extracted from hawthorn (200 mg/kg) caused a reduction in 2-Gy γ-radiation-induced stress and genotoxicity.[114]
Anti- atherosclerosis effectThe administration of sugar-free C. pinnatifida aqueous extract in atherosclerosis-induced rats resulted in the regulation of endothelial function and reduction of inflammatory responses and serum lipid levels.[115]
Cardioprotective effectThe administration of aqueous extract of C. tanacetifolia leaf (100 mg/kg) for 4 weeks in rats prevented hypertension.[116]
Cardioprotective effectThe administration of alcoholic extract of C. oxycantha (0.5 mL/100 g body weight/day) for a month prevented isoproterenol-induced myocardial infarction through a reduction in enzymes involved in the Krebs cycle. It also prevented peroxidative injury of mitochondrial lipids and preserved the mitochondrial antioxidant balance.[117]
Analgesic and central nervous system activitiesThe administration of hawthorn seed and pulp extracts (1000 mg/kg) in mice reduced pain, sleep disorders, nervousness and stress with low toxicity.[118]
Table 4. Examples of studies in humans involving hawthorn.
Table 4. Examples of studies in humans involving hawthorn.
Experimental Conditions: Clinical Trials
ActivityAdministrationMain FindingsReference
Anti-inflammatory effectPatients with diabetes (n = 37) received hawthorn vinegar (20 mL) diluted with water (40 mL) after meals for a month.The treatment reduced serum levels of triglyceride, LDL, cholesterol and glucose, as well as decreased glycated hemoglobin, blood pressure and body weight.[149]
Anti-hypertensive effectPatients (n = 21) randomly received 1000 mg, 1500 mg and 2500 mg of hawthorn extract twice per day for four days.The treatment lowered blood pressure.[150]
Anti-hypertensive effectHypertensive patients (n = 60) received 450 mg of hawthorn extract twice per day for three months.The treatment elevated the level of high-density lipoprotein and reduced the level of low-density lipoprotein, total cholesterol, diastolic blood pressure and systolic blood pressure.[151]
Antihypertensive effectThe administration of hawthorn hydroalcoholic extract in subjects with primary mild hypertension.A reduction in diastolic and systolic blood pressure after four months.[152]
Treatment of patient with New York Heart Association class II heart failureThe administration of Crataegus berry extracts (30 drops, three times per day) in subjects with NYHA class II heart failure.An improvement of confirmed tolerability and an enhancement of exercise tolerance after eight weeks.[153]
Treatment of patient with New York Heart Association class II heart failureThe administration of Crataegus extract in subjects with congestive heart failure (NYHA class II).A confirmation of the well-tolerated nature and safety of Crataegus extract based on in vitro parameters and treatment of congestive heart failure (NYHA class II) after 12 weeks.[154]

Share and Cite

MDPI and ACS Style

Nazhand, A.; Lucarini, M.; Durazzo, A.; Zaccardelli, M.; Cristarella, S.; Souto, S.B.; Silva, A.M.; Severino, P.; Souto, E.B.; Santini, A. Hawthorn (Crataegus spp.): An Updated Overview on Its Beneficial Properties. Forests 2020, 11, 564. https://doi.org/10.3390/f11050564

AMA Style

Nazhand A, Lucarini M, Durazzo A, Zaccardelli M, Cristarella S, Souto SB, Silva AM, Severino P, Souto EB, Santini A. Hawthorn (Crataegus spp.): An Updated Overview on Its Beneficial Properties. Forests. 2020; 11(5):564. https://doi.org/10.3390/f11050564

Chicago/Turabian Style

Nazhand, Amirhossein, Massimo Lucarini, Alessandra Durazzo, Massimo Zaccardelli, Santo Cristarella, Selma B. Souto, Amélia M. Silva, Patrícia Severino, Eliana B. Souto, and Antonello Santini. 2020. "Hawthorn (Crataegus spp.): An Updated Overview on Its Beneficial Properties" Forests 11, no. 5: 564. https://doi.org/10.3390/f11050564

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