Next Article in Journal
Skeletal Ryanodine Receptors Are Involved in Impaired Myogenic Differentiation in Duchenne Muscular Dystrophy Patients
Next Article in Special Issue
Fructose-Rich Diet Is a Risk Factor for Metabolic Syndrome, Proximal Tubule Injury and Urolithiasis in Rats
Previous Article in Journal
Sex Bias in Differentiated Thyroid Cancer
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Sanguiins—Promising Molecules with Broad Biological Potential

1
Student’s Scientific Association, Department of Pharmacognosy, Faculty of Pharmacy with the Division of Laboratory Medicine, Medical University of Białystok, ul. Mickiewicza 2a, 15-230 Białystok, Poland
2
Department of Pharmacognosy, Faculty of Pharmacy with the Division of Laboratory Medicine, Medical University of Białystok, ul. Mickiewicza 2a, 15-230 Białystok, Poland
3
Ludwig Boltzmann Institute for Digital Health and Patient Safety, Medical University of Vienna, Spitalgasse 23, 1090 Vienna, Austria
4
Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Jastrzębiec, 05-552 Magdalenka, Poland
5
Department of Pharmaceutical Sciences, University of Vienna, Althanstrasse 14, 1090 Vienna, Austria
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2021, 22(23), 12972; https://doi.org/10.3390/ijms222312972
Submission received: 6 November 2021 / Revised: 27 November 2021 / Accepted: 29 November 2021 / Published: 30 November 2021
(This article belongs to the Special Issue Nutraceuticals in Human Diseases: Metabolism and Interactions)

Abstract

:
Compounds of natural origin, an infinite treasure of bioactive chemical entities, persist as an inexhaustible resource for discovering new medicines. In this review, we summarize the naturally occurring ellagitannins, sanguiins, which are bioactive constituents of various traditional medicinal plants, especially from the Rosaceae family. In-depth studies of sanguiin H-6 as an antimicrobial, antiviral, anticancer, anti-inflammatory, and osteoclastogenesis inhibitory agent have led to potent drug candidates. In addition, recently, virtual screening studies have suggested that sanguiin H-6 might increase resistance toward SARS-CoV-2 in the early stages of infection. Further experimental investigations on ADMET (absorption, distribution, metabolism, excretion, and toxicity) supplemented with molecular docking and molecular dynamics simulation are still needed to fully understand sanguiins’ mechanism of action. In sum, sanguiins appear to be promising compounds for additional studies, especially for their application in therapies for a multitude of common and debilitating ailments.

1. Introduction

Most of the discovered drugs are either drugs of natural origin or synthetic derivatives of natural compounds. Thus, a multidisciplinary approach to drug discovery and molecular diversity from natural product sources needs to be combined to provide the best solution to the problems with drug discovery and development [1,2]. Plants are known to be a rich source of pharmacologically active secondary metabolites divided into structural chemical classes [3,4]. One of the pharmacologically valuable classes of phytoconstituents are ellagitannins (ETs), and belonging to them, sanguiins. ETs, water-soluble phenolics, are esters of hexahydroxydiphenic acid and a polyol, usually β-D-glucose or quinic acid [5,6,7]. ET compounds demonstrate an enormous structural variability connected with various possibilities for the linkage of hexahydroxydiphenic residues with the glucose moiety and particularly by their easy susceptibility to creating dimeric and oligomeric derivatives [8]. The polyphenol-protein system and its interactions may underlie the medicinal properties exhibited by members of the ETs family. Fruits and nuts are rich sources of ellagitannins and are important in the human diet due to their properties as micronutrients [9,10]. Due to the limited bioavailability of ellagitannins, as orally administered and the metabolic chemical changes as a result of their transit through the gastrointestinal tract, comprising of hydrolysis and gut microbiota metabolism, the activity of the produced metabolites also needs to be taken into consideration [3].
Sanguiins, members of the ET class of hydrolyzable plant polyphenols, are found mainly in the Rosaceae family and are primarily widespread in berries. The main advantage of sanguiins over other common polyphenols in the plant world is their wide distribution in food products. Therefore, their health-promoting properties can be used in a properly balanced diet [11]. In addition to the natural occurrence of sanguiin, there are reports on the synthetic production of sanguiin H-5 [12]. The structural features of sanguiins make them a demanding molecular target. Sanguiin H-1 comprises the characteristic hexahydrodiphenoyl (HHDP) moiety linked with β-D-glucose and 1,6-di-O-galloyl moieties. On the other hand, sanguiin H-2 possesses one galloyl moiety and two sanguisorboyl linking ester groups. Comparing the H-1 and H-4 sanguiins, they differ only in an additional galloyl substituent in sanguiin H-1. The structure of sanguiin H-3, a dimeric ellagitannin, contains two glucose substitutions. Furthermore, the complex structure of sanguiin H-6 includes sanguiin H-2 and pedunculagin moieties. The chemical structure of sanguiin H-10 closely resembles sanguiin H-2, except sanguiin H-10 contains an extra HHDP group. Substitution patterns of sanguiin H-11 also show similarities to sanguiin H-2. The only difference between these structures is the lack of a galloyl moiety in the sanguiin H-11 [13].
Although various bioactivities (e.g., antioxidant, anticancer, antiviral, and antimicrobial) of sanguiins, mainly sanguiin H-6, have been investigated, their pharmacological potential demonstrated in vitro, in silico, and in vivo experimental models has not been clearly organized through review articles. Thus, this manuscript summarizes the findings on the widespread bioactivities of sanguiin H-1 (SH1), sanguiin H-2 (SH2), sanguiin H-3 (SH3), sanguiin H-4 (SH4), sanguiin H-6 (SH6), sanguiin H-10 (SH10), and sanguiin H-11 (SH11) to showcase their potential to be used as therapeutic agents.

2. Methodology

A broad search strategy was used to find English language publications indexed in SCOPUS, PubMed/MEDLINE, Google Scholar, Web of Science (SCI-EXPANDED), Wiley Online Library, Taylor & Francis Online, REAXYS Database, Science Direct/ELSEVIER, and EBSCO Discovery Service (EDS) [14]. These databases were searched systematically for articles published from 1982 to 2021. Relevant publications were selected manually from the following searches: sanguiin, sanguiins, Rosaceae, traditional use, traditional medicine, folk medicine, sanguiin H-6, sanguiin H6, sanguiin H-10, sanguiin H10, sanguiin H-5, sanguiin H5, sanguiin H-2, sanguiin H2, sanguiin H-11, sanguiin H11, sanguiin H-4, sanguiin H4, sanguiin H-3, sanguiin H3, ellagitannins, tannins, Rubus, anticancer, antiviral, SARS-CoV-2, COVID-19, antioxidant, anti-inflammatory, biological activity, antimicrobial, biological potential, metabolism, clinical trials, preclinical trials, chemistry, galloyl moiety, absorption, distribution, excretion, toxicity, perspectives, trials, pharmacological, natural product, secondary metabolites, therapeutic agent, inhibitory activity, inhibitors, dose, efficacy, exposure, experimental model, quantitative analysis, qualitative analysis, geographical location, as well as each of species containing sanguiins combined with traditional use, traditional medicine, or folk medicine. The search terms operated in separate or limited combinations that considered the requirements or limitations of the database being used.

3. Natural Occurrence of Sanguiins

Among various phenolic compounds isolated from the Rosaceae family, tannins and related compounds seem to have a leading position. It is known that plants previously used in folk medicine represent a suitable beginning to discover new potent drugs to treat various human disorders [15]. Sanguiins (Figure 1), naturally occurring ET, have been isolated chiefly from Rubus species and are used as a traditional drug to cure, e.g., diarrhea, menstrual pain, menopause disorders, liver diseases, aphtha, gingivitis, as well as fever, angina, enteritis, hepatitis, concretion, eczema, rheumatism, enterocolitis, bronchitis, prostate disorders, pain, cold, cough, and fever (Table 1) [16,17]. Moreover, SH6 seems to be the most widespread within plants of the Rubus and is present in 22 species of this genus. Furthermore, the largest number of isolated and identified types of sanguiins, including SH2, SH4, SH5, SH6, and SH11, are found in Rubus coreanus [18]. Besides the Rubus genus, sanguiins and their isomers are found and reported in Alchemilla vulgaris, Alchemilla mollis [19], Duchesnea indica [20], Euphorbia fischeriana [21], Fragaria vesca, Fragaria ananassa [22], Punica granatum [23], Terminalia calamansanai [24], as well as in Sanguisorba officinalis [25], and Sanguisorba tenuijolia var. alba [18].
Among all sanguiins detected in plant material, only part of them was quantitatively analyzed. The place of harvest displays a relevant role in the amount of isolated sanguiins. For example, in Rubus fruticosus fruits, the range of detected SH6 is 135.04–547.48 mg/100 g of d.w. (dry weight) [26] and in Rubus idaeus shoots, 170.9–633.1 mg/100 g of d.w of the extract [27]. Following that, sanguiins content depends on fruits’ ripeness, harvest time, climate, geographic location, and mineral nutrition [10,28]. It is reported that in Rubus and Fragaria species, ellagitannins content represents a range of 50% to 80% of all phenolic compounds [10,29]. In this review, the list of plants that produce sanguiins and their reported traditional uses are tabulated in Table 1.

4. Chromatographic Techniques for the Analysis of Sanguiins

Chromatography displays a crucial role in the analysis of chemical compound mixtures. As a method for the separation and analysis of extracts and fractions from plants, it provides the possibility of qualitative and quantitative determination of the test substance with high resolution [67]. Chromatographic techniques and analysis conditions for detection, quantitative determination, and isolation of sanguiins and their isomers are given in Table 2.

5. Biological Potential of Sanguiins

Sanguiins, as one of the subgroups of polyphenolic ellagitannins, exhibit various pharmacological activities due to having different chemical structures. They possess a broad spectrum of pharmacological features such as anticancer, anti-inflammatory, antioxidant, osteoprotective, estrogenic, antibacterial, antifungal, and antiviral (including SARS-CoV-2), as shown in Table 3. Various in vivo and in vitro investigations on sanguiins, especially on sanguiin H-6, have elucidated their medicinal characteristics and mechanisms of action [68,69].

5.1. Antioxidant and Anti-Inflammatory Activities

One of the best-shown properties of polyphenols, and following that, sanguiins, is the potential antioxidant effect. Most references mention sanguiin H-6 as the primary compound having antioxidant activity, e.g., its influences on stress and oxidative damage were investigated. The production of peroxynitrite (ONOO-) was induced by the administration of lipopolysaccharide (LPS), followed by the induction of ischemia and reperfusion [88]. It was revealed that receiving SH6 before induction of oxidative damage could reduce the adverse effects associated with the release of ONOO- and enhance the improvement of injured kidney function [72]. Another chemical compound belonging to the sanguiins group that exhibits antioxidant activity is SH11. An examination of the protective effect of SH11 isolated from Sanguisorbae radix and its mechanism against glutamate-induced death in HT22 murine hippocampal cells exposed a significant reduction in glutamine-induced reactive oxygen radicals’ accumulation and calcium ion influx [74]. Furthermore, ellagitannins from the berries of the Rubus family, including dimeric SH6 and SH10, function both as radical scavengers (in a DPPH test) and as antioxidants toward lipid oxidation in food emulsions (studied in bulk and emulsified methyl linoleate, in human low-density lipoprotein in vitro) [75]. The impact of sanguiins on the inflammation process was investigated by measuring their effect on rat neutrophils’ chemotaxis. SH11 and SH6 effectively inhibited the cytokine-induced neutrophil chemoattractant migration process by 10.7% and 33%, respectively, in comparison with the control. Additionally, the study showed no toxic effect of sanguiin on neutrophils [70]. Furthermore, at a concentration of 2.5 μM, SH6 completely inhibited the release of IL-8 induced by tumor necrosis factor α and interleukin-1β and inhibited TNFα stimulated NF-κB transcription [71]. SH6 caused a concentration-dependent reduction in nitrite production, regression in induced NO synthase (iNOS) activity, and an increase in cell viability. Moreover, SH6 showed an apparent scavenging effect for NO generated from sodium nitroprusside (NO donor) [76].

5.2. Osteoclastogenesis Inhibitory Activity

In a subsequent in vitro study, the action of Rubus parvifolius L. and its main component, SH6, was tested as the inhibitor of osteoclastogenesis and bone resorption. Sanguiin influence was based on the reduction in osteoclast differentiation and bone resorption, a decrease in the production of reactive oxygen species, as well as the inhibition of the nuclear translocation of the nuclear factor of activated T cells cytoplasmic-1 (NFATc1), c-Fos, and nuclear factor-κB. Additionally, sanguiin reduced the levels of NFATc1, cathepsin K, c-Src, and inhibited in vivo TNF-α-mediated osteoclastogenesis [47].

5.3. Antibacterial Activity

The growing resistance of bacteria to currently used antibiotics is a growing problem in current medicine [89]. Increasingly emerging research on sanguine antibacterial properties gives hope for the discovery of antibacterial agents with the lack of unpleasant side effects. Examination of the antibacterial activity of fruits of selected Rubus species and compounds (SH6 and ellagic acid) against selected Gram-negative and Gram-positive bacteria allowed assessment of their usefulness in the fight against microorganisms. The results showed that SH6 was active against Streptococcus A (MIC = 0.5 mg/mL), Streptococcus pneumoniae (MIC = 0.5 mg/mL), Corynebacterium diphtheriae (MIC = 0.03 mg/mL), Bacillus subtilis (MIC = 0.5 mg/mL), Clostridium sporogenes (MIC = 0.06 mg/mL), Staphylococcus aureus (MIC = 0.25 mg/mL), Staphylococcus epidermidis (MIC = 0.125 mg/mL), and Moraxella catarrhalis (MIC = 0.5 mg/mL) [27].
Additionally, another study showed that SH6 exhibited a significant inhibition level against S. aureus, E. coli, and C. perfringens [77]. Rubus ulmifolius fruit extract containing SH10, showed an antibacterial effect against Escherichia coli, Morganella morganii, and Proteus mirabilis, but higher extract concentrations were required: MIC = 5 mg/mL, MIC = 5 mg/mL, and MIC = 10 mg/mL, respectively [78].

5.4. Antifungal Activity

Moreover, Rubus ulmifolius fruit extract was tested as an antifungal agent. It was proved that the extract containing SH6 exhibited fungistatic activity against Candida albicans. The minimum inhibitory concentration was 5 mg/mL. Unfortunately, the extract did not show any fungicidal activity, achieving a result of >20 mg/mL [78].

5.5. Antiviral Activity (Including SARS-CoV-2)

Viruses, as pathogenic microorganisms, show significant genetic variability and the ability to mutate. Often, they do not show signs of infection at first. Currently, an increasing number of drug-resistant strains, as well as the toxicity of previously known drugs, force researchers to develop new antiviral substances [90]. In recent months, the entire world has been severely affected by the SARS-CoV-2 pandemic, which has led scientists to focus their attention on potential candidates against its eradication. More and more recent research conducted worldwide shows that sanguiins may be a potential candidate in the fight against viral diseases, including COVID-19 [91,92]. One of the studies predicted that SH6 is a compound that binds very well to the S1 and S2 subunits of the SARS-CoV-2 virus spine, which is responsible for entering the host cells and causing infection. SH6 showed the best binding energy among all tested compounds in the molecular docking assay. Additionally, SH2, also mentioned in the study, showed a lower result than the one mentioned above. Moreover, sanguiin has been proposed to act not only against the spike subunits of the SARS-CoV-2 virus [93]. Another molecular docking examination of polyphenolic compounds against the SARS-CoV-2 virus Mpro protease revealed that SH6 had the best result of all tested compounds in the in silico model [80]. Moreover, the study performed by S. Luo et al. concerned the verification of bacterial neuraminidase inhibitory properties by nine compounds isolated from mock strawberry (Duchesnea indica Andr.). SH4 exhibited significant inhibitory activity in an in vitro model, which offers potential for its use as a new antiviral substance [20].

5.6. Anticancer Activity

Additionally studied features of sanguiins are their potential anticancer activity. Several investigations on SH6 have explained its anticancer effect due to its promising competency in inhibiting DNA topoisomerases I and II. Moreover, the compound acted as a blocker to HeLa cells. It inhibited their growth at an effective dose of 12 µM and also had a dose-dependent effect on intracellular topoisomerase activity. SH6 also exhibited significant antiangiogenic potential [82]. A study by Lee S. et al. on HT1080 human fibrosarcoma cells showed that this compound blocked KDR/Flk-1-Fc binding to VEGF165 in a dose-dependent manner. Moreover, the compound obstructed the VEGF-induced proliferation of HUVEC cells (IC50 ca. = 7.4 µg/mL) but was not active against HT1080 human fibrosarcoma cells [83].
The potential antitumor properties of sanguiins were also tested on PRMI-7951 melanoma cells. A moderate selective cytotoxicity was shown by SH2, SH6, and SH11 with ED50 results of 0.44, 0.5, and 5.0 µg/mL, respectively [68]. Furthermore, anticancer activity was tested with SH4 isolated from Terminalia calamansanai leaves against large tumor cells lines, including human promyelocytic leukemia HL-60 cells. The compound induced a decrease in human poly (ADP-ribose) polymerase [79] (PARP) associated with the cleavage of procaspase-3 and exhibited strong activation of proapoptotic caspase-3 in HL-60 cells. It is worth mentioning that SH4 does not affect healthy cells, suggesting this compound is selective against cancer cells [24]. In another examination, SH6 was responsible for modulating the Smad 2/3 signaling pathway by TGF-β1, increasing the expression of the epithelial marker E-cadherin, repressing the expression of Snail and the mesenchymal marker N-cadherin during TGF-β1-induced EMT (epithelial-mesenchymal transition), and regulating the expression of EMT-dependent genes induced by TGF-β1. In summary, SH6 inhibits the migration and invasion of A549 lung cancer in vitro by inhibiting TGF-β1 induction of EMT [84].
Moreover, SH6 showed a large number of antiproliferative, antimigration, and cytotoxic effects against human breast carcinoma cells. A study performed by Berdowska et al. proved that the tested compound exhibited an inhibitory effect on adriamycin-resistant cells (MCF-7/Adr) [85]. It also showed antimetastatic properties in MDA-MB-231 cells by reducing the expression of vascular endothelial growth factor (VEGF), phosphorylated Akt, and kinase 1/2 (ERK1/2) regulated by extracellular signals [86]. In addition, SH6 increased the ratio of Bax to Bcl-2 in both MCF-7 and MDA-MB-231 cells [79].
SH6 was also studied for its activity against A2780 human ovarian carcinoma cells. The tested compound induced an antiproliferative effect and a morphological change similar to apoptotic cell death but did not arrest the cancer cell cycle. Moreover, SH6 showed an early apoptotic effect, caspase activation, PARP cleavage, activation of mitogen-activated protein kinases (MAPKs), especially p38, and an increase in truncated p15/BID [87].

5.7. Estrogenic Activity

SH6 has also been tested for estrogenic activity against MCF-7 human breast cancer cells. The E-screen examination and the molecular docking analysis showed that the SH6 from Rubus coreanus exhibited the best binding energy of −250,149 kcal/mol. Additionally, at 100 μg/mL, R. coreanus extract significantly stimulated cell proliferation (574.57% ± 8.56%). The study results indicated that SH6 contributed to the estrogenic activity of R. coreanus by activating the ERα coactivator binding site [81].

5.8. Neuroprotective Activity

Rubus L. subgenus R. watson, R. brigantinus, and R. vagabundus extracts containing SH2, SH6, and SH10 were tested for their potential neuroprotective properties against SK-N-MC neuroblastoma cells. All digested extracts after 2 and 24 h of preincubation reduced basal ROS production. Rubus brigantinus and R. vagabundus extracts increased the mitochondrial transmembrane potential and the integrity of the cell membrane. Moreover, the extracts increased GSH levels while not changing the GSH/GSSG ratio. It is worth mentioning that there is insufficient evidence for the interaction of brain endothelial cells with polyphenol metabolites, which makes it difficult to determine the level of the passage of the compound across the blood–brain barrier [62].

5.9. Clinical Trials

As mentioned above, the efficacy of sanguiins is mainly limited to preclinical studies. However, there has been some research on black raspberry and pomegranate food products in clinical trials. Considering the fact that these products are rich in ellagitannins, it can be concluded that the biological activity may also be connected with the occurrence of sanguiins in the juice from berries and pomegranate. Nevertheless, there is a lack of information on clinical studies that use only sanguiins in medical treatment [44,94,95].

6. Pharmacokinetics of Sanguiins

Sanguiins, belonging to the ellagitannin group, show similar pharmacokinetics. In vitro studies have shown that ellagitannins are stable in the gastric environment, and in the presence of gastric enzymes, they are not hydrolyzed to ellagic acid. In addition, the absorption of ellagitannins in the stomach is impracticable due to their complex chemical structure. However, free ellagic acid molecules can be absorbed in the stomach. On the other hand, the intestinal environment, together with the gastrointestinal microbiota, creates suitable conditions for their hydrolysis and decomposition into urolithins and their derivatives, which pass through the intestinal wall into the enterohepatic circulation [96]. In addition, in vivo studies have shown that the metabolism of SH6 and SH10 in the liver is partly based on conjugation with glucuronic acid and sulfuric acid, leading to the formation of compounds such as urolithin A-O-glucuronide, urolithin A-sulfate, and urolithin B-3-O-glucuronide. Moreover, urolithins were detected in the unconjugated form. Conjugation of derivatives occurs at different rates and intensities; Tmax of plasma urolithin glucuronides and sulfates is achieved in the vast majority of compounds 24 h after administration. Ultimately, conjugated and unconjugated compounds are excreted in the urine at varying intervals, up to 48 h after ingestion. Further in vivo clinical studies linked to full pharmacokinetic analysis are necessary to fully determine the participation of urolithins in the therapeutic effects of ellagitannin-rich plants [3,97,98].

7. Conclusions

The isolation and structure determination, accompanied by the measurement of the diverse pharmacological activities of each isolated sanguiin, has brought about a marked change in the concept of these compounds as active components of medicinal plants. In summary, sanguiins, especially sanguiin H-6, show evidence of promising action in various biological contexts, particularly in respect of their anticancer, antiradical, and antiviral properties. Apart from that, further studies involving drug delivery may improve the effectiveness of these compounds toward the drug target sites. Furthermore, it is worth considering performing a supplementary survey on their metabolism and toxicology patterns with molecular docking and molecular dynamics simulation to understand their mechanisms of action fully.

Author Contributions

Conceptualization, K.J., J.G. and M.T.; methodology, K.J. and J.G.; formal analysis, M.T. and A.G.A.; writing—original draft preparation, K.J. and J.G.; writing—review and editing, M.T. and A.G.A.; visualization, K.J., and J.G.; supervision, M.T. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data are contained within the article.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

SH2sanguiin H-2
SH3sanguiin H-3
SH5sanguiin H-5
SH6sanguiin H-6
SH10sanguiin H-10
SH11sanguiin H-11
ADMET absorption, distribution, metabolism, excretion, toxicity
CINC-1cytokine-induced neutrophil chemoattractant
TNFα tumor necrosis factor α
IL-1βinterleukin-1β
d.w.dry weight
ONOO-peroxynitrite
LPS lipopolysaccharide
NFATc1nuclear factor of activated T cells 1
NF-κB nuclear factor-κB
c-Src proto-oncogene tyrosine-protein kinase Src
TNF-α tumor necrosis factor
PARP poly (ADP-ribose) polymerase
EMT epithelial-mesenchymal transition
TGF-β1transforming growth factor beta 1
VEGF vascular endothelial growth factor
iNOS inducible NO synthase
ETsellagitannins
DPPH2,2-diphenyl-1-picrylhydrazyl
ROSreactive oxygen species
ABTS2,29-azinobis-(3-ethylbenzo-thiazoline-6-sulfonic acid)
FRAPferric reducing ability of plasma
MRSAmethicillin-resistant Staphylococcus aureus
MSSAmethicillin-sensitive Staphylococcus aureus
MICminimum inhibitory concentration;
MBCminimum bactericidal concentration
GSHglutathione
GSSG glutathione disulfide
BMMsbone marrow macrophages
tBIDtruncated BID
HUVECshuman umbilical vein endothelial cells
MCF-7/wt MCF-7 human breast cancer cell (wild type)
FAformic acid
TBAthiobarbituric acid
ACNacetonitrile
AMCacceptable means of compliance
iNOSinducible nitric oxide synthase
TRAPtartrate-resistant acid phosphatase
ES/BSeroded surface/bone surface
BMMbone marrow-derived macrophages
NFATC1
Tmax
nuclear factor of activated T cells 1
time take to reach maximum concentration

References

  1. Newman, D.J.; Cragg, G.M. Natural products as sources of new drugs over the 30 years from 1981 to 2010. J. Nat. Prod. 2012, 75, 311–335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Atanasov, A.G.; Zotchev, S.B.; Dirsch, V.M.; Orhan, I.E.; Banach, M.; Rollinger, J.M.; Barreca, D.; Weckwerth, W.; Bauer, R.; Bayer, E.A.; et al. Natural products in drug discovery: Advances and opportunities. Nat. Rev. Drug Discov. 2021, 20, 200–216. [Google Scholar] [CrossRef] [PubMed]
  3. Kiss, A.K.; Piwowarski, J.P. Ellagitannins, gallotannins and their metabolites—The contribution to the anti-inflammatory effect of food products and medicinal plants. Curr. Med. Chem. 2016, 25, 4946–4967. [Google Scholar] [CrossRef]
  4. Riaz, A.; Rasul, A.; Hussain, G.; Zahoor, M.K.; Jabeen, F.; Subhani, Z.; Younis, T.; Ali, M.; Sarfraz, I.; Selamoglu, Z. Astragalin: A bioactive phytochemical with potential therapeutic activities. Adv. Pharmacol. Sci. 2018, 2018, 9794625. [Google Scholar] [CrossRef]
  5. Clifford, M.N.; Scalbert, A. Ellagitannins—Nature, occurrence and dietary burden. J. Sci. Food Agric. 2000, 80, 1118–1125. [Google Scholar] [CrossRef]
  6. Prothmann, J.; Sun, M.; Spégel, P.; Sandahl, M.; Turner, C.; Scheuba, J.; Wronski, V.K.; Rollinger, J.M.; Grienke, U.; Santos-Buelga, C.; et al. Relationship between phenolic compounds, anthocyanins content and antioxidant activity in colored barley germplasm. J. Agric. Food Chem. 2017, 53, 1713. [Google Scholar]
  7. Su, X.; Surry, D.S.; Spandl, R.J.; Spring, D.R. Total synthesis of sanguiin H-5. Org. Lett. 2008, 10, 2593–2596. [Google Scholar] [CrossRef]
  8. Niemetz, R.; Gross, G.G. Enzymology of gallotannin and ellagitannin biosynthesis. Phytochemistry 2005, 66, 2001–2011. [Google Scholar] [CrossRef]
  9. Feldman, K.S.; Sambandam, A. Ellagitannin chemistry. the first total chemical synthesis of an O(2),O(3)-Galloyl-Coupled ellagitannin, sanguiin H-5. J. Org. Chem. 1995, 60, 8171–8178. [Google Scholar] [CrossRef]
  10. Bakkalbasi, E.; Mentes, O.; Artik, N. Food ellagitannins-occurrence, effects of processing and storage. Crit. Rev. Food Sci. Nutr. 2009, 49, 283–298. [Google Scholar] [CrossRef]
  11. Koponen, J.M.; Happonen, A.M.; Mattila, P.H.; Törrönen, A.R. Contents of anthocyanins and ellagitannins in selected foods consumed in Finland. J. Agric. Food Chem. 2007, 55, 1612–1619. [Google Scholar] [CrossRef] [PubMed]
  12. Su, X.; Surry, D.; Spandl, R.; Spring, D. Synthesis of Sanguiin H-5. Synfacts 2008, 2008, 1130. [Google Scholar] [CrossRef]
  13. Nonaka, G.; Tanaka, T.; Nita, M.; Nishioka, I. A dimeric hydrolyzable tannin, sanguiin H-6 from Sanguisorba officinalis L. Chem. Pharm. Bull. 1982, 30, 2255–2257. [Google Scholar] [CrossRef]
  14. Jakimiuk, K.; Wink, M.; Tomczyk, M. Flavonoids of the Caryophyllaceae. Phytochem. Rev. 2021, 20, 1–40. [Google Scholar] [CrossRef]
  15. Landete, J.M. Ellagitannins, ellagic acid and their derived metabolites: A review about source, metabolism, functions and health. Food Res. Int. 2011, 44, 1150–1160. [Google Scholar] [CrossRef]
  16. Patel, A.; Rojas-Vera, J.; Dacke, C.G. Therapeutic constituents and actions of Rubus species. Curr. Med. Chem. 2004, 11, 1501–1512. [Google Scholar] [CrossRef]
  17. Moreno-Medina, B.L.; Casierra-Posada, F.; Cutler, J. Phytochemical composition and potential use of Rubus species. Gesunde Pflanz. 2018, 70, 65–74. [Google Scholar] [CrossRef]
  18. Okuda, T.; Yoshida, T.; Hatano, T.; Iwasaki, M.; Kubo, M.; Orime, T.; Yoshizaki, M.; Naruhashi, N. Hydrolysable tannins as chemotaxonomic markers in the Rosaceae. Phytochemistry 1992, 31, 3091–3096. [Google Scholar] [CrossRef]
  19. Duckstein, S.M.; Lotter, E.M.; Meyer, U.; Lindequist, U. Phenolic constituents from Alchemilla vulgaris L. and Alchemilla mollis (Buser) Rothm. at different dates of harvest. Z. Naturforsch. C 2012, 67, 529–540. [Google Scholar] [CrossRef] [Green Version]
  20. Luo, S.; Guo, L.; Sheng, C.; Zhao, Y.; Chen, L.; Li, C.; Jiang, Z.; Tian, H. Rapid identification and isolation of neuraminidase inhibitors from mockstrawberry (Duchesnea indica Andr.) based on ligand fishing combined with HR-ESI-Q-TOF-MS. Acta Pharm. Sin. B 2020, 10, 1846–1855. [Google Scholar] [CrossRef]
  21. Lee, S.-H.; Tanaka, T.; Nonaka, G.; Nishioka, I.; Zhang, B. Allose gallates from Euphorbia fischeriana. Phytochemistry 1991, 30, 1251–1253. [Google Scholar] [CrossRef]
  22. Vrhovsek, U.; Guella, G.; Gasperotti, M.; Pojer, E.; Zancato, M.; Mattivi, F. Clarifying the identity of the main ellagitannin in the fruit of the strawberry, Fragaria vesca and Fragaria ananassa Duch. J. Agric. Food Chem. 2012, 60, 2507–2516. [Google Scholar] [CrossRef] [PubMed]
  23. Mena, P.; Calani, L.; Dall’Asta, C.; Galaverna, G.; Garcia-Viguera, C.; Bruni, R.; Crozier, A.; Del Rio, D. Rapid and comprehensive evaluation of polyphenolic compounds in pomegranate (Punica granatum L.) juice by UHPLC-MS. Molecules 2012, 17, 14821–14840. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Chen, L.G.; Huang, W.T.; Lee, L.T.; Wang, C.C. Ellagitannins from Terminalia calamansanai induced apoptosis in HL-60 cells. Toxicol. Vitr. 2009, 23, 603–609. [Google Scholar] [CrossRef]
  25. Tanaka, T.; Tachibana, H.; Nonaka, G.; Nishioka, I.; Hsu, F.-L.; Kohda, H.; Tanaka, O. Tannins and related compounds. CXXII. New dimeric, trimeric and tetrameric ellagitannins, lambertianins A-D, from Rubus lambertianus SERINGE. Chem. Pharm. Bull. 1993, 41, 1214–1220. [Google Scholar] [CrossRef] [Green Version]
  26. Verma, R.; Gangrade, T.; Punasiya, R.; Ghulaxe, X. Rubus fruticosus (blackberry) use as an herbal medicine. Pharmacogn. Rev. 2014, 8, 101–104. [Google Scholar] [CrossRef] [Green Version]
  27. Krauze-Baranowska, M.; Majdan, M.; Hałasa, R.; Głód, D.; Kula, M.; Fecka, I.; Orzeł, A. The antimicrobial activity of fruits from some cultivar varieties of Rubus idaeus and Rubus occidentalis. Food Funct. 2014, 5, 2536–2541. [Google Scholar] [CrossRef]
  28. Yoshida, T.; Amakura, Y.; Yoshimura, M. Structural features and biological properties of ellagitannins in some plant families of the order myrtales. Int. J. Mol. Sci. 2010, 11, 79–106. [Google Scholar] [CrossRef] [Green Version]
  29. Aires, A. (Ed.) Tannins: Structural Properties, Biological Properties and Current Knowledge, 1st ed.; IntechOpen: London, UK, 2020; pp. 21–53. [Google Scholar]
  30. Zhu, M.; Dong, X.; Guo, M.; Ferreira, I.; McPhee, D.J. Phenolic profiling of Duchesnea indica combining macroporous resin chromatography (MRC) with HPLC-ESI-MS/MS and ESI-IT-MS. Molecules 2015, 20, 22463–22475. [Google Scholar] [CrossRef] [Green Version]
  31. Liberal, J.; Francisco, V.; Costa, G.; Figueirinha, A.; Amaral, M.T.; Marques, C.; Girão, H.; Lopes, M.; Cruz, M.T.; Teresa, B.M. Bioactivity of Fragaria vesca leaves through inflammation, proteasome and autophagy modulation. J. Ethnopharmacol. 2014, 158, 113–122. [Google Scholar] [CrossRef]
  32. Yoshida, T.; Tanaka, K.; Chen, X.-M.; Okuda, T. Dimeric ellagitannins, laevigatins E, F and G, from Rosa laevigata. Phytochemistry 1989, 28, 2451–2454. [Google Scholar] [CrossRef]
  33. Cui, C.; Zhao, Q.-C.; Cai, B.; Yao, X.-S.; Osadsa, H. Two new and four known polyphenolics obtained as new cell-cycle inhibitors from Rubus aleaefolius poir. J. Asian Nat. Prod. Res. 2002, 4, 243–252. [Google Scholar] [CrossRef] [PubMed]
  34. Hu, J.; Zhao, J.; Chen, W.; Lin, S.; Zhang, J.; Hong, Z. Hepatoprotection of 1β-hydroxyeuscaphic acid—The major constituent from Rubus aleaefolius against CCl4-induced injury in hepatocytes cells. Pharm. Biol. 2013, 51, 686–690. [Google Scholar] [CrossRef] [Green Version]
  35. Mertz, C.; Cheynier, V.; Günata, Z.; Brat, P. Analysis of phenolic compounds in two blackberry species (Rubus glaucus and Rubus adenotrichus) by high-performance liquid chromatography with diode array detection and electrospray ion trap mass spectrometry. J. Agric. Food Chem. 2007, 55, 8616–8624. [Google Scholar] [CrossRef] [PubMed]
  36. Gancel, A.; Feneuil, A.; Acosta, O.; Mercedes, A.; Vaillant, F. Impact of industrial processing and storage on major polyphenols and the antioxidant capacity of tropical highland blackberry (Rubus adenotrichus). Food Res. Int. J. 2011, 44, 2243–2251. [Google Scholar] [CrossRef]
  37. Hukkanen, A.; Kostamo, K.; Karenlampi, S.; Kokko, H. Impact of agrochemicals on Peronospora sparsa and phenolic profiles in three Rubus arcticus cultivars. J. Agric. Food Chem. 2008, 56, 1008–1016. [Google Scholar] [CrossRef]
  38. Martins, A.; Barros, L.; Carvalho, A.M.; Santos-Buelga, C.; Fernandes, I.P.; Barreiro, F.; Ferreira, I.C.F.R. Phenolic extracts of Rubus ulmifolius Schott flowers: Characterization, microencapsulation and incorporation into yogurts as nutraceutical sources. Food Funct. 2014, 5, 1091–1100. [Google Scholar] [CrossRef]
  39. Puupponen-Pimiä, R.; Nohynek, L.; Suvanto, J.; Salminen Juha-Pekka Seppänen-Laakso, T.; Tähtiharju, J.; Honkapää, K.; Oksman-Caldentey, K.-M. Natural antimicrobials from cloudberry (Rubus chamaemorus) seeds by sanding and hydrothermal extraction. ACS Foods Sci. Technol. 2001, 1, 917–927. [Google Scholar] [CrossRef]
  40. Wang, B.; Koivumäki, T.; Kylli, P.; Heinonen, M.; Poutanen, M. Protein-phenolic interaction of tryptic digests of β-lactoglobulin and cloudberry ellagitannin. J. Agric. Food Chem. 2014, 62, 5028–5037. [Google Scholar] [CrossRef]
  41. Thiem, B. Rubus chamaemorus L.—A boreal plant rich in biologically active metabolites: A review. Biol. Lett. 2003, 40, 3–13. [Google Scholar]
  42. Grochowski, D.M.; Paduch, R.; Wiater, A.; Dudek, A.; Pleszczynska, M.; Tomczykowa, M.; Granica, S.; Polak, P.; Tomczyk, M. In vitro antiproliferative and antioxidant effects of extracts from Rubus caesius leaves and their quality evaluation. Evid. Based Complement. Altern. Med. 2016, 2016, 5698685. [Google Scholar] [CrossRef]
  43. Oszmiański, J.; Wojdyło, A.; Nowicka, P.; Teleszko, M.; Cebulak, T.; Wolanin, M. Determination of phenolic compounds and antioxidant activity in leaves from wild Rubus L. species. Molecules 2015, 20, 4951–4966. [Google Scholar] [CrossRef] [Green Version]
  44. Gu, J.; Ahn-Jarvis, J.H.; Riedl, K.M.; Schwartz, S.J.; Clinton, S.K.; Vodovotz, Y. Characterization of black raspberry functional food products for cancer prevention human clinical trials. J. Agric. Food Chem. 2014, 62, 3997–4006. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Lim, T.K. Edible Medicinal and Non-Medicinal Plants: Fruits, 1st ed.; Springer: Dordrecht, The Netherlands, 2012; pp. 570–580. [Google Scholar]
  46. Krauze-Baranowska, M.; Głód, D.; Kula, M.; Majdan, M.; Hałasa, R.; Matkowski, A.; Kozłowska, W.; Kawiak, A. Chemical composition and biological activity of Rubus idaeus shoots—A traditional herbal remedy of Eastern Europe. BMC Complement. Altern. Med. 2014, 14, 1–12. [Google Scholar] [CrossRef] [PubMed]
  47. Sakai, E.; Aoki, Y.; Yoshimatsu, M.; Nishishita, K.; Iwatake, M.; Fukuma, Y.; Okamoto, K.; Tanaka, T.; Tsukuba, T. Sanguiin H-6, a constituent of Rubus parvifolius L., inhibits receptor activator of nuclear factor-κB ligand-induced osteoclastogenesis and bone resorption in vitro and prevents tumor necrosis factor-α-induced osteoclast formation in vivo. Phytomedicine 2016, 23, 828–837. [Google Scholar] [CrossRef]
  48. Jong-Won, C.; Yeong-Min, Y.; Min-Young, K.; Jung-Hwan, N.; Agung, N.; Hee-Juhn, P. Anti-hyperglycemic and anti-hyperlipidemic effects of the triterpenoid-rich fractions from Rubus coreanus and Rubus crataegifolius and their main component, niga-ichigoside f1, in streptozotocin-induced diabetic rats. Nat. Prod. Sci. 2008, 14, 260–264. [Google Scholar]
  49. Li, X.; Sun, J.; Chen, Z.; Jiang, J.; Jackson, A. Characterization of carotenoids and phenolics during fruit ripening of Chinese raspberry (Rubus chingii Hu). RSC Adv. 2021, 11, 10804–10813. [Google Scholar] [CrossRef]
  50. Jia-Yun, S.; Si-Qi, W.; Kao-Hua, L.; Bo, Z.; Qiao-Yan, Z.; Lu-Ping, Q.; Jian-Jun, W. Rubus chingii Hu: An overview of botany, traditional uses, phytochemistry, and pharmacology. Chin. J. Nat. Med. 2020, 18, 401–4016. [Google Scholar]
  51. Sun, Z.-L.; Zhang, Y.; Wan, A.-H.; Zhang, X.-L.; Feng, J. A new active compound against kidney deficiency from the fruits of Rubus corchorifolius. J. Asian Nat. Prod. Res. 2011, 13, 68–74. [Google Scholar] [CrossRef]
  52. Gasperotti, M.; Masuero, D.; Vrhovsek, U.; Guella, G.; Mattivi, F. Profiling and accurate quantification of Rubus ellagitannins and ellagic acid conjugates using direct UPLC-Q-TOF hdms and HPLC-DAD analysis. J. Agric. Food Chem. 2010, 58, 4602–4616. [Google Scholar] [CrossRef]
  53. Piwowarski, J.P.; Granica, S.; Zwierzyńska, M.; Stefańska, J.; Schopohl, P.; Melzig, M.F.; Kiss, A.K. Role of human gut microbiota metabolism in the anti-inflammatory effect of traditionally used ellagitannin-rich plant materials. J. Ethnopharmacol. 2014, 155, 801–809. [Google Scholar] [CrossRef]
  54. Zhang, G.; Liu, Y.; Hai, P. The complete chloroplast genome of tibetan medicinal plant Rubus phoenicolasius Maxim. Mitochondrial DNA Part B 2021, 6, 886–887. [Google Scholar] [CrossRef]
  55. Kool, M.M.; Comeskey, D.J.; Cooney, J.M.; McGhie, T.K. Structural identification of the main ellagitannins of a boysenberry (Rubus loganbaccus × baileyanus Britt.) extract by LC-ESI-MS/MS, MALDI-TOF-MS and NMR spectroscopy. Food Chem. 2010, 119, 1535–1543. [Google Scholar] [CrossRef]
  56. Lee, Y.A.; Lee, M.W. Tannins from Rubus coreanum. Korean J. Pharmacogn. 1995, 26, 27–30. [Google Scholar]
  57. Pang, K.C.; Kim, M.S.; Lee, M.W. Hydrolyzable tannins from the fruits of Rubus coreanum. Korean J. Pharmacogn. 1996, 27, 366–370. [Google Scholar]
  58. Kim, M.S.; Pang, K.C.; Lee, S.M. Tannins from the leaves of Rubus coreanum. Korea Sci. 1996, 40, 666–669. [Google Scholar]
  59. Kim, L.S.; Youn, S.H.; Kim, J.Y. Comparative study on antioxidant effects of extracts from Rubus coreanus and Rubus occidentalis Lee. J. Korean Soc. Food Sci. Nutr. 2014, 43, 1357–1362. [Google Scholar] [CrossRef] [Green Version]
  60. Kosmala, M.; Jurgoński, A.; Juśkiewicz, J.; Karlińska, E.; Macierzuński, J.; Rój, E.; Zduńczyk, Z. Chemical composition of blackberry press cake, polyphenolic extract, and defatted seeds, and their effects on cecal fermentation, bacterial metabolites, and blood lipid profile in rats. J. Agric. Food Chem. 2017, 65, 5470–5479. [Google Scholar] [CrossRef]
  61. Sparzak, B.; Merino-Arevalo, M.; Vander Heyden, Y.; Krauze-Baranowska, M.; Majdan, M.; Fecka, I.; Głód, D.; Ba̧czek, T. HPLC analysis of polyphenols in the fruits of Rubus idaeus L. (Rosaceae). Nat. Prod. Res. 2010, 24, 1811–1822. [Google Scholar] [CrossRef]
  62. Tavares, L.; Figueira, I.; McDougall, G.; Vieira, H.; Stewart, D.; Alves, P.; Ferreira, R.; Santos, C. Neuroprotective effects of digested polyphenols from wild blackberry species. Eur. J. Nutr. 2013, 52, 225–236. [Google Scholar] [CrossRef]
  63. Nonaka, G.I.; Tanaka, T.; Nishioka, I. Tannins and related compounds. Part 3. A new phenolic acid, sanguisorbic acid dilactone, and three new ellagitannins, sanguiins H-1, H-2, and H-3, from Sanguisorba officinalis. J. Chem. Soc. Perkin Trans. 1 1982, 1067–1073. [Google Scholar] [CrossRef]
  64. Zhao, Z.; He, X.; Zhang, Q.; Wei, X.; Huang, L.; Fang, J.; Wang, X.; Zhao, M.; Bai, Y.; Zheng, X. Traditional uses, chemical constituents and biological activities of plants from the genus Sanguisorba L. Am. J. Chin. Med. 2017, 45, 199–224. [Google Scholar] [CrossRef] [PubMed]
  65. Arun, N.; Road, R.; Pradesh, U. Punica granatum: A review on pharmacological and therapeutic properties. Int. J. Pharm. Sci. Res. 2012, 3, 1240–1245. [Google Scholar]
  66. Lia, Y.-N.; He, J.; Zhang, J.; Shi, Y.-X.; Guo, L.-B.; Peng, Z.-C.; Yang, T.; Ding, K.; Zhang, W.-K.; Xu, J.-K. Existing knowledge on Euphorbia fischeriana Steud. (Euphorbiaceae): Traditional uses, clinical applications, phytochemistry, pharmacology and toxicology. J. Ethnopharmacol. 2021, 275, 114095. [Google Scholar] [CrossRef] [PubMed]
  67. Juszczak, A.M.; Zovko-Končić, M.; Tomczyk, M. Recent trends in the application of chromatographic techniques in the analysis of luteolin and its derivatives. Biomolecules 2019, 9, 731. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Kashiwada, Y.; Nonaka, G.I.; Nishioka, I.; Chang, J.J.; Lee, K.H. Tannins and related compounds as selective cytotoxic agents. J. Nat. Prod. 1992, 55, 1033–1043. [Google Scholar] [CrossRef]
  69. Jang, E.; Inn, K.S.; Jang, Y.P.; Lee, K.T.; Lee, J.H. Phytotherapeutic activities of Sanguisorba officinalis and its chemical constituents: A review. Am. J. Chin. Med. 2018, 46, 299–318. [Google Scholar] [CrossRef]
  70. Konishi, K.; Urada, M.; Adachi, I.; Tanaka, T. Inhibitory effect of sanguiin H-11 on chemotaxis of neutrophil. Biol. Pharm. Bull. 2000, 23, 213–218. [Google Scholar] [CrossRef] [Green Version]
  71. Sangiovanni, E.; Vrhovsek, U.; Rossoni, G.; Colombo, E.; Brunelli, C.; Brembati, L.; Trivulzio, S.; Gasperotti, M.; Mattivi, F.; Bosisio, E.; et al. Ellagitannins from Rubus berries for the control of gastric inflammation: In vitro and in vivo studies. PLoS ONE 2013, 8, e71762. [Google Scholar] [CrossRef] [Green Version]
  72. Yokozawa, T.; Chen, C.P.; Rhyu, D.Y.; Tanaka, T.; Park, J.C.; Kitani, K. Potential of sanguiin H-6 against oxidative damage in renal mitochondria and apoptosis mediated by peroxynitrite in vivo. Nephron 2002, 92, 133–141. [Google Scholar] [CrossRef]
  73. Mullen, W.; McGinn, J.; Lean, M.E.J.; MacLean, M.R.; Gardner, P.; Duthie, G.G.; Yokota, T.; Crozier, A. Ellagitannins, flavonoids, and other phenolics in red raspberries and their contribution to antioxidant capacity and vasorelaxation properties. J. Agric. Food Chem. 2002, 50, 5191–5196. [Google Scholar] [CrossRef]
  74. Song, J.H.; Kim, S.Y.; Hwang, G.S.; Kim, Y.S.; Kim, H.Y.; Kang, K.S. Sanguiin H-11 from Sanguisorbae radix protects HT22 murine hippocampal cells against glutamate-induced death. Bioorgan. Med. Chem. Lett. 2019, 29, 252–256. [Google Scholar] [CrossRef] [PubMed]
  75. Kähkönen, M.; Kylli, P.; Ollilainen, V.; Salminen, J.P.; Heinonen, M. Antioxidant activity of isolated ellagitannins from red raspberries and cloudberries. J. Agric. Food Chem. 2012, 60, 1167–1174. [Google Scholar] [CrossRef] [PubMed]
  76. Yokozawa, T.; Chen, C.P.; Tanaka, T.; Kitani, K. Effects of sanguiin H-6, a component of Sanguisorbae radix, on lipopolysaccharide-stimulated nitric oxide production. Biochem. Pharmacol. 2002, 63, 853–858. [Google Scholar] [CrossRef]
  77. Puljula, E.; Walton, G.; Woodward, M.J.; Karonen, M. Antimicrobial activities of ellagitannins against Clostridiales perfringens, Escherichia coli, Lactobacillus plantarum and Staphylococcus aureus. Molecules 2020, 25, 3714. [Google Scholar] [CrossRef] [PubMed]
  78. Da Silva, L.P.; Pereira, E.; Pires, T.C.S.P.; Alves, M.J.; Pereira, O.R.; Barros, L.; Ferreira, I.C.F.R. Rubus ulmifolius Schott fruits: A detailed study of its nutritional, chemical and bioactive properties. Food Res. Int. 2019, 119, 34–43. [Google Scholar] [CrossRef] [Green Version]
  79. Park, E.J.; Lee, D.; Baek, S.E.; Kim, K.H.; Kang, K.S.; Jang, T.S.; Lee, H.L.; Song, J.H.; Yoo, J.E. Cytotoxic effect of sanguiin H-6 on MCF-7 and MDA-MB-231 human breast carcinoma cells. Bioorgan. Med. Chem. Lett. 2017, 27, 4389–4392. [Google Scholar] [CrossRef]
  80. Bhatia, S.; Giri, S.; Singh, S.; Lal, A.F. Identification of potential inhibitors of dietary polyphenols for SARS-CoV-2 M protease: An in silico study. One Health Bull. 2020, 1, 21–29. [Google Scholar]
  81. Trinh, T.A.; Park, E.-J.; Lee, D.; Song, J.H.; Lee, H.L.; Kim, K.H.; Kim, Y.; Jung, K.; Kang, K.S.; Yoo, J.-E. Estrogenic activity of sanguiin H-6 through activation of estrogen receptor α coactivator-binding site. Nat. Prod. Sci. 2019, 25, 28. [Google Scholar] [CrossRef]
  82. Bastow, K.F.; Bori, I.D.; Fukushima, Y.; Kashiwada, Y.; Tanaka, T.; Nonaka, G.; Nishioka, I.; Lee, K.H. Inhibition of DNA topoisomerases by sanguiin H-6, a cytotoxic dimeric ellagitannin from Sanguisorba officinalis. Planta Med. 1993, 59, 240–245. [Google Scholar] [CrossRef]
  83. Lee, S.J.; Lee, H.K. Sanguiin H-6 blocks endothelial cell growth through inhibition of VEGF binding to VEGF receptor. Arch. Pharm. Res. 2005, 28, 1270–1274. [Google Scholar] [CrossRef] [PubMed]
  84. Ko, H.; Jeon, H.; Lee, D.; Choi, H.K.; Kang, K.S.; Choi, K.C. Sanguiin H6 suppresses TGF-β induction of the epithelial-mesenchymal transition and inhibits migration and invasion in A549 lung cancer. Bioorgan. Med. Chem. Lett. 2015, 25, 5508–5513. [Google Scholar] [CrossRef]
  85. Berdowska, I.; Zieliński, B.; Saczko, J.; Sopel, M.; Gamian, A.; Fecka, I. Modulatory impact of selected ellagitannins on the viability of human breast cancer cells. J. Funct. Foods 2018, 42, 122–128. [Google Scholar] [CrossRef]
  86. Park, E.-H.H.; Park, J.Y.; Yoo, H.-S.S.; Yoo, J.-E.E.; Lee, H.L. Assessment of the anti-metastatic properties of sanguiin H-6 in HUVECs and MDA-MB-231 human breast cancer cells. Bioorgan. Med. Chem. Lett. 2016, 26, 3291–3294. [Google Scholar] [CrossRef] [PubMed]
  87. Lee, D.; Ko, H.; Kim, Y.-J.; Kim, S.-N.; Choi, K.-C.; Yamabe, N.; Kim, K.H.; Kang, K.S.; Kim, H.Y.; Shibamoto, T. Inhibition of A2780 human ovarian carcinoma cell proliferation by a Rubus component, sanguiin H-6. J. Agric. Food Chem. 2016, 64, 801–880. [Google Scholar] [CrossRef]
  88. Yokozawa, T.; Chen, C.P.; Tanaka, T. Direct scavenging of nitric oxide by traditional crude drugs. Phytomedicine 2000, 6, 453–463. [Google Scholar] [CrossRef]
  89. Frieri, M.; Kumar, K.; Boutin, A. Antibiotic resistance. J. Infect. Public Health 2017, 10, 369–378. [Google Scholar] [CrossRef] [Green Version]
  90. Oh, D.Y.; Hurt, A.C. A review of the antiviral susceptibility of human and avian influenza viruses over the last decade. Scientifica 2014, 2014, 430629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Tu, Y.-F.; Chien, C.-S.; Yarmishyn, A.A.; Lin, Y.-Y.; Luo, Y.-H.; Lin, Y.-T.; Lai, W.-Y.; Yang, D.-M.; Chou, S.-J.; Yang, Y.-P.; et al. A review of SARS-CoV-2 and the ongoing clinical trials. Int. J. Mol. Sci. 2020, 21, 2657. [Google Scholar] [CrossRef] [Green Version]
  92. Lim, Y.X.; Ng, Y.L.; Tam, J.P.; Liu, D.X. Human coronaviruses a review of virus-host interactions. Diseases 2016, 4, 26. [Google Scholar] [CrossRef]
  93. Arnica, F.; Sabeena, G.; Sonam, B.; Shaminder, S.; Sonia, M. Identification of ellagitannins as natural inhibitors of spike proteins of COVID19 virus: An in silico- based study for drug development. Afr. J. Health Sci. 2020, 33, 78–97. [Google Scholar]
  94. Stoner, G.D.; Sardo, C.; Apseloff, G.; Mullet, D.; Wargo, W.; Pound, V.; Singh, A.; Sanders, J.; Aziz, R.; Casto, B.; et al. Pharmacokinetics of anthocyanins and ellagic acid in healthy volunteers fed freeze-dried black raspberries daily for 7 days. J. Clin. Pharmacol. 2005, 45, 1153–1164. [Google Scholar] [CrossRef]
  95. Cho, J.M.; Chae, J.; Jeong, S.R.; Moon, M.J.; Ha, K.C.; Kim, S.; Lee, J.H. The cholesterol-lowering effect of unripe Rubus coreanus is associated with decreased oxidized LDL and apolipoprotein B levels in subjects with borderline-high cholesterol levels: A randomized controlled trial. Lipids Health Dis. 2020, 19, 1–12. [Google Scholar] [CrossRef] [PubMed]
  96. Lipińska, L.; Klewicka, E.; Sójka, M. The structure, occurrence and biological activity of ellagitannins: A general review. Acta Sci. Pol. Technol. Aliment. 2014, 13, 289–299. [Google Scholar] [CrossRef] [Green Version]
  97. Espín, J.C.; Larrosa, M.; García-Conesa, M.T.; Tomás-Barberán, F. Biological significance of urolithins, the gut microbial ellagic acid-derived metabolites: The evidence so far. Evid.-Based Complement. Altern. Med. 2013, 2013, 270418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Ludwig, I.A.; Mena, P.; Calani, L.; Borges, G.; Pereira-Caro, G.; Bresciani, L.; Del Rio, D.; Lean, M.E.J.; Crozier, A. New insights into the bioavailability of red raspberry anthocyanins and ellagitannins. Free Radic. Biol. Med. 2015, 89, 758–769. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Sanguiins presented in natural sources. (1) Sanguiin H-1, (2) sanguiin H-2, (3) sanguiin H-3, (4) sanguiin H-4, (5) sanguiin H-6, (6) sanguiin H-10, and (7) sanguiin H-11.
Figure 1. Sanguiins presented in natural sources. (1) Sanguiin H-1, (2) sanguiin H-2, (3) sanguiin H-3, (4) sanguiin H-4, (5) sanguiin H-6, (6) sanguiin H-10, and (7) sanguiin H-11.
Ijms 22 12972 g001
Table 1. Plants containing sanguiin and its traditional uses.
Table 1. Plants containing sanguiin and its traditional uses.
PlantFamilyGeographical LocationType of SHAmount of SHTraditional Medicine UsesReferences
Alchemilla vulgarisRosaceaeGermanySH6
SH10
isomers
not givenwounds, eczema, and inflamed mucosa[19]
Alchemilla mollisRosaceaeGermanySH6
SH10
isomers
not givenwounds, eczema, and inflamed mucosa[19]
Duchesnea indicaRosaceaeChinaSH40.0046 mg/g of dried fruitsfever, inflammation, cancer[20,30]
Fragaria vescaRosaceaeItalySH6not giveninflammation-related diseases[22,31]
Fragaria ananassaRosaceaeItalySH6not givennot found[22]
Rosa laevigataRosaceaeChinaSH40.03 mg/g of dried pecarpsurinary incontinence, diarrhea, pain, burns, toothache[32]
Rubus aleaefoliusRosaceaeChinaSH2
ethyl ester
0.028 mg/g of dried rootsvarious types of hepatitis[33,34]
Rubus adenotrichusRosaceaeCosta Rica, Trinidad
Ecuador
SH64.2 mg/g of dried berries not found[35,36]
Rubus arcticusRosaceaepropagated
vegetatively
SH5
SH6
SH10
not givennot found[37]
Rubus ulmifoliusRosaceaePortugalSH10
isomer
not givendiarrhea, menstrual pain, menopause disorders, liver diseases, aphtha, gingivitis[38]
Rubus chamaemorusRosaceaeFinlandSH6
SH10
isomers
not givenscurvy and diarrhea[39,40,41]
Rubus caesiusRosaceaePolandSH65.79 mg/g of dried leavesuterine relaxant, stimulant during confinement, diarrhea and similar enteric disorders, an astringent[16,42,43]
Rubus hirsutusRosaceaeJapanSH6
SH11
73.92 mg/g of dried leavesnot found[18]
not given
Rubus occidentalisRosaceaePolandSH610.78–50.45 mg/g of plant extract from shootscommon cold, fever and flu-like infections, management of impotence, spermatorrhea, enuresis, asthma, allergic diseases[27,44,45,46]
Rubus lambertianusRosaceaeTaiwan, JapanSH2
SH6
SH11
not givennot found[18,25]
Rubus parvifoliusRosaceaeJapanSH2
SH6
SH11
not givenfever, angina, enteritis, hepatitis, concretion, eczema, rheumatism[18,25,47]
Rubus crataegifoliusRosaceaeJapanSH2
SH6
SH11
not givendiabetes mellitus[18,25,48]
Rubus pedatusRosaceaeJapanSH6
SH11
not givennot found[18]
Rubus palmatusRosaceaeJapanSH2
SH6
SH11
not givennot found[18,25]
Rubus chingiiRosaceaeJapanSH2
SH6
SH11
not giveninvigorating Qi, losing weight, blackening hair, tonifying kidney, enriching essence, impotence [18,25,49,50]
Rubus sieboldiiRosaceaeJapanSH2not givennot found[25]
Rubus corchorifoliusRosaceaeJapanSH2not givenimpotence, seminal
emission
[25,51]
Rubus palmatus var.
coptophyllus
RosaceaeJapanSH2not givennot found[25]
Rubus idaeusRosaceaeJapan
Poland
Italy
SH2not givenenterocolitis, bronchitis, prostate disorders, analgesic, cold, cough, fever[25,27,52,53]
SH61.7–6.33 mg/g of plant extract from shoots
Rubus mesogeanusRosaceaeJapanSH2not givennot found[25]
Rubus calycinoidesRosaceaeTaiwanSH2not givennot found[25]
Rubus phoenicolasiusRosaceaeJapanSH2
SH6
SH11
not givenrheumatism, irregular menstruation, kidney ailments[18,25,54]
Rubus loganbaccus x Rubus baileyanusRosaceaeNew ZealandSH2
SH6
SH10
not givennot found[55]
Rubus glaucusRosaceaeTrinidad, Costa Rica, Ecuador SH62.45 mg/g of dried berries diarrhea, wounds, burns[17,35]
Rubus coreanusRosaceaeKorea,
Japan
SH2
SH5
SH4
SH6
SH11
not givenimpotence, pollution, premature ejaculation, frequency of urination[18,56,57,58,59,60]
Rubus fruticosusRosaceaePoland,
Japan
SH61.35–5.47
mg/g of dried
berries
dysentery, diarrhea,
whooping cough, colitis,
toothache, pain
[18,26,61]
SH11not given
SH2 isomernot given
Rubus irirasemRosaceaeJapanSH6
SH11
not givennot found[18]
Rubus hiraseanusRosaceaeJapanSH6
SH11
not givennot found[18]
Rubus vagabundusRosaceaePortugalSH2
SH6
SH10
not givennot found[62]
Rubus brigantinusRosaceaePortugalSH2
SH6
SH10
not givennot found[62]
Rubus radulaRosaceaePolandSH616.66 mg/g of dried leavesnot found[43]
Rubus montanusRosaceaePolandSH616.95 mg/g of dried leavesnot found[43]
Rubus gracilisRosaceaePolandSH618.07 mg/g of dried leavesnot found[43]
Rubus macrophyllusRosaceaePolandSH614.48 mg/g of dried leavesnot found[43]
Rubus pericrispatusRosaceaePolandSH614.49 mg/g of dried leavesnot found[43]
Rubus subcatusRosaceaePolandSH659.79 mg/g of dried leavesnot found[43]
Rubus ambrosiusRosaceaePolandSH621.11 mg/g of dried leavesnot found[43]
Rubus fasciculatusRosaceaePolandSH623.24 mg/g of dried leavesnot found[43]
Rubus nessensisRosaceaePolandSH612.22 mg/g of dried leavesnot found[43]
Rubus glivicensisRosaceaePolandSH648.46 mg/g of dried leavesnot found[43]
Rubus bifronusRosaceaePolandSH639.48 mg/g of dried leavesnot found[43]
Rubus praecoxRosaceaePolandSH618.49 mg/g of dried leavesnot found[43]
Rubus perrobustusRosaceaePolandSH653.02 mg/g of dried leavesnot found[43]
Rubus parthenocissusRosaceaePolandSH611.41 mg/g of dried leavesnot found[43]
Rubus pseudidaeusRosaceaePolandSH615.07 mg/g of dried leavesnot found[43]
Rubus constrictusRosaceaePolandSH624.38 mg/g of dried leavesnot found[43]
Rubus wimmerianusRosaceaePolandSH664.44 mg/g of dried leavesnot found[43]
Rubus orthostachysRosaceaePolandSH645.60 mg/g of dried leavesnot found[43]
Rubus plicatusRosaceaePolandSH658.48 mg/g of dried leavesnot found[43]
Rubus pedemontanusRosaceaePolandSH663.51 mg/g of dried leavesnot found[43]
Rubus grabowskiRosaceaePolandSH649.77 mg/g of dried leavesnot found[43]
Sanguisorba tenuifolia var. parvifloraRosaceaeJapanSH2
SH11
not givennot found[25]
Sanguisorba officinalisRosaceaeJapanSH1not givenleukopenia,
hemorrhaging, burns
[13,25,63,64]
SH2not given
SH3not given
SH61.6 mg/g of dried leaves
SH11not given
Sanguisorba tenuijolia
var. alba
RosaceaeJapanSH6
SH11
not givennot found[18]
Punica granatumLythraceaeSpainSH10
isomers
not giveninflammation, rheumatism, pain, snakebites, diabetes, burns, leprosy, vermifugal and taenicidal agent[23,65]
Euphorbia
fischeriana
EuphorbiaceaeChinaSH50.072 mg/g of dried rootsdyspepsia, abdominal distension, abdominal pain, cough, external applications as a cure for scabies and tuberculosis of lymph nodes[21,66]
Terminalia
calamansanai
CombretaceaeTaiwanSH40.098 mg/g of dried leaveslithotriptic[24]
Table 2. Chromatographic techniques for the analysis of sanguiins.
Table 2. Chromatographic techniques for the analysis of sanguiins.
CompoundStationary Phase/ColumnMobile PhaseConditions (Flow Rate, Injection Volume)DetectionReferences
SH6, SH10 isomersSunFire C18 RP1% FA and ACN/H2O (9:1, v/v)0.21 mL/min; 5 μL280 nm[19]
SH4Phenomenex Gemini C18; Waters Symmetry C18; Phenomenex Kinetex C18; Phenomenex Luna C181% FA and MeOH1–15 mL/min310 nm[20]
Toyopearl HW-40F70% MeOH--[32]
LiChroprep RP C180.05% TFA and CH3CN (95:5)1 mL/min280 nm[24]
SH2, ethyl esterODSMeOH–H2O (35:65)--[33]
SH6Lichrospher ODS-2 RP 2% FA and ACN/H2O/FA (80:18:2, v/v/v)0.5 mL/min; 10 μL200–600 nm[35]
Discovery HS C18 0.1% TFA and 0.1% TFA in a mixture of H2O:ACN (50:50 v/v)0.3 mL/min; 1 μL520 nm[27]
Fuji-gel ODS-G3MeOH–H2O (7:3)--[25]
UPLC BEH C18 4.5% FA and ACN0.45 mL/min; 10 μL240 nm[43]
SH5, SH6, SH10ODS Hypersil ACN and 1% FA 2 mL/min; 15 μL280 nm[37]
SH10 isomerSpherisorb S3 ODS-2 C181% FA and ACN0.5 mL/min;280 nm[38]
BlueOrchid C18; Hypersil Gold C18; Kinetex PFPACN + 1% FA and H2O0.2 mL/min; 5 μL-[23]
SH2MCI-gel CHP 20Pmixture of MeOH and H2O--[25]
SH5Sephadex LH-20mixture of MeOH and H2O--[21]
SH6, SH11Superspher Si 60hexane-MeOH-THF-HCO2H + (COH)2O1.5 mL/min;280 nm[18]
SH2, SH6, SH10Synergy Hydro RP C18 ACN:H2O10 mL/min; 50–200 μL280 nm[55,62]
Table 3. Bioactivities of sanguiins reported in in vitro and in vivo experimental models.
Table 3. Bioactivities of sanguiins reported in in vitro and in vivo experimental models.
ActivityExperimental ModelExposureConcentrationEfficacyReferences
Anti-inflammatoryRat neutrophils60 min chemotaxis and 2 h toxicity in
in vitro assays
0, 1, 2.5, 5, and 10 μM SH11, SH6, and SH2
  • IC50 of SH2, SH6 and SH11 of inhibitory activity on CINC-1-dependent neutrophil chemotaxis was about: 10, 4, and 2.5 μM, respectively
  • 95% of the cells were living after 2 h-incubation with sanguiins
[70]
Human AGS gastric
epithelial cells
1 h for NF-κB nuclear translocation, 6 h for NF-κB-driven transcription, and 6 h for IL-8 release in in vitro assays0.25–10 μM
SH6
  • IC50: 0.87 ± 0.16 µM—without stimulation and 1.9 ± 0.23 µM with IL-1β
  • IC50: 1.5 ± 0.35 µM—TNFα stimulated and 2.7 ± 0.30 µM—IL-1β stimulated
  • At 2.5 µM SH6 completely inhibited release of IL-8 with IC50: 0.58 ± 0.05 μM—TNFα-induced and 1.03 ± 0.06 μM—IL-1β-induced
[71]
AntioxidantMale LWH Wistar ratsIn vivo, rats were fed orally with SH6 for 30 days10 mg/kg body weight/day
  • Level of 3-nitrotyrosine in plasma reduced from 607.6 ± 15.6 to 294.8 ± 26.1 pmol/mL
  • TBA-reactive substance decreased from 1.31 ± 0.30 to 0.83 ± 0.14 nmol/mg protein
  • GSH level increased from 1.44 ± 0.25 to 2.44 ± 0.26 nmol/mg (sham treatment—3.35 ± 0.25)
  • Glutathione peroxidase level increased from 107.6 ± 5.2 to 115.6 ± 6.0 U/mg (sham treatment—141.3 ± 16.0)
  • DNA fragmentation level decreased from 23.4% ± 2.0% to 16.9% ± 1.6%
  • Caspase-3 decreased from 8.26 ± 0.71 to 5.95 ± 0.36 pmol AMC/mg protein/min
  • Urea nitrogen decreased from 75.2 ± 3.1 to 59.5 ± 2.3 mg/dL
  • Cr decreased from 1.84 ± 0.13 to 1.34 ± 0.12 mg/dL
[72]
Fremy’s salt20 min electron spin
resonance spectroscopy in situ assay
extracts diluted to 5% (v/v) with ethanol and water (12:88, v/v);
1.0 mL portion
  • 1.7 × 1017 per gram f.w. Fremy’s radicals reduced by SH6
[73]
HT22 murine hippocampal cells8 h in vitro assay0, 10, and 20 μM SH11
  • Intracellular ROS: viability of cells I creased at a concentration: 20 µM (glutamine present), 10 µM (glutamine absent), and 20 µM (glutamine absent).
  • At a 10 µM with glutamine present observed slight decrease in viability
[74]
DPPH, methyl linoleate and diene hydroperoxide15 min, 72 h, and 2 h in situ assays2, 5, 10, 50, and 250 μM of
raspberry
ET dimers and trimers
  • DPPH test (ROS %):
    • 2 μM: raspberry ET dimers and trimers: 20 ± 0.4; cloudberry ET dimers and trimers: 21 ± 0.1
    • 5 μM: raspberry ET dimers and trimers: 40 ± 0.1; cloudberry ET dimers and trimers: 47 ± 0.2
    • 10 μM: raspberry ET dimers and trimers: 79 ± 0.3; cloudberry ET dimers and trimers: 74 ± 1.7
  • Methyl linoleate: inhibition %
    • 50 μM; raspberry ET dimers and trimers: 24 ± 4.9; cloudberry ET dimers and trimers: 21 ± 4.9
    • 100 μM: raspberry ET dimers and trimers: 37 ± 0.0; cloudberry ET dimers and trimers: 13 ± 3.2
    • 250 μM: raspberry ET dimers and trimers: 37 ± 3.2; cloudberry ET dimers and trimers: 59 ± 3.2
  • Emulsion: inhibition (%) of conjugate diene hydroperoxide formation:
    • 50 μM: raspberry ET dimers and trimers: 90 ± 0.7; cloudberry ET dimers and trimers: 91 ± 0.0
    • 250 μM: raspberry ET dimers and trimers: 96 ± 0.4; cloudberry ET dimers and trimers: 95 ± 0.0
[75]
ABTS and FRAP
assays
6 min in situ ABTS assay, 8 min in situ FRAP assaynot given
  • ABTS radical scavenging (mmol TE/g dm): R. pedemontanus—212.69 and R. parthenocisus—c.a. 203
  • FRAP ability: R. pedemontanus—192.91 and R. parthenocissus—192.53
[43]
mice
macrophage
and sodium nitroprusside
24 in vitro
macrophage incubation,
150 min in situ
sodium
nitroprusside
assay
0, 12.5, 25, and
50 μM of SH6
in macrophage assay, 0, 2.5, 5, 12.5, 25, 50,
and 100 μM
of SH6 in sodium nitroprusside assay
  • Macrophage’s assay (µM):
    • Nitrite level reduced above 50% at concentrations 12.5, 25, and 50
    • Cell viability (%) increased at concentrations 12.5, 25, and 50
    • The enzymatic activity of iNOS (pmol/mg protein/min) was: 12.5 µM SH6-19.98; 25 µM SH6—9.80; 50 µM SH6—7.01
  • Decreased NO generation from sodium nitroprusside: 0 μM: 13.15 ± 0.11; (2.5 μM): 8.29 ± 0.07; 5 μM: 8.16 ± 0.09; 12.5 μM: 8.07 ± 0.10; 25 μM: 7.69 ± 0.07; 50 μM: 6.91 ± 0.10; 100 μM: 4.78 ± 0.05
[76]
Osteoclastogenesis inhibitory8-week-old male C57BL/6J miceintraperitoneal injections for
5 days
10 μg/body weight(g)/day
of SH6
  • Mice treated with both TNF-α and SH6—TRAP-positive amount of osteoclasts significantly reduced and the percentage of ES/BS (eroded surface/bone surface)
[47]
bone marrow macrophages (BMMs)72 h
in vitro
assay
0, 1, 5, 10, and
25 μM of SH6
  • SH6 at concentrations >5 μM downregulated the expression of NFATc1 and its target proteins, c-Src, and cathepsin K
RAW-D cells72 h
in vitro
assay
5 μM of SH6
  • SH6 strongly inhibited the nuclear translocation of NFATc1, phosphorylated-c-Fos, and NF-κB
BMMs and RAW-D cells72 h
in vitro
assay
0–50 μM
of SH6 in BMM and RAW-D
cells assays
  • Dose-dependent inhibition of multinucleated osteoclast formation in BMM cells; cytotoxicity was observed at 25 and 50 μM. The number of TRAP-positive RAW-D-derived osteoclasts decreased significantly after treatment with >0.1 μM SH6; cytotoxicity was observed at >10 μM SH6
AntibacterialStreptococcus group A, B, C
S. pneumoniae
E. faecalis
C. diphtheriae
B. subtilis
C. sporogenes
S. aureus
S. epidermidis
N. meningitidis
M. catarrhalis
H. influenzae
H. pylori
K. pneumoniae
48 h
in vitro
assay
SH6
concentrations: geometric
progression from 0.015 to
1 mg/mL
  • MIC (mg/mL):
    • Streptococcus group A: 0.5
    • S. pneumoniae: 0.5, C. diphtheriae: 0.03
    • B. subtilis: 0.5
    • C. sporogenes: 0.06
    • S. aureus: 0.25
    • S. epidermidis: 0.125
    • M. catarrhalis: 0.5
2.
MBC (mg/mL):
  • Streptococcus group A: 0.5
  • S. pneumoniae: 0.5
  • C. diphtheriae: 0.03
  • S. epidermidis: 0.125
[27]
C. perfringens
E. coli
L. plantarum
S. aureus
24 h in vitro
incubation
0.5 mM
of SH6
  • S. aureus inhibition: reduction in the growth from 109 CFU/mL to 103 CFU/mL
  • E. coli inhibition: reduction in the growth from 109 CFU/mL to 107 CFU/mL
  • 3.L. plantarum inhibition: reduction in the growth from 8.0 × 108 CFU/mL to 6.0 × 108 CFU/mL
  • 4.C. perfringens inhibition: reduction in the growth from 7.0 × 108 CFU/mL to 2.0 × 108 CFU/mL
[77]
E. coli, E. faecalis
K. pneumoniae,
M. morganii,
P. mirabilis,
P. aeruginosa,
L. monocytogenes,
MRSA, MSSA
not given100 mg/mL (stock solution) R. ulmifolius
extract; SH10: 9.6 ± 0.1 mg/g
  • MIC: E. coli, M. morganii, E. faecalis
    L. monocytogenes, MSSA: 5 mg/mL
    Proteus mirabilis, MRSA: 10 mg/mL
    P. aeruginosa, K. pneumoniae: >20 mg/mL
[78]
AntifungalC. albicansnot given100 mg/mL (stock solution) R. ulmifolius
extract; SH10: 9.6 ± 0.1 mg/g
  • MIC: 5 mg/mL
AntiviralNA from
C. perfringens
30 min
in situ assay
SH4 solution
  • Inhibitory activity of SH4 on NA from Clostridium perfringens: IC50 (μmol/L): 17.48 ± 2.9
[20]
spike glycoprotein of SARS-CoV-2
virus
in silico
molecular docking assay
SH6 and SH2 molecular
structures
  • SH6: docking score of—9.8 kcal/mol
  • SH2: docking score of—8.7 kcal/mol
[79]
Mpro protease and
spike glycoprotein of SARS-CoV-2
virus
in silico
molecular docking assay
SH6
molecular
structure
  • Mpro protease docking score: −10.3 kcal/mol
  • Spike glycoprotein docking score: −9.8 kcal/mol
[80]
EstrogenicMCF-7
human breast
adenocarcinoma cell
144 h
in vitro
proliferation assay
SH6 at 0, 25, 50 and 100 μM, Rubus coreanus: 0, 5, 10, 25, 50, and 100 μg/mL
  • SH6: 127.41% ± 0.26% cell proliferation at 100 μM;
  • R. coreanus: 574.57% ± 8.56% cell proliferation at 100 μg/mL
[81]
Estrogen
Receptor α
in silico
molecular docking assay
SH6
molecular
structure
  • SH6: docking score of—250.149 kcal/mol
NeuroprotectiveSK-N-MC
neuroblastoma cells
2 and 24 h
in vitro assay
commercial blackberry and R. vagabundus:
0, 0.25, 0.5,
and 1 µg GAE/mL,
R. brigantinus:
0, 0.1, 0.2, and 0.4 µg GAE/mL
  • All blackberry digested extracts at 2 and 24 h preincubation reduced basal ROS production. Under oxidative stress conditions, blackberry extracts did not reduce ROS production above 20%
  • The best activity (20%) exhibited R. brigantinus extract with a concentration of 0.4 µg GAE/mL)
[62]
24 h
in vitro assay
Commercial blackberry and R. vagabundus:
0, 0.25, 0.5,
and 1 µg GAE/mL,
R. brigantinus:
0, 0.1, 0.2,
and 0.4 µg GAE/mL
  • R. brigantinus and R. vagabundus extracts simultaneously increased mitochondrial transmembrane potential and cell membrane integrity
  • Preincubation with the IN fractions from R. brigantinus and R. vagabundus, although not changing GSH/GSSG ratio, increased GSH levels
AnticancerHeLa cells72 h
in vitro assay
Cytotoxicity:
0–25 µM of
SH6 DNA
cleavage: 10, 15, and 25 µM
  • Growth inhibitory effects of SH2 against HeLa cells occurred over a narrow dose range, with an ED50 of 12 µM
  • SH6 interfered with drug-stimulated DNA break formation in a dose-dependent fashion. This effect was quite similar against both DNA topoisomerases with IC50 values of ~15 µM
[82]
Topoisomerase
I and II
30 min
in situ assay
Topoisomerase I: 0, 19, 38, and 75 nM of SH6
Topoisomerase II: 0, 0.05, 0.1 0.2, 0.4, and 0.8 µM of SH6
  • SH6 interfered with topoisomerase I-mediated DNA cleavage: IC50 value = 0.02 µM
  • Topoisomerase Il-dependent DNA cleavage of linear DNA induced by the inhibitor VP-16 was prevented by simultaneous exposure to SH6. IC50 value = 0.l6 µM
Topoisomerase
I and II
30 min
in situ assay
0, 0.1, 0.2, 0.4, 0.6, 1.2, and 2.4 µM of SH6
  • Reaction of topoisomerase I-dependent DNA relaxation with IC50 value = 1 µM
  • Topoisomerase II was completely inhibited at 0.5 µM of SH6. IC50 = 0.01 µM
  • Relative potency of SH6 was 100-fold greater for topoisomerase II than for I
HUVECs and HT1080 cells72 h in vitro XTT
incorporation assay
SH6: concentrations up to
20 µg/mL
  • SH6 efficiently blocked the VEGF-induced HUVEC proliferation in a dose-dependent manner (IC50 = 7.4 µg/mL)
[83]
PRMI-7951 melanoma cellsin vitro
cytotoxicity
assay
SH2, SH6, and SH11 solutions
  • ED50 against melanoma RPMI-795 l:
    • SH2: 0.44 µg/mL
    • SH6: 5.00 µg/mL
    • SH11: 0.50 µg/mL
[68]
HL-60 and PBMCs12 h in vitro treatmentHL-60: 100 µM,
PBMCs:
400 µM of SH4
  • Inhibition of cell growth: cell values:
    • 93.0% ± 0.42% (HL-60)
    • 45.6% ± 0.30% (PBMCs)
[24]
AGS, HeLa,
Hep G2,
HT 29, and T 24
cell lines
24 h in vitro treatment100 µM of SH4
  • Inhibition of cell growth: cell values
    • 2.69% ± 2.44% (AGS)
    • 24.34% ± 4.73% (HeLa)
    • 38.99% ± 2.19% (Hep G2)
    • 8.10% ± 6.37% (HT 29)
    • 80.58% ± 5.98% (T 24)
HL-60 cells12 h in vitro
assay
serial dilution concentrations from 0 to 400 µM of SH4
  • Cytotoxic effect of SH4 was more pronounced in the leukemia HL-60 cells than in the normal PBMCs
25, 50, and 100 µM of SH4
  • SH4 showed significantly inhibited DNA fragmentation in a dose-dependent manner
100 µM of SH4
  • Treatment with SH4 showed a decrease in the 116 kDa PARP and a dose-dependent increase in inactive PARP
50 and 100 µM of SH4
  • SH4 showed a significant activation of caspase-3 in HL-60 in dose-dependent manner
A549 lung cancer cells48 h in vitro
assay
5 and 10 µM
of SH6
  • SH6 blocked the migration and invasion capabilities of the A549 cells during TGF-β1 induction of the EMT
[84]
48 h in vitro
assay
5 and 10 µM
of SH6
  • Significant decreases in the expression levels of nine genes
2 h in vitro
pretreatment
5 and 10 µM
of SH6
  • Snail expression was decreased by SH6 treatment in a dose-dependent manner.
  • Plasminogen activator inhibitor type-1 (PAI-1) expression decreased after SH6 treatment in a dose-dependent manner
5 and 10 µM
of SH6
  • SH6 antagonizes the phosphorylation of Smad2 and Smad3
5 and 10 µM
of SH6
  • TGF-β1 induction of the mesenchymal phenotype was inhibited
48 h in vitro
assay
1, 2.5, 5, 10, 25, 50, 75, and 100 µM of SH6
  • Concentrations of SH6 ≤ 25 µM did not affect the proliferation of A549 cells.
  • Proliferation of A549 cells was inhibited with ≥ 50 µM
MCF-7/Adr and MCF-7/wt cells 48 h in vitro
incubation; MTT assay
10, 20, 40, 79, 157, and 313
µM of SH6
  • SH6 inhibited the viability of MCF-7/Adr cell line within the whole concentration range. (EC50 = 38 µM).
  • SH6 caused fluctuations around the 100% control viability of MCF-7/wt cells
[85]
MDA-MB-231 human breast cancer cells24 h in vitro
assay
0 and 6.25 µM
of SH6
  • SH6 decreased the protein expression of VEGF, phosphorylated Akt, and ERK1/2
0, 6.25, 12.5, 25, 50, 100, and 200
µM of SH6
  • Treatment with up to 25 µM had no effect on MDA-MB-231 cells.
  • Treatment with 200 µM decreased cell viability
HUVECs0 and 6.25 µM
of SH6
  • The percentage inhibition of migration of 6.25 µM SH6-treated cells was 37.6% of that observed in the control group.
  • SH6 at a concentration of 6.25 µM significantly blocked tube formation (41.5% of control)
[86]
0, 6.25, 12.5, 25, 50, 100, and
200 µM of SH6
  • 12.5 µM with no effect on the HUVECs.
  • Treatment with 25 to 200 µM decreased cell viability
MCF-7 and MDA-MB-231 cells24 h in vitro
assay
0, 50, and 100 µM of SH6 for MCF-7 and MDA-MB-231 cells
  • SH6 increased Bax expression in MCF-7 cells
  • SH6 decreased Bcl-2 expression in MDA-MB-231 cells
[79]
50 and 100 µM of SH6
  • SH6 increased the cleavage of caspase-8, caspase-3, and PARP, but not that of caspase-9 in MCF-7 cells.
  • SH6 increased the cleavage of caspase-8, caspase-9, and caspase-3, as well as that of PARP in MDA-MB-231 cells
0, 5, 10, 25, 50, and 100 µM of SH6
  • SH6 at a concentration of 100 µM for MCF-7 and MDA-MB-231 significantly reduced viabilities to approximately 69% and 63%, respectively.
  • SH6 reduced the viabilities of both cell lines in a concentration-dependent manner
A2780 human ovarian carcinoma cells24 h in vitro
assay
0, 10, 20, and
40 µM of SH6
  • Increasing amount of: cleaved caspase-8, cleaved caspase-3, tBID cleaved RARP, and p-p38 with increasing SH6 dose
[87]
20 and 40 µM
of SH6
  • Treatment of A2780 cells with SH6 induced an increase in the fraction (Annexin V+/PI-) of early apoptotic cells from 4.17% to 41.76%
0, 10, 20, and
40 µM of SH6
  • Treatment of A2780 cells with SH6 induced a decrease in cell viability in a dose-dependent manner
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gesek, J.; Jakimiuk, K.; Atanasov, A.G.; Tomczyk, M. Sanguiins—Promising Molecules with Broad Biological Potential. Int. J. Mol. Sci. 2021, 22, 12972. https://doi.org/10.3390/ijms222312972

AMA Style

Gesek J, Jakimiuk K, Atanasov AG, Tomczyk M. Sanguiins—Promising Molecules with Broad Biological Potential. International Journal of Molecular Sciences. 2021; 22(23):12972. https://doi.org/10.3390/ijms222312972

Chicago/Turabian Style

Gesek, Jakub, Katarzyna Jakimiuk, Atanas G. Atanasov, and Michał Tomczyk. 2021. "Sanguiins—Promising Molecules with Broad Biological Potential" International Journal of Molecular Sciences 22, no. 23: 12972. https://doi.org/10.3390/ijms222312972

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