ISOORIETIN

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 23

2582

Send Orders for Reprints to reprints@benthamscience.net


Current Pharmaceutical Design, 2021, 27, 2582-2604
REVIEW ARTICLE
ISSN: 1381-6128
eISSN: 1873-4286

Extracts and Flavonoids of Passiflora Species as Promising Anti-inflammatory and


Antioxidant Substances
Impact
Factor:
3.116

Marcin Ożarowski1, and Tomasz M. Karpiński2,*

1
Department of Biotechnology, Institute of Natural Fibres and Medicinal Plants, Poznań, Poland; 2Chair and Department of Medical
Microbiology, Faculty of Medical Sciences, Poznań University of Medical Sciences, Poznań, Poland

Abstract: There is increasing interest in evaluating anti-inflammatory activities of plant substances such as ex-
tracts and flavonoid rich fractions. A promising source of new medicinal drugs may be species from the Passi-
floraceae family. The most interesting group of principal chemical substances in Passiflora species are polyphe-
nolic compounds, including flavonoids (quercetin, rutin, apigenin, luteolin, chrysin, and C-glycosylflavones i.e.,
vitexin, isovitexin, orientin, isoorientin), due to their antioxidant activity demonstrated in various studies. How-
ARTICLE HISTORY ever, each extract from Passiflora spp. as multi-component mixtures should be estimated for chemical composi-
tion (in the standardization process) and its activity using in vitro and in vivo tests. The current standard for drug
Received: February 28, 2020 discovery and development from plants indicates that only collective assessment allows estimating plant sub-
Accepted: April 19, 2020
stances by definition of the origin of raw materials and their quality, methods of extractions, and metabolite pro-
files. Increasingly, due to complex phytochemical procedures to obtain extracts, individual flavonoid compounds
DOI:
are also tested for anti-inflammatory action. However, it should be emphasized that various sources of potential
10.2174/1381612826666200526150113
new drugs from plant origin are not mutually exclusive, but are complementary. A review of bibliographic data
includes the following information about Passiflora species, such as distribution, classification, phytochemical
compounds, the anti-inflammatory activity of the extracts, the anti-inflammatory activity of flavonoids, and anti-
Current Pharmaceutical Design

oxidant potential. The review concluded that extracts and flavonoids (mainly quercetin, apigenin, and vitexin)
from Passiflora spp. can be a valuable source of anti-inflammatory and anti-oxidative medications for the preven-
tion and treatment of many diseases, which occur with complex inflammatory processes.
Keywords: Passifloraceae, Passiflora, flavonoids, anti-inflammatory, antioxidant, chrysin.

The effectiveness of available anti-inflammatory therapy using


1. INTRODUCTION
nonsteroidal anti-inflammatory drugs (NSAIDs) is still not satisfac-
Inflammatory processes have a multifactorial course, including tory. Moreover, this group of medicinal drugs belongs to the most
chain reactions in various pathways at the subcellular, cellular, and widely prescribed medicines. However, there are many adverse
tissue level with various signaling molecules and immune cells. effects observed in patients after treatment of frequent inflamma-
However, it should be noted that inflammation is a physiological, tory diseases i.e., serious cardiovascular events, gastrointestinal
biological defense process against infection and tissue damage [1]. complications (mucosal injury, ulcers, bleeding), hypertension, and
It is well known that these processes are adapted by inflammatory renal disturbances [8, 9]. This situation may be more complicated
mediators such as cytokines: interleukins, tumor necrosis factor-α, because mild-to-moderate inflammatory diseases have a long time
prostaglandins, leukotrienes released by macrophages and leukocytes, of duration, and very often their symptoms return after remission
reactive oxygens species; moreover, during pathophysiological proc- like in ulcerative colitis [10], rheumatoid arthritis [11], multiple
esses of inflammatory diseases, they may occur with antioxidative sclerosis [12]. For this reason, patients have to use NSAIDs for a
stress [2]. long time. Additionally, many therapies have not shown therapeutic
In the 21st century, there is a need for novel, innovative medici- benefits for a variety of reasons [13, 14]. Thus, there is an increas-
nal products, not only against diseases associated with inflamma- ing demand for new drugs with no side effects, showing better effi-
tory processes for which there are generally no effective medica- cacy.
tions, e.g., inflammatory bowel disease (IBD, including ulcerative In recent years, interdisciplinary research has focused on many
colitis and Crohn’s disease), cancers, rheumatoid arthritis, asthma, levels to develop more effective and safer therapeutics. It is also
diabetes, psoriasis, hypertension, neuropathic pain, neurodegenera- happening in the development of more favorable therapies for in-
tive disorders (including Alzheimer’s and Parkinson’s diseases), flammatory diseases, including the search for new medicinal drugs
neuroinflammatory diseases like sclerosis multiplex [1, 3-7], but from natural sources, plants, and their extracts containing promising
also for diseases that may occur in the future. It should be noted chemical compounds, i.e. flavonoids which possess anti-
that inflammatory diseases are increasingly prevalent in various inflammatory activity [2, 15-17]. Passiflora spp. can have a great
populations [1]. value in this aspect.

2. PASSIFLORA SPECIES: DISTRIBUTION, CLASSIFICA-


*Address correspondence to this author at the Chair and Department of TION, FRUITS AND EXTRACTS MARKET
Medical Microbiology, Faculty of Medical Sciences, Poznań University of Ornamental and medicinal plants from the family Passi-
Medical Sciences, Wieniawskiego 3, 61-712 Poznań, Poland;
Tel: +48-61-854-61-38; E-mail: tkarpin@ump.edu.pl
floraceae Juss. ex DC. grow in tropical, subtropical, and temperate

1873-4286/21 $65.00+.00 © 2021 Bentham Science Publishers


Extracts and Flavonoids of Passiflora Species as Promising Anti-inflammatory Current Pharmaceutical Design, 2021, Vol. 27, No. 22 2583

regions, mainly in South America (Brazil, Colombia, Peru, Bolivia, for their therapeutic application, extracts should always be stan-
etc.) [18, 19]. Approximately 96% of the species are distributed in dardized to a defined content of a constituent or a group of sub-
the Americas (mainly in Brazil and Colombia). However, there are stances with known therapeutic activity during the manufacturing
records of species in India, China, Southeastern Asia, Australia, the processes and quality control [35]. Such standardized (phytochemi-
Pacific islands, and neighboring regions [19]. Few species are dis- cally characterized) herbal extracts and their therapeutical applica-
tributed in Europe i.e. P. caerulea (the blue passionflower) now tions are the basis of rational phytotherapy.
grows wildly in Spain [20] and is cultivated in gardens of Poland Species from the Passifloraceae family deliver two kinds of
[21]. valuable raw materials: nutritious fruits and aerial parts containing
Currently, species of the family Passifloraceae Juss. ex Roussel leaves and flowers, which have active chemical compounds. It
sensu stricto, belong to 36 plant genera, and the Plant List includes needs to be highlighted that today, not only fruits but also leaves of
932 accepted species names [22]. Passiflora L. is the most repre- Passiflora are accepted as a component of a healthy diet used in the
sentative genus in the Passifloraceae family, containing more than prevention or reduction of symptoms of many diseases. Studies
500 species [23]. It should be noted that new species from the Pas- showed that fruits of P. edulis contain flavonoids (quercetin and
sifloraceae family are still being discovered. Additionally, more derivatives, rutin, kaempferol, chrysin, apigenin, luteolin), C-
than 330 hybrids have been described [24]. In North America and glycosylflavones (orientin, isoorientin, vitexin, schaftoside,
Europe, the most well-known species with a long tradition of me- isoschaftoside) [39, 40], carotenoids and vitamin C [37]. Moreover,
dicinal use worldwide is Passiflora incarnata L. (the purple pas- the fresh pulp of the fruits of P. subpeltata contains epicatechins,
sionflower), described in the European Medicines Agency, the carbohydrates, and proteins [41]. Whereas, the leaves of cultivated
European Pharmacopoeia, and the British Herbal Pharmacopoeia species of Passiflora are renewable, and this green biomass can be
[25]. In contrast, in Brazil and Colombia - P. alata Curtis (the fra- further used as a bioactive food additive and as a source of promis-
grant grenadilla) and P. edulis Sims (the purple and yellow passion- ing anti-inflammatory pharmaceuticals.Many studies showed vari-
fruit or maracujá) are official species in the Brazilian Pharmaco- ous flavonoids (flavon-C-, and C,O-, and O-glycosides) present in
poeia [26]. the extracts from the leaves of Passiflora [42-51].
Numerous Passiflora species, in addition to phytotherapeutic Recently, phytochemical comparison of alcoholic extracts from
use, are characterized by the growing agronomic significance not leaves of three species of Passiflora (P. alata, P. caerulea, P. in-
only in Brazil and Colombia but also in India, as they are cultivated carnata) revealed that flavonoids such as O,C-glycosides of api-
plants, being the source of nutritious fruits, hence they are of great genin, luteolin and chrysin dominated in these species (>60% for P.
importance in the global fruit industry (m.in. P. alata Curtis, P. incarnata, 50% for P. caerulea, 40% for P. alata). C-glycosides
cincinnata Mast., P. edulis Sims, P. quadrangularisL., P. ligularis have been observed in a high level in P. caerulea (>30%) and P.
A. Juss., P. maliformis L., P. nitida Kunth, P. setacea D.C.) [27, incarnata (>20%) [52]. Moreover, other studies phytochemically
28]. Previously, the world’s production of passion fruits showed an examined extracts from fruit pulps, from fruit peels (flavedo and
annual production that exceeds 900,000 tons in Brazil [29]. Addi- albedo), leaves, bark, and seeds [53, 54].
tionally, according to the market research report, the global passion- It has been concluded that quercetin (3,3’,4’,5,7-pentahy-
flower extracts market value was estimated to be USD 2.55 billion droxyflavone) (Fig. 1) is present in some extracts of Passiflora i.e.,
in 2018. The global passionflower extracts market includes phar- from the leaves of Passiflora subpeltata [48]. Moreover, glycosides
maceutical, nutraceutical, personal care, food, and beverage sectors of quercetin were found in the extracts of P. caerulea and P. alata
[30]. [49], P. leschenaultii [51], P. edulis var. flavicarpa [55], and P.
subpeltata [41]. Rutin (3,3′,4′,5,7-pentahydroxyflavone-3-
3. PASSIFLORA EXTRACTS AND THEIR PHYTOCHEMI-
rhamnoglucoside;rutoside) was detected in the extract of Passiflora
CAL COMPOUNDS
leschenaultii [96], P. subpeltata [48], P. incarnata [56]. Chrysin
Currently, there is an intense scientific interest in drug discov- (5,7-dihydroxyflavone) and its glycosides are found in extracts i.e.,
ery and development based on herbal medicines. At the same time, Passiflora caerulea and P. incarnata [39,49, 57, 58]. Luteolin (3',
one should not forget that therapies of complementary and alterna- 4', 5,7-tetrahydroxyflavone) and its C-glycosides belong to the
tive medicine are becoming increasingly popular around the world group of flavonoids present in the extracts of P. incarnata [49, 59,
[31, 32]. These two tendencies are not mutually exclusive, and they 60], P. edulis [47,61], P. alata and P. caerulea [49]. Few studies
indicate that a detailed study of plant sources is comprehensive and revealed their anti-inflammatory activity in various tests [62-67].
complementary in the searching for a new therapeutic solution,
Apigenin had been isolated from the aerial parts of P. incar-
including diseases with inflammatory processes. According to
nata [68] and from the pericarp of P. quadrangularis [69]. How-
Atanasov et al. [32], developing trends indicate that natural prod-
ever, different glycosides of apigenin like vitexin and isovitexin
ucts will be among the most important sources of new therapeutic
derivatives are more often detected in various Passiflora species
drugs in the future.
[38, 39,49]. Vitexin, a well-known flavonoid, is present in P.
One of the most popular forms of plant substances is the ex- foetida [70], P. caerulea [71, 72], P. edulis var. flavicarpa, P.
tracts that can be an interesting source of pharmacologically active manicata, and P. tripartita var. mollissima [46, 54]. However, gly-
compounds for future applications. It has been known for a long cosides of vitexin were detected in P. alata [73], P. quadrangularis,
time that crude and fractionated plant extracts are a promising com- P. manicata [46], P. caerulea, and P. incarnata [49]. Other C-
plex composition of primary and secondary metabolites, showing glycosylflavonoids, such as isovitexin, orientin, and isoorientin
activity through synergy in their action [32, 33]. The herbal drug are characteristic chemical compounds detected in extracts from
preparations, mainly extracts, differ in chemical compositions, be- many species of Passiflora (i.e. P. edulis) [49, 54, 72, 74-77]. Be-
cause they are obtained from various parts of plants growing in sides this, Ożarowski et al., [49] revealed that extracts from leaves
different climate and ecological conditions. It was observed that the of Passiflora species contain compounds with terpenoid structures:
populations of the same Passiflora spp. growing in different geo- blumenols B and C (megastigmanes), and cyclopassifloic acid glu-
graphical areas would have different profiles in their chemical coside, the activity of which is poorly studied. On the other side, it
compounds [34]. Furthermore, plant extracts are obtained using is well known, that flavonoids and phenolic acids occurring in vari-
various processes such as extraction, fractionation, distillation, ous extracts from Passiflora are taken into account as novel thera-
fermentation, hydrolysis using different reagents, and many variants peutic agents in the prevention and treatment of Alzheimer’s dis-
of technological equipment and parameters (i.e. temperature and ease, disorders associated with inflammation [16], liver injury with
time of extraction) in standard conditions [35-38]. For this reason,
2584 Current Pharmaceutical Design, 2021, Vol. 27, No. 22 Ożarowski and Karpiński

Quercetin

Rutin

Chrysin Luteolin Apigenin

Isovitexin
Vitexin

Isoorientin
Orientin
Fig. (1). Chemical structure of the main flavonoids of Passiflora spp.

inflammatory processes [67], colitis [78], or bacterial infections results also demonstrated that not only leaf extracts possess activi-
[79]. ties but also peels of Passiflora edulis, which improve insulin sensi-
tivity [100] and have shown intestinal anti-inflammatory effects in
4. RECENT PROFILE OF ACTIVITIES - IN VITRO, IN VIVO animal models of colitis [53, 54, 97], with antioxidant activity [89].
AND CLINICAL TRIALS OF PASSIFLORA EXTRACT Moreover, the bark of the same species reduced dyslipidemia [61].
Biological and pharmacological activities of preparations from Few randomized-controlled clinical trials confirmed some pharma-
Passiflora species depend on the presence of phenolic compounds, cological activities of these plant substances [18], i.e. P. incarnata
mainly flavonoids. In the past years, several preclinical experiments showed anxiolytic effect [101-103], improved sleep efficiency and
demonstrated that alcoholic extracts of aerial parts of various spe- quality [104, 105], relieved nervous restlessness [106], exerted anti-
cies of Passiflora, including flowers and leaves, have many activi- hypertensive properties [107] and alleviated osteoarthritis symp-
ties such as sedative, anxiolytic, antidepressant [80], memory- toms (pain) [108]. Furthermore, peel of Passiflora edulis f. flavi-
enhancing activity [81], anti-diarrhoeal, spasmolytic [72], antidia- carpa decreased insulin resistance in patients with diabetes type 2
betic [82, 83], and anti-allodynic effects in neuropathic pain [84], [109].
with antinociceptive [85], anti-leukemic [49], hepato-, nephro-, Today, extracts of Passifloraspecies (mainly P. incarnata, and
gastroprotective [86, 87], neuroprotective effects [88, 89], = antimi- P. edulis) are popularly used for the prevention and treatment of
crobial [90] and antioxidative activity shown in various in vitro and various central nervous system diseases such as anxiety, insomnia,
in vivo tests [20, 40, 41, 50, 73, 75, 82, 91-96]. Nowadays, there is and depression [18, 98]. However, a large number of patients from
growing scientific evidence that the extracts of Passiflora have anti- America use products containing extracts of Passifloraspecies for
inflammatory properties [45,48, 54, 55, 72, 85, 95, 97-99]. Recent the treatment of wounds, boils, earaches, and liver diseases. In con-
Extracts and Flavonoids of Passiflora Species as Promising Anti-inflammatory Current Pharmaceutical Design, 2021, Vol. 27, No. 22 2585

trast, in Europe, these products are preferred in order to provide that abnormal composition in the gut microbiota can be associated
relieve from agitation and restlessness. According to prognosis for with chronic inflammatory bowel disease. In this context, Jang et
the development of the market for herbal medicinal products, such al., [118] observed in mice the simultaneous amelioration of colitis
applications are anticipated to create growth opportunities for the by Bifidobacterium longum and Lactobacillus plantarum, and these
extracts of Passiflora species in the near future [110]. strains inhibited the activation of NF-κB pathway. They may also
It needs to be highlighted, that mechanisms of action of natural diminish the growth of Escherichia coli.
chemical compounds, including antioxidant and anti-inflammatory Using a mouse model of experimental colitis induced by the
pathways might create a new therapeutic window for many dis- dextran sodium sulfate, the authors further examined the prebiotic
eases, i.e. colitis [53, 54, 111], hypertension [107], osteoarthritis effects of the flour from peels of P. edulis fruits, which was given
[108], neuropathic pain [84, 85] or Alzheimer’s disease [16, 112- to the female mice (8 mg/mL in the drinking water) [54]. Results of
115], occurring with pathogenetic processes such as inflammation this study revealed that dietary fibers and polyphenols containing
and oxidative stress. C-glycosyl flavonoids (isoorientin > vicenin > orientin > vitexin >
isovitexin, lucenin-2, schaftoside, violanthin) downregulated the
5. ANTI-INFLAMMATORY ACTIVITY OF PASSIFLORA pro-inflammatory cytokines IL-1β, IL-6, and IL-17. However, peels
SPECIES of P. edulis did not affect the TNF-α and IL-12 contrary to previous
Several studies have shown that various species of Passiflora studies [97]. Moreover, a biochemical analysis revealed that the
containing phenolic compounds revealed anti-inflammatory proper- peel’s intake decreased the expression of chemokine MCP-1, the
ties during in vitro and in vivo tests, such as in non-obese diabetic adhesion molecule ICAM-1, and increased the expression of the
mice, models of mouse colitis, pleurisy, air pouch inflammation, mucin protein MUC-2 and MUC-3, thus leading to improvement of
and paw edema. Many species of Passiflora are a source of juice the epithelial barrier before bacteria, microbial products, and toxins
and pulp containing fiber and polyphenols, including flavonoids, during the inflammatory process. Another study also exerted that
however, a significant amount of waste is generated [54]. Recent polyphenolic compounds (i.e. naringenin, naringin) in the diet may
extensive studies have reported several novel aspects of extracts improve the integrity and stabilization of the epithelial barrier [119,
from fruit pulps, and also from fruit peels (flavedo and albedo), 120]. This observation is innovative because there is no cure and
leaves, bark, and seeds, which are studied for their healthy proper- therapies owhich would decrease the inflammatory process and
ties. It was observed that single flavonoids also demonstrated this restore the physiological function of the intestinal barrier [120].
effect very often together with antioxidant activity. State of the art Furthermore, Cazarin et al., [54] observed that peels of P. edulis
in this field shows the potential beneficial effects of these by- decreased the expression of these metalloproteinases (MMP-2 and
products of the fruit industry in many diseases occurring with in- MMP-9), which are associated with various physiological proc-
flammatory processes. esses.
Other studies have reported that not only P. edulis is an interest-
5.1. Studies of Anti-Inflammatory Activity of Complex Extracts ing species in this field, but also others such as P. caerulea, which
5.1.1. The Action of Extracts During the Inflammatory Process in is a very representative medicinal plant in Brazil. The ethanolic
Colitis extract of P. caerulea with identified flavonoids isoorientin, vitexin,
isovitexin, and vicenin-2, was also examined in the experimental
Many clinical studies described pathological processes during rats model of colitis induced by acetic acid [72]. Results showed
inflammatory bowel diseases (IBD), including ulcerative colitis and that the extract at a dose of 250 mg/kg, p.o. reduced macroscopic
Crohn’s disease, which are chronic and multifactorial disorders of and microscopic damages of the colon. Besides anti-inflammatory
the gastrointestinal tract and may lead to progression of colon can- properties of the extract of P. caerulea, there were also antidiar-
cer [116]. Furthermore, these diseases with uncontrolled inflamma- rhoeal and spasmolytic activities.
tion can be progressive and challenging to treat using conventional
therapy [117]. Today, several studies have been carried out to Recently, it was revealed that also acetone extract from other
search the new anti-inflammatory drugs, including flavonoid-rich species P. subpeltata, prevalent in Mexico, had anti-inflammatory
extracts from leaves and peels of fruits of many species of Passi- and protective activity on indomethacin-induced experimental ul-
flora, which were tested in various animal models of colitis. cerative colitis in rats [51]. This extract contained phenolic and
flavonoid compounds such as gallic acid, apigenin, catechin, luteo-
Anti-inflammatory properties of extracts from leaves and fruit lin, and quercetin 3-b-D glucoside. Indomethacin (10 mg/kg s.c.)
peels of Passiflora edulis were observed in a series of studies by was administered fortwo days and next, the extract was given to
Cazarin et al. [53, 54, 97]. The activity of aqueous extract of Passi- rats until 11 days. The biochemical study showed that extract at a
flora edulis leaves was investigated in colitis in rats induced by dose of 400 mg/kg possessed more effective activity in an experi-
intracolonic administration of 2,4,6-trinitrobenzene sulphonic acid. mental model. More detailed information from these studies is
The leaf extract containing significant amounts of isoorientin, ori- shown in Table 1.
entin, isovitexin, and vitexin, was administered to rats at a dose of
1100 µg mL−1 for 2 weeks. Biochemical analyses showed that the 5.1.2. The Activity of Extracts in the Model of Paw Edema and
extract reduced the level of pro-inflammatory mediators in intesti- Air Pouch Inflammation
nal tissues such as fivefold reduction of IL-1βand two-fold for Currently, Saravanan Shanmugam et al. [95] showed that Passi-
TNF-α [97]. More detailed information taken from these studies is flora leschenaultii growing in India exerted anti-inflammatory ac-
shown in Table 1. tivity using carrageenan for the induction of paw edema, the cotton
In the subsequent study [53], in animals with colitis, which was pellet to a model of inflammation and pyrexia model induced by the
developed through the application of 2,4,6-trinitrobenzenesulphonic suspension of brewer’s yeast in rats. Phytochemical analysis of
acid, powdered peels of fruits of Passiflora edulis were tested. various extracts (petroleum ether, chloroform, methanol, acetone,
However, the macroscopic and microscopic evaluations of the in- hot water) from leaves revealed the presence of tannins, phenolics
flamed mucosa did not show attenuation of the damage of tissues in such as chlorogenic acid, and caffeic acid, and also flavonoids such
the model of colitis after the consumption of a diet with peels. On as quercetin-3-O-galactoside and rutin. The animals were pre-
the other site, peels of fruits of Passiflora edulis as a source of fla- treated with the extract 1 h before the application of 0.1 mL of 1%
vonoids and fibers, increased the count of Bifidobacterium and carrageenan. Assessment of anti-inflammatory properties of the
Lactobacillus, hence intake of peels exhibited an effect, modulating extracts using doses of 200 and 400 mg/kg b.w. per os, revealed
the microbiota in the intestinal area. Currently, it is well known
2586 Current Pharmaceutical Design, 2021, Vol. 27, No. 22 Ożarowski and Karpiński

Table 1. Anti-inflammatory activities of extracts of Passiflora spp.

Passiflora species / Dose and method of Effects on proinflammatory


Pharmacological activities against the pathological status Refs.
preparation administration cytokines

The activity of extracts during the inflammatory process in colitis

- improving the body weight, the weight of proximal


duodenum, distal jejunum, proximal ileum, whole cae-
cum, proximal colon
- reducing the percentage of the damaged area of the
gastrointestinal tract and reducing the number of - suppression of NO and
200 mg/kg
P. subpeltata / extract lesions, necrosis and mucosal infiltrations after a higher TNF-α production in LPS-
400 mg/kg dose of an extract [51]
from leaves induced RAW 264.7 cells,
per os - reduction in the level of neutrophil accumulation after 100 μg/mL of extract
- inhibiting the MPO enzyme levels after a higher dose
of the extract
- decreasing the level of lipid peroxides
- preservation of the enzymes SOD, CAT and GSH
levels

- reduces the expression of


- increasing the expression of the mucin protein MUC-2 chemokine - MCP-1
and MUC-3
8 mg/mL in the - decreasing the ICAM-1
drinking water - the protective effect in the colon mucosa
- reduction in the expres-
P. edulis / flour from (around 40 mg/mice) - decreasing the expression of MMP-2 and MMP-9 sion of IL-1β, Il-6, and IL- [54]
dried peels
per os - promoting the short-chain fatty acids production 17
- enhancing the intestinal protective barrier - no affect on the expression
- revealed prebiotic effects of the TNF-α and the IL-
12 in the colon

- improving the content of serum proteins, albumin, and


antioxidant status - the five-fold reduction in
1100 µg leaves mL−1 - enhancement of the GSH activity by 1.3- fold and the IL-1β level
P. edulis / extract for 2 weeks GR activity by 1.8-fold in the liver of the tea colitis - the two-fold decrease in
[97]
from leaves per os group compared to the control colitis group IL-6 and TNF-α levels in
- no differences in damage of the colon tissue in macro- the colon intestinal tissue
scopic and microscopic level between the tea colitis of the tea colitis group 
group and control colitis group

diets with 50% of the - protection of the colon tissue from lipid peroxidation
cellulose substituted - improving serum antioxidative status and trend to
with dietary fiber enhance the level of the antioxidant enzymes
from passion fruit - increasing the GR activity
P. edulis / peels flour (PFF) for 84 - no studied [53]
- rising the count of Bifidobacterium and Lactobacillus
days
after PFF used in the prevention protocol
per os
- no attenuation of the macroscopic and microscopic
tissue damage in animals with colitis 

- reduction in macroscopic and microscopic damages of


colon
P. caerulea / extract 125 mg/kg
- decreasing the MPO level in the colon
from leaves 250 mg/kg
- reduction in the lipid peroxidation - no studied [72]
per os
- reduction in the severity of tissue damage
- antidiarrhoeal (in vivo) and spasmolytic (in vitro)
activities
(Table 1) Contd.....
Extracts and Flavonoids of Passiflora Species as Promising Anti-inflammatory Current Pharmaceutical Design, 2021, Vol. 27, No. 22 2587

Passiflora species / Dose and method of Effects on proinflammatory


Pharmacological activities against the pathological status Refs.
preparation administration cytokines

The activity of extracts in the model of paw edema and air pouch inflammation

- inhibition of the levels of


cytokine-like MIP-2 by
78.0% after aqueous re-
- decreasing the migration of leucocytes by 48% after P. sidual fraction at a dose of
edulis aqueous lyophilized extract at a dose of 100 100 mg/kg (most active)
P. edulis f. flavicarpa 50 mg/kg mg/kg (most active)
- repression in the levels of
/ extract from leaves
100 mg/kg - decreasing the neutrophils by 58.2% after butanolic IL-1β by 74.0% after
and fractions of the [42]
per os fraction at a dose of 50 mg/kg (most active) aqueous residual fraction
extract
- inhibiting the MPO level by 48.2% after butanolic at a dose of 100 mg/kg
fraction at a dose of 50 mg/kg (most active) (most active)
- inhibiting the NO level by
61.7% after aqueous re-
sidual fraction at a dose of
100 mg/kg (most active)

- 200 mg/kg and 400 mg/kg showed 42.07% and 81.37%


inhibition of edema induced by carrageenan, respecti-
vely
P. leschenaultii / 200 mg/kg - 56% and 79.04% granuloma inhibition after 200 mg/kg
extract from leaves 400 mg/kg and 400 mg/kg of an extract, respectively
- no studied [95]
per os - antipyretic activity - decreasing the rectal temperature
after 200 mg/kg and 400 mg/kg of extract by 1.05 and
2.42°C, respectively (after 5h)
- analgesic effect (in additional tests)
- antioxidant activity (in vitro)

- 200 mg/kg and 400 mg/kg presented 47.08% and


81.54% inhibition of edema induced by carrageenan,
P. subpeltata / extract 200 mg/kg respectively
from leaves 400 mg/kg - antipyretic effects - decreasing the rectal temperature
- no studied [48]
per os after 200 mg/kg and 400 mg/kg of extract by 1.68 and
2.55°C, respectively (after 5h)
- analgesic effect (in additional tests)
- antioxidant activity (in vitro)

- 200 mg/kg presented inhibition of edema induced by


P. foetida / extract 100 mg/kg
carrageenan
from leaves 200 mg/kg - no studied [45]
- anti-inflammatory effects against histamine
per os
- analgesic effect (in additional tests)

- reduction in the carrageenan-induced hind paw edema


in mice after all doses

100 mg/kg - reduction in edema induced by histamine after 400


mg/kg dose of the extract
P. cincinnata / extract 200 mg/kg
- reduction in total leukocytes and reduction in neutro- - inhibiting the production [85]
from leaves and stems 400 mg/kg
phil migration after treatment with the 100 and 400 of NO
per os mg/kg doses of the extract
- reduction in the number of mononuclear cells after 200
mg/kg dose of an extract
- analgesic effect (in additional tests)
(Table 1) Contd.....
2588 Current Pharmaceutical Design, 2021, Vol. 27, No. 22 Ożarowski and Karpiński

Passiflora species / Dose and method of Effects on proinflammatory


Pharmacological activities against the pathological status Refs.
preparation administration cytokines

The activity of extracts during the inflammatory process in diabetes

- reduction in the numbers of inflammatory cells in the


pancreatic islets:
- reduction in the proliferation of T lymphocytes at the
doses of 500, 800 and 1000 μg/mL of extract
P. alata / extract from
- decreasing the numbers of CD4+ T and CD8+ T cells
leaves extract in chow diet in the pancreas islets - no studied [126]
(15 g leaves/L water) - reducing the infiltration of CD4+ T and CD8+ T cells
in the pancreatic islets
- decrease of 28.6% in diabetes incidence
- protection of the beta cells
- antioxidant activity: decreasing the lipid peroxidation

- increasing the reduced glutathione concentration


- decreasing numbers of CD4+, CD8+ and CD11c+ cells
P. alata / extract from extract in chow diet
in the pancreas islets
leaves (15 g leaves/L water) - no studied [73]
- reducing the level of insulitis
- antioxidant activity
- decreasing of injury of pancreatic islets

The activity of extracts during inflammation of the pleurae induced by carrageenan

- diminishing the level of


- inhibition of the activity of the MPO (64% of inhibi-
TNF-α (79% of inhibition)
tion) using 250 mg/kg dose of an extract
and IL-1β (59% of inhibi-
- reduction in the leukocyte influx (67% of inhibition) tion) in pleurisy induced
P. edulis var. flavi- 250-500 mg/kg using 250 mg/kg dose of an extract by carrageenan using 250
carpa / extract from mg/kg dose of the extract [43]
intraperitoneal route - decreasing the neutrophils (85% of inhibition) using
leaves
250 mg/kg dose of an extract - inhibiting the level of
- inhibition of the leukocyte migration nitric oxide (47% of inhi-
bition) using 250 mg/kg
dose of the extract

P. alata P. alata
- decreasing the leukocyte migration from ca. 29 to 65% - reducing the C-reactive
- inhibition in the neutrophil influx from 27 to 96% protein level by 64% using
100 mg/kg dose of an
- inhibition of myeloperoxidase by 61% using 100 mg/kg extract
100 – 300 mg/kg of dose
P. alata / extract from P. alata extract P. edulis
leaves - decreasing the adenosine deaminase activity by 57%
intraperitoneal route using 100 mg/kg dose - decreasing the C-reactive
P. edulis / extract protein level by 66% using [44]
from leaves 100 - 1000 mg/kg of P. edulis 250 mg/kg dose of an
P. edulis extract - decreasing the leukocyte migration from 19 to 75% extract
intraperitoneal route - inhibition of the neutrophil influx from 24 to 78%
- inhibition of myeloperoxidase by 49% using 250 mg/kg - no significant changes in
dose nitric oxide levels (P. ala-
- decreasing the adenosine deaminase activity by 77% ta, P. edulis)
using 250 mg/kg dose
Abbreviations (Tables 1 and 2): CAT - catalase, COX-2 - cyclooxygenase-2, GR - reduced glutathione, GSH - glutathione, GSH-Px - glutathione peroxidase,
GST - glutathione S-transferase, HO-1 - heme oxygenase-1, IL-1β, IL-6 – interleukins, iNOS - inducible nitric oxide synthase, ICAM-1 - intercellular adhesion
molecule-1, i.p. - intraperitoneal administration, 5-LOX - 5-lipoxygenase, LPS - lipopolysaccharide, LTB4 - leukotriene, MCP-1 - monocyte chemoattractant
protein-1, MDA - malondialdehyde, MMP-2 - matrix metalloproteinases-2, MMP-9 - matrix metalloproteinases-9, MPO - myeloperoxidase, NF-κB - nuclear
factor kappa-light-chain-enhancer of activated B cells, NO - nitric oxide, Nrf2 - nuclear factor erythroid 2-related factor 2, ox-LDL - oxidized low-density
lipoprotein, PGE2 - prostaglandin E2, PKC - protein kinase C, p.o. - per os administration, ROS - reactive oxygen species, SOD - superoxide dismutase, TGF-
β1- transforming growth factor-β1, TNF-α - tumor necrosis factor-α, TXB2 - thromboxane, VCAM-1 - vascular cell adhesion molecule 1.

that all plant substances showed similar activities in a dose- minished the formation of granuloma by 79% in comparison with
dependent manner. Still, the most active was the acetone extract at a indomethacin. This extract also showed antioxidant, antipyretic,
dose of 400 mg/kg, because it inhibited inflammation during the and analgesic activities. It was discussed that the extract with fla-
late phase (5 h) by 81% in edema induced by carrageenan, and di- vonoids (i.e. quercetin-3-O-galactoside or hyperin) may lead to
Extracts and Flavonoids of Passiflora Species as Promising Anti-inflammatory Current Pharmaceutical Design, 2021, Vol. 27, No. 22 2589

anti-inflammatory action by decreasing the mediators of inflamma- to reduce the proliferation of T lymphocytes, which have been
tion through various pathways [121-125]. stimulated by concanavalin A, and decrease the numbers of CD4+
A similar study was performed earlier by Saravanan Shan- T and CD8+ T cells and their infiltration in the pancreatic islets.
mugam et al., [48] using the extract from leaves of Passiflora sub- Accordingly, these in vivo findings indicate that anti-inflammatory
peltata, which is used in folk medicine in India. The author used the properties exerted by P. alata extract containing phenolic com-
same test and doses of acetone extract (200 and 400 mg/kg b.w. per pounds protected beta cells. In the previous study of these authors
os) in rats. The extract contained flavonoids (apigenin, quercetin, [73], it was similarly observed that the same aqueous extract (con-
rutin), and tannin compounds (gallic acid and catechin). The rats taining vitexin, isovitexin, isoorientin) in diet after 28 weeks de-
were pre-treated with the extract 1 h before the administration of creased the injury of pancreatic islets, infiltration of inflammatory
0.1 ml of 1% carrageenan. Analysis of pharmacological results cells, and reduced the incidence of diabetes in non-obese diabetic
showed that this extract at doses of 200 and 400 mg/kg presented mice.
47.08% and 81.54% inhibition of edema after the administration of
5.1.4. The Activity of Extracts During Inflammation of the Pleu-
carrageenan. Moreover, the extract exhibited antipyretic effect,
rae Induced by Carrageenan
analgesic activity, and the antioxidant potential in vitro, similarly as
acetone extract of Passiflora leschenaultii [95]. The anti-inflammatory effects of aqueous extract from leaves of
Passiflora edulis var. flavicarpa with a high flavonoid content
Previously, anti-inflammatory properties have also been inves-
(4.4%)at doses 250-500 mg/kg i.p. were previously observed by
tigated for ethanol extract of Passiflora foetida leaves in a model
Montanher et al., [43]. This study demonstrated several mecha-
using 0.1 ml of 1% carrageenan and histamine (in 1% CMC w/v)
nisms of anti-inflammatory effects of extract in the mouse model of
for the induction of acute paw edema in rats [45]. Based on other
pleurisy induced by a single intrapleural injection of 0.1 mL of
studies, it was found that the extract may contain rutin, quercetin,
carrageenan. The extract (250 mg/kg) administered 0.5 h before
luteolin, hesperidin, and bioflavonoids. The extract, which was
inflammation, diminished level of pro-inflammatory cytokines such
given at a dose of 100 mg/kg b.w., per os, produced a reduction in
as TNF-α and IL-1β, inhibited the activity of myeloperoxidase, and
inflammatory processes. In contrast, the extract at a dose of 200
migration of leukocyte and neutrophils. Vargas et al., [44] proved
mg/kg produced analgesic activity. The anti-inflammatory action of
that not only aqueous extracts from leaves of Passiflora edulis
the extract was observed in both models; however, only on paw
(with 1.9% w/w of total flavonoid content) but also of Passiflora
edema induced by histamine, this extract was active as indometha-
alata (with 4.04% w/w of total flavonoid content) inhibited in-
cin [45].
flammation in a mouse model of pleurisy induced by a single intra-
In the current study performed by de Lavor et al., [85], the pleural injection of 0.1 ml of carrageenan (1%) through the influ-
ethanolic extract from leaves and stems of Passiflora cincinnata ence of this extracts (100 or 250 mg/kg, i.p.) on the migration of
was tested in a similar model of paw edema, which has been in- leukocytes and inhibition of myeloperoxidase and adenosine-
duced using carrageenan (20 μl of 1% suspension in 0.9% sterile deaminase activities, and decreasing C-reactive protein in serum.
saline solution) and histamine (at a concentration of 100 Detailed information is shown in Table 1.
μg/paw).The presence of flavonoids like isoorientin, vitexin, and
isovitexin was indicated in the extract. In this study, three doses of 5.1.5. Other studies
the extract, 100, 200, and 400 mg/kg, were tested. Pharmacological Apart from the above discussed studies [55], the anti-
results showed that extract inhibited the volume of the edema in inflammatory activity of methanolic extract from fruits of three
both tests. However, the most effective was the extract in a dose of species, such as Passiflora edulis var. flavicarpa, P. edulis var.
400 mg/kg. Sims, P. ligularis var. Juss, was tested using cell culture of colorec-
Similarly, as a result of Benica’s study [42], the mechanism of tal adenocarcinoma (Caco-2). The extracts contain mainly cyanidin
action of this extract was based mainly on the inhibition of the leu- 3-rutinoside, (+)-catechin, and ferulic acid in quantifiable amounts.
cocytes migration and the reduction of neutrophils. In addition to Additionally, quercetin 3-glucoside was detected only in Passiflora
this, the extracts from the leaves of Passiflora edulis f. flavicarpa edulis var. flavicarpa, rosmarinic acid in Passiflora edulis var.
containing isovitexin, vitexin, orientin, and isoorientin, has also Sims, epigallocatechin, and epigallocatechin gallate in Passiflora
been investigated in the model of the air pouch inflammation by ligularis var. Juss. The study focusing on the impact of extracts on
Beninca et al. [42]. The inflammatory process was induced by us- the dysfunction of the intestinal barrier after application of cytoki-
ing carrageenan, histamine, and substance P in mice. Results nes and LPS revealed that the highest inhibitory activity was exhib-
showed similar activity dependent on aqueous extract at a dose of ited by the extract of Passiflora ligularis var. Juss. in comparison
100 mg/kg, i.p. (AE), butanolic fraction of extract (BuOH: 50 with other extracts at the same concentration at 10 mg/dL. This
mg/kg, i.p.), and an aqueous residual fraction (AR: 100 mg/kg, i.p.) extract makes the value of transepithelial electrical resistance to
used in tests. It was revealed that the extracts diminished the migra- near 73% of the initial value. The authors concluded that all the
tion of leucocytes (by 10%-48%), neutrophils (by 29.2%-58.2%). extracts had the ability to inhibit cell barrier dysfunction in in vitro
Moreover, the extracts and fraction decreased levels of cytokines model used in experiments. Results may show implications of the
such as MIP-2 (by 31%-78%) and IL-1β (38.5%-74%). extract in the prevention of many inflammatory diseases involving
disorders of the intestinal barrier. At present, several studies have
5.1.3. The Activity of Extracts During the Inflammatory Process shown anti-inflammatory properties of polyphenolics using the in
in Diabetes vitro cell model of an inflammatory process in the epithelium of
Figueiredo et al., [126] studied anti-inflammatory and anti- human intestines [96, 127-132].
diabetogenic properties of aqueous extract from the leaf of P. alata
and its effectiveness in non-obese diabetic mice. The objectives of 6. ANTI-INFLAMMATORY STUDIES OF FLAVONOIDS
this study have been justified by the fact that inflammatory cells There is ample evidence that flavonoids have broad pharmacol-
occurring in the pancreatic islets can promote the destruction of ogical activities, hence their growing popularity and multipoint
beta cells and consequently can decrease the secretion of insulin medical use. Many studies have focused on testing of flavonoids for
during the progression of type 1 diabetes. After the administration their not only cardioprotective, neuroprotective, anti-hypertensive,
of P. alata extract (containing vitexin, isovitexin, and isoorientin) and anticancer properties but also anti-inflammatory activity [49,
for 30 weeks in diet, a tendency towards reduced numbers of in- 133, 134]. Many flavonoids and phenolic acids present in various
flammatory cells in the islets and a decrease of 28.6% in diabetes extracts of Passiflora may lead to anti-inflammatory actions by
incidence were observed. Moreover, the extract of P. alata was able numerous mechanisms of biological action and by influencing me-
2590 Current Pharmaceutical Design, 2021, Vol. 27, No. 22 Ożarowski and Karpiński

diators and effectors of inflammatory processes [48, 49, 50]. The synthesis of nitric oxide and prostaglandin E2, diminished the ex-
advancement in the knowledge of molecular mechanisms of fla- pression of inducible synthase of nitric oxide and cyclooxygenase-2
vonoids has been observed for many years. These natural chemical in vitro. Moreover, Odontuya et al., [149] observed that luteolin
compounds decreased the concentration of pro-inflammatory pros- and their derived C-glycosides inhibited the biosynthesis of throm-
taglandins and leukotrienes by inhibition of the activity of cy- boxane and leukotrienes. Aziz et al., [65] summed up that luteolin
clooxygenase, lipoxygenase, and phospholipase (Table 2). Moreo- exerts strong anti-inflammatory activity in vitro and in vivo, in
ver, flavonoids inhibited phosphodiesterases and kinases and re- silico and in clinical studies, and its mechanisms of action include
duced the expression of cyclooxygenase COX-2 [121, 123]. It is few pathways for the nuclear factor, mitogen-activated protein
important to emphasize that flavonoids that have anti-inflammatory kinase, the signal transducer and activator of transcription 3.
activity can prevent a disorder of the biological structure and Over the years, several investigations have suggested that api-
physiological function of the intestinal barrier [55,111]. genin may show beneficial effects in various diseases with inflam-
One of the most investigated flavonoids in this field is quer- matory conditions. In recent years, a lot of studies were carried out
cetin (3,3’,4’,5,7-pentahydroxyflavone), which showed multiple to explain the mechanism of the anti-inflammatory activity of api-
mechanisms of anti-inflammatory activity in various models such genin. The medical database of PubMed contains 280 scientific
as in animal and human cell lines, and in vivo model. Lee et al., articles on this field of 2010 till 2020. Apigenin has been studied in
[135] observed that quercetin inhibited in vitro synthase induced by various models in vitro and in vivo. Many tests were performed
nitric oxide, production of nitric oxide, and interleukin-6, the nu- using human cell lines like i.e. human endothelial cells [150], BV2
clear translocation of nuclear factor-B (NF-B), which are bio- microglia [151], bronchial epithelial cells [152], lung epithelial
markers of inflammation. Other studies showed that quercetin also cells [153], human and mouse macrophages [154, 155], and human
inhibited mediators such as MAPK/AP-1 and IKK/NF-B, protein pluripotent stem cell model of Alzheimer’s disease [156]. Moreo-
kinase C vascular superoxide production, and also tyrosine phos- ver, for the assessment of anti-inflammatory effects of apigenin,
phorylation [136-138]. Many studies on human cell lines have few in vivo studies were carried out such as on the model of asth-
shown that quercetin exerted anti-inflammatory action by down- matic mice [157], on the model of polycystic ovary syndrome in
regulation of vascular cell adhesion molecule 1 [139], by reducing rats [158], colonic inflammation in a mouse model of diet-induced
the gene expression of tumor necrosis factor  and interleukins obesity [159], on a rat model of Parkinson's disease [160], on mice
such as IL-1, IL-8 and IL-6 [140], and by decreasing the concen- with liver injury [161], on rats model of lung injury [162], on the
tration and activity of cyclooxygenase-2 [141] and lipooxygenase model of arthritis in rats [163], and a rat’s model of sepsis [164].
[142, 143]. Lim et al., [144] also observed that quercetin reduced It is well-known that apigenin acts not only as an antioxidant
the induction of metalloproteinase-1, activation of extracellular but also as an anti-inflammatory molecule through inhibition of
signal-regulated protein kinase. Furthermore, Bahareh et al., [77] mediators and signaling pathways [17]. Several studies have re-
showed that quercetin significantly inhibited the productions of the ported that anti-inflammatory mechanism of apigenin action is
tumor necrosis factor, nitric oxide, and myeloperoxidase in human based on inhibition/downregulation of the signaling pathways such
neutrophils. as the nuclear factor-kappa B (NF-κB) [143, 154, 162; 165-167],
Another flavonoid, chrysin (5,7-dihydroxyflavone), also ex- STAT3-NF-κB [168] the TLR4/NF-κB [169], and P2X7/NF-κB
erted anti-inflammatory activity in vitro [145-148]. Previously, it [163] leading to decreasing cytokines release. Moreover, apigenin
was shown that this flavonoid inhibited expression cyclooxygenase- influenced the COX2/PGE2 axis [155, 170], activated the
2 via IL-6 signaling [145]. Recently, it was observed that this fla- GSK3β/Nrf2 [151], and the Nrf2 signaling pathways [166]. It was
vone inhibited the inflammatory process in endothelial cells via the shown that apigenin subsequently attenuated the synthesis of in-
NF-κB signaling pathway and decreased the levels of pro- flammatory cytokines, like TNF-α, IL-1β, and IL-6 [151, 154, 159,
inflammatory mediators [148]. Chrysin also diminished the expres- 165], and it was revealed that this flavonoid decreased the expres-
sion of vascular cell adhesion molecule-1 in endothelial cells, inhib- sion of pro-inflammatory cytokines and adhesion molecules by 30-
ited nuclear factor-κB, protein kinase activated by p38 mitogen, and 70% [167]. Furthermore, it reduced the expression of LOX-1 and
c-Jun N-terminal kinase. Because chrysin decreased the adhesion of NLRP3 and inhibited adhesion of leukocytes in vitro [150], and
leukocytes to the endothelium, thus this anti-inflammatory effect is down-regulated the release of nitric oxide from inflammatory cells
considered as therapeutic potential in the treatment of multiple scle- [156]. It should be noted that the anti-inflammatory action of api-
rosis and other neurodegenerative disorders [146]. genin was more effective in comparison with the other 13 flavon-
Next, a natural chemical compound such as luteolin (3', 4', 5,7- oids tested [152]. Results of studies revealed that apigenin includ-
tetrahydroxyflavone) at doses of 30 and 50 mg/kg used in rat paw ing above mechanisms of action, inhibited neuro- [148, 151, 160]
edema decreased serum levels of proinflammatory cytokines i.e. and colonic inflammation [159,171], attenuated atherogenesis [172]
tumor necrosis factor-, interleukins (IL-1β, IL-6) and myeloper- and arthritis [163] and decreased inflammatory processes in the
oxidase [67]. These effects were observed previously by Chen et al. liver [161], lungs [162], spleen [164] and testicular cells [165].
[62]. Also another study [64] showed that luteolin at a concentra- Based on the above considerations, it can be concluded that api-
tion of 0.5 μM inhibited not only TNF- but also a nuclear factor, genin is a universal molecule with a wide anti-inflammatory effect
expression of protein-1 in the chemokine monocytes and adhesion observed during many models of disorders, and may be considerate
of various molecules to vascular cells. Biochemical results may as an interesting drug candidate to new treatments of not only coli-
suggest that luteolin is a promising molecule that can inhibit in- tis but also of Parkinson’s and Alzheimer’s disease and many other
flammation by decreasing the signaling pathway of NF-κB and by diseases occurring with inflammation.
improving the function of vascular cells in vitro. Similar effects The previous study showed that not only quercetin, luteolin,
were observed in mice fed a diet containing 0.6% luteolin for 3 and apigenin, but also rutoside (3,3′,4′,5,7-pentahydroxyflavone-3-
weeks. Moreover, when luteolin and one of its glycosidic forms - rhamnoglucoside; rutin) produced anti-inflammatory activity [173,
luteolin-7-O-glucoside (50 mg/kg b.w.) were given daily for 3 174]. Nikfarjam et al., [175] revealed that this compound signifi-
weeks to mice with liver injury, it was observed that these natural cantly diminished the production of nitric oxide and tumor necrosis
compounds protected liver cells by regulation of mediators of in- factor- in peripheral blood neutrophils. Moreover, it was observed
flammation and phase II enzymes [66], however, luteolin-7-O- that rutoside reduced myeloperoxidase activity in vitro. In the
glucoside only changed the activation of NF-κB [63]. Previously, model of acute and chronic inflammation, this flavonoid (80 mg/kg,
Park et al., [63] and Chen et al., [62] showed that luteolin and one i.p.) inhibited the chronic phase the most in comparison with
of its glycosidic forms, luteolin-7-O-glucoside, also inhibited the
Extracts and Flavonoids of Passiflora Species as Promising Anti-inflammatory Current Pharmaceutical Design, 2021, Vol. 27, No. 22 2591

Table 2. Anti-inflammatory activities of flavonoids in various experimental models.

Concentration or dose Effect of flavonoids on inflammatory mediators


Flavonoid Experimental model Refs.
with dosage regimen / other factors / functions

Animal model of inflammation

- decreasing the protein level of TNF-α, IL-1β, and MCP-1 in a


dose-dependent manner
40 mg/kg, p.o., rats with abdominal - reduction in expressions and protein levels of MMP-2 and
aortic aneurysms indu- MMP-9 - max. the inhibitory effect after 40 mg/kg
Apigenin 80 mg/kg, p.o., [143]
ced by 0.5 mol/L of
for 2 months CaCl2 - reduction in phosphorylation of IκB and phosphorylation of
IKKβ
- blocking the activity of NF-κB

- decreasing the percentage of eosinophils, as well as neutrophil


infiltration in the lungs of asthmatic mice
20 mg/kg, p.o. BALF mice with asthma
- down-regulating the expression of NF-κB p65 submit
Apigenin induced by particulate [157]
for 7 days matter (PM2.5) - reducing the levels of total serum immunoglobulin IgE by
about 78% and IL-4 by about 64%, IL-13 by about 33%, IL-17
by about 44.2%

20 mg/kg, p.o. - reducing the TNF-a level by about 28% after apigenin at 20
polycystic ovary syn- mg/kg, and by about 37.3% after apigenin at 40 mg/kg
Apigenin 40 mg/kg, p.o. [158]
drome in the rat model - reducing the IL-6 level by about 32% after apigenin at 20
for 21 days mg/kg and by about 41% after apigenin at 40 mg/kg

- reducing the MDA level by 22.7%


- decreasing the colonic levels of IL-1β by 57% and IL-6 by
10 mg/kg, p.o. C57BL/6J mice fed a
Apigenin 74% [159]
for 8 weeks high-fat diet
- diminishing the eosinophil infiltration
- decreasing the iNOS expression

- inhibiting the release of pro-inflammatory cytokines: TNF-α


by about 18.5%, IL-6 by about 23.5%, NF-κB by about 13%
and pro-inflammatory enzyme iNOS-1 by about 16% after api-
10 mg/kg, i.p. a rotenone-induced rat genin at 10 mg/kg with rotenone
Apigenin 20 mg/kg, i.p. model of Parkinson's [160]
- decreasing the lipid peroxides level, the substantia nigra after
for 14 days disease
apigenin at 10 and 20 mg/kg with rotenone
- increasing the SOD and CAT activity in the striatum after
apigenin at 10 and 20 mg/kg with rotenone

- decreasing the hepatic TNF-α content by about 19% after


apigenin at 100 mg/kg and by about 23.5% after apigenin at
200 mg/kg
100 mg/kg, p.o. d-galactosamine / LPS- - increasing the SOD level by about 23.5% after apigenin at 100
Apigenin 200 mg/kg, p.o. induced liver injury in mg/kg and by 17.6% after 200 mg/kg, CAT level by 15.8% at [161]
mice 200 mg/kg
for 7 day
- increasing the GST level and GR level after apigenin at 200
mg/kg
- reducing the hepatic NF-κ p65 protein expression and level

- reducing the expression in the left lung:


- the TNFα expression by 70% after a single dose and by 71.7%
after double dose apigenin
acute ischemia- - the iNOS expression by 79.4% after a single dose and by 66%
25 mg/kg, i.p.
Apigenin reperfusion double dose apigenin [162]
single or double dose
injury of the lung in rats - the IL-6 expression by 85% after a single dose and by 76.5%
after double dose apigenin
- reducing the NF-κB activation by 40.8% after a single dose
and by 66.1% at double dose apigenin
(Table 2) Contd....
2592 Current Pharmaceutical Design, 2021, Vol. 27, No. 22 Ożarowski and Karpiński

Concentration or dose Effect of flavonoids on inflammatory mediators


Flavonoid Experimental model Refs.
with dosage regimen / other factors / functions

- decreasing the serum concentration of TNF-α by about 24%


after 5 mg/kg, by about 14% at 20 mg/kg, and by about 20.5%
at 40 mg/kg
5 mg/kg, p.o., - decreasing the serum concentration of IL-6 by about 43% (5
20 mg/kg, p.o. Freund’s complete adju- mg/kg), by about 13% (20 mg/kg), by about 30% (40 mg/kg)
Apigenin vant-induced arthritis in [163]
40 mg/kg, p.o. rats - up-regulating the expression of p-NF-κBp65, p- IKKα, P2X7,
for 7 days p-IKKβ, p-IκBα of ankle joint tissue
- protection for synovial inflammatory cell infiltration, articular
cartilage lesion, epithelial cell degeneration, and synovial tis-
sue edema after apigenin at 40 mg/kg

- reducing the proinflammatory cytokine levels:


- TNFα by 7.5%, IL-1β by 15% and IL-6 by 19% after pretreat-
ment with apigenin at 20 mg/kg
20 mg/kg, p.o., polymicrobial sepsis rat
- TNFα by 22.6%, IL-1β by 20.7% and IL-6 by 37% after pre-
model induced by cecal
Apigenin 40 mg/kg, p.o., treatment with apigenin at 40 mg/kg [164]
ligation and puncture
60 mg/kg, p.o. (CLP) - decreasing the lipid peroxidase (LPO) levels by apigenin at 40
mg/kg, and catalase (CAT) levels by apigenin at 20 and 40
mg/kg,
- increasing the SOS activity by all doses

- decreasing the increase of TNF-α by about 30.4% and 32%,


IL-6 by about 37% and 40%, and IL-1β by about 31.7% and
234 mg/kg, p.o. acrylonitrile-induced 33% in rat testes after 234 and 468 mg/kg of apigenin, respec-
Apigenin 468 mg/kg, p.o. inflammation in testicu- tively [165]
for 12 weeks lar cells in rats - decreasing the protein levels of NF-κB by about 45% and
27.4% in rat testes after 234 and 468 mg/kg, respectively
- down-regulating the caspase-3 levels in testicular tissue

- inhibiting the ulcerative colon damage


- inhibiting the production of TNF-α by about 37.5% and 61%,
IL-1β by about 52% and 66.5%, IL-6 by about 38.5% and
200 mg/kg, p.o., ulcerative colitis indu- 60%, MCP-1 by about 41.5% and 63.5% after 200 mg.kg and
Apigenin 300 mg/kg, p.o. ced by dextran sulfate 300 mg/kg of apigenin, respectively [168]
for 20 days sodium in mice - decreasing the expression of COX-2 by about 32% and 53%
after 200 mg/kg and 300 mg/kg of apigenin, respectively
- reducing the MPO activities by about 26% and by 36% after
200 and 300 mg/kg of apigenin, respectively

- reducing the oxidative stress by increasing the level of gluta-


thione (GSH), the activity of glutathione peroxidase (GSH-Px)
117 mg/kg, p.o. after all doses of apigenin
234 mg/kg, p.o. acrylonitrile-induced - increasing the SOD activity after apigenin at 234 mg/kg
Apigenin neuroinflammation in [169]
351 mg/kg, p.o. - downregulating the TLR4/NF-κB signaling pathway
rats
for 28 days - decreasing the concentration of TNF-α and IL-6 after 117
mg/kg
- decreasing the mRNA expression of NF-κB after all doses

- alleviating the injuries such as leukocyte infiltration, eryth-


rocyte aggregation, and interstitial edema after pretreatment of
40 mg/kg, p.o. LPS - induced vascular
Chrysin chrysin at 40 mg/kg and 80 mg/kg [148]
80 mg/kg, p.o. inflammation in mice
- suppressing the leukocyte-endothelial cell adhesion and
inflammation

Isoorientin 15 mg/kg, p.o. carrageenan-induced - inhibiting inflammation between 28-37% (at 15 mg/kg) and
[187]
30 mg/kg, p.o. hind paw edema of mice 36-43% (at 30 mg/kg doses)

(Table 2) Contd....
Extracts and Flavonoids of Passiflora Species as Promising Anti-inflammatory Current Pharmaceutical Design, 2021, Vol. 27, No. 22 2593

Concentration or dose Effect of flavonoids on inflammatory mediators


Flavonoid Experimental model Refs.
with dosage regimen / other factors / functions

- decreasing TNF-α by about 50% (at 50 mg/kg) and 68% (at 100
mg/kg) and I L-6 production by about 43% (at 50 mg/kg) and
70.6% (at 100 mg/kg)
- reducing the MPO activity by about 38% (at 50 mg/kg) and 58%
(at 100 mg/kg), and MDA content by about 51% (at 50 mg/kg)
LPS-induced acute and 68% (at 100 mg/kg)
50 mg/kg, i.p. [177]
Isovitexin lung
100 mg/kg, i.p. - increasing SOD by about 95% (at 100 mg/kg) and GSH levels by
Injury in mice about 90.5% (at 50 mg/kg) and 105% (at 100 mg/kg)
- inhibiting the expression of iNOS by about 28.3% (at 50 mg/kg)
and 56.6% (at 100 mg/kg) and COX-2 by about 32% (at 50
mg/kg) and 54% (at 100 mg/kg)
- diminishing the ROS generation

- reducing the increased circulating levels of MCP-1/JE, KC (the


C57BL/6 mice treated mouse homolog of human MCP-1 and IL-8, respectively), and
Luteolin diet containing 0.6%
with TNF-α at 25 sICAM-1 in mice by 61%, 93%, and 35%, respectively [64]
luteolin
μg/kg (i.p.) - complete blocking of the F4/80-positive monocytes-derived
macrophages

- attenuating the inflammatory cell infiltration and necrosis by the


3-week administration of luteolin
Luteolin galactosamine/LPS-
- attenuating the inflammatory mediators and their transcription
50 mg/kg, p.o. induced hepatitic ICR [66]
factors, NF-κB and AP-1
mice
- ameliorating the increased TNF-α production by about 51% and
COX-2 expression by about 69%

- decreasing the paw edema in dose dependent manner (max.


protective effect after 24 h with 50 mg/kg luteolin)
- decreasing the serum level of TNF-α (from 68.24 ng/mL to 39.28
Luteolin 30 mg/kg, i.p., monosodium urate
ng/mL), IL-1β (from 20.18 ng/mL to 12.07 ng/mL), and IL-6
crystal-induced rats [67]
50 mg/kg, i.p., (from 39.35 ng/mL to 24.72 ng/mL) after 50 mg/kg of luteolin
paw edema
- reduction of lipid peroxidation by 52%, level of glutathione
peroxidase by 40%, level of SOD by 36%, level of CAT by
37.5% after 50 mg/kg of luteolin

- decreasing the edema produced in the acute phase after rutin by


53% and quercetin by 59%
Rutoside adjuvant - carragee-
- decreasing the edema produced in the chronic phase after rutin by
and 80 mg/kg, i.p. nan-induced inflam- [174]
73% after 30 days, but quercetin without significant activity
Quercetin mation in rats
- suppressing the arthrogram scores by 100% after rutin and by
50% after quercetin

Rutoside - the highest anti-inflammatory effect after quercetin = inhibiting


150 mg/kg i.p. of rutin, carrageenan-induced
and the edema by 66% after 1 h and by 63% after 7 h [176]
75 mg/kg i.p. of quercetin paw edema in mice
Quercetin - no significant activity after rutin

Rutoside - decreasing the edema produced in the chronic phase after rutin by
adjuvant arthritis test 53% after 3 h (6 days) and by 61% after 10 days
and 75 mg/kg i.p. [176]
in mice - reducing the edema produced in the chronic phase after quercetin
Quercetin by 59% after 3 h (6 days) and by 70% after 10 days

Rutoside
50 mg/kg s.c. of rutin, cotton pellet granu- - inhibiting the granuloma formation by 24% after rutin
and [176]
25 mg/kg s.c. loma test in rats - inhibiting the granuloma formation by 26% after quercetin
Quercetin

- inhibiting the production of TNF-α by about 36.5%, IL-1β by


about 78%, IL-6 by about 35.5%, IL-33 by about 33%
Vitexin 10 mg/kg, i.p., carrageenan-induced - enhancing the production of anti-inflammatory cytokine (IL-10)
[76]
single dose inflammation in mice by about 77%
- prevention in the carrageenan-induced decrease of GSH levels
(10 mg/kg, i.p.,)
(Table 2) Contd....
2594 Current Pharmaceutical Design, 2021, Vol. 27, No. 22 Ożarowski and Karpiński

Concentration or dose Effect of flavonoids on inflammatory mediators


Flavonoid Experimental model Refs.
with dosage regimen / other factors / functions

5 mg/kg, p.o., carrageenan, zymo-


san LPS-induced - max. inhibitory effects on TNF-α level at the dose of 30mg/kg
15 mg/kg, p.o.,
Vitexin leukocytes migration (69.9%) and IL-1β level at the dose of 15mg/kg (56.4%) [180]
30 mg/kg p.o. to the peritoneal - max. reduction in NO concentration at 15mg/kg (70.9%)
single dose cavity in mice

- decreasing the levels of TNF-α by about 29%, iNOS by about


2 μM, 36.5%, nNOS by about 68.5%, COX-2 by about 26.5%, p-38 by
acrylamide-induced
10 μM, neuroinflammation in about 69% after 10 μM of vitexin
Vitexin [181]
20 μM, zebrafish larvae - up-regulating the nuclear translocation of Nrf2-mediated expressions
(Danio rerio) of SOD by about 66% and CAT by about 50% after 10 μM of vite-
for 24 h
xin

- decreasing the concentrations of TNF-α by about 35%, IL-1β by


10 mg/kg, i.p. LPS- induced acute
Vitexin about 55%, and IL-6 by about 50% [185]
single dose lung injury in mice
- reducing the ROS levels by 44%

- attenuating the migration of eosinophil, neutrophil, and mononuclear


cells and Th2 cytokines level in bronchoalveolar lavage fluid
- reducing the total leukocytes (87%), eosinophils (95%), neutrophils
0.2 mg/kg, murine ovalbumin- (89%), mononuclear cells (79%)
induced allergic
Vitexin 1 mg/kg, - reducing the concentrations of the three cytokines at all doses of [186]
asthma model in
5 mg/kg, mice vitexin with maximum effect for IL-4 (64%) at 1 mg/kg, for IL-5
(96%) at 0.2 mg/kg and for IL-13 (65%) at 1 mg/kg of vitexin
- decreasing the plasma IgE concentration at all doses (79% at 0.2
mg/kg)

Another model for inflammatory processes

- attenuating the increased contents of TNFα by about 34% and 52%,


of IL-1β by about 43.5% and 55.5%, and of IL-6 by about 37.5%
and 50% after 100 μM and 200 μM of apigenin, respectively
human renal epithe- - decreasing the level of MDA by about 32% and 54% after 100 μM
100 μM,
Apigenin lial cell HK-2 incuba- and 200 μM of apigenin, respectively [166]
200 μM ted with D-glucose - increasing the activity of SOD by about 55.5% and 116%, and of
CAT by about 156% and 200% after 100 μM and 200 μM of apige-
nin, respectively
- increasing the mRNA expression of an Nrf2 response gene

- diminishing the leukocyte adhesion and expression of adhesion


ISO-HAS human molecules, i.e. VCAM-1 after 30 μM of apigenin (132%) and 50 μM
10 μM, endothelial cells apigenin (127%), ICAM-1 after 30 μM of apigenin (131%) and 50
Apigenin 30 μM, exposed to trimethy- μM of apigenin (128%) [150]
50 μM lamine-N-oxide
(TMAO) - reducing the protein expression of LOX-1 and NLRP3 after 50 μM
of apigenin

- suppressing the production of these inflammatory cytokines in a


10 μM, LPS-stimulated dose-dependent manner, maximal effect after apigenin at 40 μM:
Apigenin 20 μM, murine BV2 micro- TNF-α by about 83.5%, IL-6 by about 81%, IL-1ß by about 79% [151]
40 μM glial cells - up-regulating the expression of GSK3b, Nrf2, and HO-1
- inhibiting the LPS-induced NF-κB activation

- reducing levels of the IL-6 by about 52.4% after apigenin > by about
40.5% after luteolin
- reducing the level of IL-8 by about 54.5% after luteolin > by about
LPS-stimulated 38.5% after apigenin
Apigenin human bronchial
10-6 - 10-4 M - inhibiting the MCP-1 release by 68% after luteolin > by about 54% [152]
Luteolin epithelial cell line
after apigenin
(BEAS-2B cells)
- decreasing the LPS-induced activity of NF-κB p65 by about 45.5%
after apigenin with LPS
- inhibiting the level of MDA by about 35% after apigenin with LPS
(Table 2) Contd....
Extracts and Flavonoids of Passiflora Species as Promising Anti-inflammatory Current Pharmaceutical Design, 2021, Vol. 27, No. 22 2595

Concentration or dose Effect of flavonoids on inflammatory mediators


Flavonoid Experimental model Refs.
with dosage regimen / other factors / functions

- decreasing the LPS - induced mRNA levels of iNOS and COX-2


in a dose-dependent manner - inhibition in iNOS by 10% and
COX2 by 26% after apigenin at 40 μM
LPS-stimulated human - decreasing the NO production and release in a dose-dependent
Apigenin lung alveolar type II manner
10 μM – 100 μM [153]
epithelial cells (A549 - inhibiting the LPS - induced expression of pro-inflammatory
cells) cytokines (IL-1β, IL-2, IL-6, IL-8, and TNF-α) in a dose-
dependent manner after apigenin at 20 μM and 40 μM
- decreasing the AP-1 proteins: mRNA transcripts of c-Jun, c-Fos,
and JunB in a dose-dependent manner (20 μM and 40 μM)

- inhibiting the IL-6 by about 94% after 25 μM of apigenin and by


6.25 μM, 60% after 12.5 μM of apigenin,
LPS - stimulated human
Apigenin 12.5 μM, - decreasing the IL-1β by about 49% after 25 μM of apigenin and [154]
THP-1 monotypic cells
25 μM by about 37% after 12.5 μM of apigenin,
- reducing the TNF-α by 53% only after 25 μM of apigenin

functional neurons from - promoting a global down-regulation of cytokine and nitric oxide
human induced pluripo- release in inflammatory cells
tent stem cell (iPSC)
5 μM, - protection against apoptosis, neuronal death by apigenin at 50 μM
model,
Apigenin 10 μM, - reducing the caspase-3/7 activity levels by apigenin at 50 μM [156]
RAW264.7 cells,
50 μM - inhibiting the levels of 23 cytokines i.e.
C8B4 cells
IC50 for TNF-α = 75 (RAW264.7), 27 (C8B4), 19 (PM),
primary microglial
(PM) culture IC50 for IL-6 = 46 (RAW264.7), 63 (C8B4), 38 (PM)

human umbilical vein - suppressing the production of ROS (30% relative to control)
endothelial cells using 10 μM of apigenin
Apigenin 0–100 μM (HUVECs) incubated - decreasing the protein expression of proinflammatory cytokines [167]
with glycation end (IL-6, TGF-β1) and adhesion molecules (MCP-1) by 30–70% in a
products (AGEs) dose-dependent manner

- blocking the adhesion of monocytes to bEnd.3 cells exposed to


LPS by 83.9%
- attenuating the IκBα degradation from 29.7% to 94.6% at 100 µM
LPS - stimulated brain - decreasing the phosphorylation of p38 mitogen-activated protein
Chrysin 10 μM –100 µM endothelial cells kinase (MAPK) from 289.8 % to 173.2% at 100 µM [146]
(bEnd.3 cells) - decreasing the phosphorylation of -Jun N-terminal kinase (JNK)
from 240.2% to 92.0% at 100 µM
- reducing the VCAM-1 mRNA expression in a concentration-
dependent manner from 435.8% to 201.4%

- suppressing the increase in mRNA expression and protein levels


human umbilical vein of ICAM-1, VCAM-1, and E-selectin
1 mM, 3 mM, 10 mM,
Chrysin endothelial cells [148]
30 mM - inhibiting the induction of the p-IkBa levels by treatment with 10
(HUVECs)
mM or 30 mM of chrysin

- inhibiting the LTB4 synthesis after 50 µg/mL (33.6%), 100


µg/mL (28.24%)
peritoneal leukocytes of
Isoorientin 25, 50 and 100 µg/mL - inhibiting the TXB2synthesis after 25 µg/mL (54.33%), 50 µg/mL [149]
rats
(56.34%), 100 µg/mL (66.42%)
- inhibiting the COX-2 and 5-LOX activity

- diminishing the AngII-induced increase of the superoxide anion


production in the smooth muscle cells after isorhamnetin (10
Isorhamnetin 1 or 10 mmol/L rings of thoracic aortae mmol/L) [136]
- preventing the in AngII-induced endothelial dysfunction after
incubation with isorhamnetin (10 mmol/L)

- prevention of the endothelin-1 increased intracellular [137]


Isorhamnetin 1 and 10 µM rings of thoracic aortae O2•−production and A23187-stimulated O2•−generation after incu-
bation with 10 µM of isorhamnetin
(Table 2) Contd....
2596 Current Pharmaceutical Design, 2021, Vol. 27, No. 22 Ożarowski and Karpiński

Concentration or dose Effect of flavonoids on inflammatory mediators


Flavonoid Experimental model Refs.
with dosage regimen / other factors / functions

- reducing the NO production with IC50 = 58.5 µM


Isovitexin LPS-stimulated RAW
- inhibiting IKK kinase activity and decreasing the iNOS protein
5 µM - 100 µM 264.7 murine macro- [74]
expression in a dose-depending manner
phages
- preventing the translocation of NF-κB

- reducing the secretion of TNF-α by about 57.5% and IL-6 by


about 54.5% after 50 µg/mL
- decreasing the expression of iNOS by about 93% and COX-2 by
25 μg/mL, LPS-stimulated RAW about 67.7% after 50 µg/mL
Isovitexin 264.7 murine macro- [177]
50 μg/mL - inhibiting the phosphorylation of MAPK, reducing the nuclear
phages
translocation of NF-κB,
- upregulating the expression of Nrf2 and HO-1
- suppressing intracellular ROS generation

- suppressing the LPS-induced gene expressions of TNF- α, IL-6,


iNOS, and COX-2 in a dose-dependent manner
mouse alveolar
macrophages stimula- - reducing the DNA binding activity of NF-κB
Luteolin [62]
5.0 μM – 50 μM ted by LPS (MH-S - blocking the degradation of IκB-α and NF-κB p65 subunit
and RAW 264.7 - attenuating the LPS-mediated protein kinase B
cells)
- attenuating the production of reactive ROS after preincubation
with luteolin (5, 10 and 25 μM)

- suppressing the NO (IC50 = 13.9 μM) and PGE2 (IC50 = 7.4 μM)
Luteolin LPS-stimulated RAW
- inhibiting the LPS-induced expression of iNOS and COX-2 in a
5 μM – 50 μM 264.7 murine macro- [63]
dose-dependent manner
phages
- inhibiting the Akt phosphorylation

- blocking the inflammation-induced adhesion of monocytes to


endothelial cells by luteolin at 20 μM
Luteolin EA.hy926 endothelial - inhibiting the TNF-α-induced gene expression of MCP-1,
0.5 μM – 20 μM [64]
cells VCAM-1, ICAM-1, and chemokine by luteolin at 0.1 μM
- complete prevention of the TNF-α-induced gene expression of
MCP-1, VCAM-1, ICAM-1, and chemokine by luteolin at 2 μM

- inhibiting the LTB4 synthesis after 25 µg/mL (95.29%), 50


25 µg/mL, µg/mL (97.71%), 100 µg/mL (96.27%)
Luteolin peritoneal leukocytes
50 µg/mL, - inhibiting the TXB2 synthesis after 25 µg/mL (39.38%), 50 [149]
of rats
100 µg/mL µg/mL (64.76%), 100 µg/mL (76.88%)
- inhibiting the COX and 5-LOX activity

- decreasing 86.09%, in NO production in the cells during a 4-h


incubation period with quercetin (25 μM)
Quercetin cultured neutrophils [77]
- inhibition of 79.31% in TNF-α production after incubation with
1.0 μM - 100 μM isolated from human
quercetin (25 μM)
blood
- reducing the MPO activities by 83.7% in the cells treated with
quercetin

- reducing the NO production in a dose-dependent manner after


pretreatment with 12.5-25 μM of quercetin
- decreasing the iNOS expression after pretreatment with 6.25-25
LPS - stimulated μM of quercetin
Quercetin
6.25μM - 25 μM RAW264.7 murine - reducing the IL-6 production in a dose-dependent manner after [135]
macrophages pretreatment with 6.25-25 μM of quercetin
- attenuating the nuclear translocation of NF-κB
- no effect on the production of TNF-α, IL-6, and expression of
COX-2
(Table 2) Contd....
Extracts and Flavonoids of Passiflora Species as Promising Anti-inflammatory Current Pharmaceutical Design, 2021, Vol. 27, No. 22 2597

Concentration or dose Effect of flavonoids on inflammatory mediators


Flavonoid Experimental model Refs.
with dosage regimen / other factors / functions

- diminishing the AngII-induced increase of the superoxide anion


production in the smooth muscle cells after quercetin (10
Quercetin 1.0 mmol/L, rings of thoracic mmol/L) [136]
10 mmol/L aortae
- preventing the in AngII-induced endothelial dysfunction after
incubation with quercetin (1 or 10 mmol/L)

- prevention of the endothelin-1 increased intracellular


O2•−production and A23187-stimulated O2•−generation after incu-
Quercetin 1.0 μM, rings of thoracic [137]
bation with 10 µM of quercetin
2.0 10 µM aortae
- inhibition of the increased PKC activity induced by endothelin-1
after incubation with 10 µM of quercetin

- inhibiting the LPS-induced NO production in a concentration-


dependent manner,

Quercetin LPS - stimulated - inhibiting the secretion of PGE2, iNOS, COX-2, TNF-α, IL-1β,
5.0 μM – 20 µM RAW 264.7 murine IL-6, and GM-CSF mRNA and protein expressions at higher con- [138]
macrophages centrations
- inhibiting the LPS-induced NF- κB activation by quercetin in a
time- and dose-dependent manner

- decreasing the expression of VCAM-1 and cluster of differentia-


Quercetin human umbilical vein tion after pretreatment of quercetin at 210 μM
0 - 210 μM endothelial cells [139]
(HUVECs) - the protective effects against oxidative damage at concentrations
of 90-210 μM

- decreasing the mRNA levels of IL-6, IL-8, monocyte chemoat-


tractant protein-1, interferon-γ inducible protein-10, IL-1b, TNF-
Quercetin 0.3 μM, 3.0 μM, human U937 mono- α and cyclooxygenase-2 (COX-2) after the 5-h incubation time [140]
10 μM, 30 μM cytes
- decreasing the phosphorylation of JNK and c-Jun after 2.5 h
pretreatment of quercetin in a dose-dependent manner

- suppression of the arsenite-induced the COX-2 expression mainly


by blocking the activation of the PI3K signaling pathway after
pretreatment with quercetin at a concentration of 10 or 20 μM
10 μM, rat liver epithelial
Quercetin - reduction in the production of PGE2 by quercetin at 10 or 20 μM [141]
20 μM (RLE) cells
- attenuation of the arsenite-induced PI3K activity by binding with
PI3K, and the suppression of activation of Akt, p70S6K, and
ERK

- inhibition of activity of the COX-2 by 68% after quercetin at 100


10 μM, epidermal tissue of μM (IC 50 = 50 μM)
Quercetin [142]
100 μM guinea-pigs - inhibition of activity of the LOX by 61% after quercetin at 100
μM (IC50 = 30 μM) (1%) as

- inhibition of the activity of the MMP-1 (IC50 = 39.6 μM)


- suppression of activation of the transcription factor (AP-1) after
human dermal fibro-
Quercetin 0 – 100 μM 25 and 12.5 μM of quercetin [144]
blast (HDF) culture
- inhibition of extracellular signal-regulated protein kinase (ERK)
and p38 mitogen-activated protein kinase (MAPK) activation

- decreasing the production of NO by about 86%


human peripheral
Rutoside 25 μM - decreasing the production of TNF-α by about 71% [175]
blood neutrophils
- reducing the MPO activity by about 80%

- decreasing 86.74% NO production in the cells after vitexin (25


μM) during a 4-h incubation period
cultured neutrophils
- inhibiting 80.94% TNF-α production after incubation with vitexin
Vitexin 1 μM - 100 μM isolated from human [77]
(25 μM)
blood
- reducing the MPO activities by 87.3% in the cells treated with
vitexin
(Table 2) Contd....
2598 Current Pharmaceutical Design, 2021, Vol. 27, No. 22 Ożarowski and Karpiński

Concentration or dose Effect of flavonoids on inflammatory mediators


Flavonoid Experimental model Refs.
with dosage regimen / other factors / functions

- inhibiting the ox-LDL-induced overexpression of IL-1β, IL-6,


TNF- α, E-selectin, ICAM1 and VCAM1 after 20 µM of vitexin
human umbilical vein
- inhibiting the production of ROS and MDA, and by promoting
Vitexin 1 µM - 50 µM endothelial cells [179]
the expression of SOD
(HUVECs)
- increasing the expression of p-AMPK and decreasing the expres-
sion of p-mTOR

- max. inhibitory effects on TNF-α level (26.7%) and IL-1β level


25 μg/ml, (59.1%) at the concentration of 100 μg/ml of vitexin
LPS - stimulated
Vitexin 50 μg/ml, RAW 264.7 murine - increasing the IL-10 level by 22.8% at 100 μg/ml of vitexin [180]
100 μg/ml macrophages - decreasing the NO level by 95.1% at 100 μg/ml of vitexin
- max. inhibitory effects on PGE2 by 75.2% at 25 μg/ml of vitexin

5 μM, 10 μM, - decreasing the IL-1β-induced production of PGE2 and NO (152.6


IL-1β-stimulated pg/mL and 72.8 Μmol/L, respectively) after 100 μM of vitexin
Vitexin 50 μM, 100 μM, chondrocytes from [183]
osteoarthritis patients - inhibiting the IL-1β-induced expressions of IL-6, TNF-α,
for 24 h MMP-1, MMP-3, MMP-13 after 10 μM and 50 μM of vitexin

quercetin and hesperidin [174]. A similar effect was shown by Ro- phils, neutrophils, mononuclear cells, and inhibited the release of
telli et al. [176]. several cytokines [186].
There is evidence that C-glycosylflavonoids, characteristic
7. ANTIOXIDANT POTENTIAL OF PASSIFLORA SPECIES
chemical compounds detected in extracts from various species of
Passiflora, such as vitexin [76, 77], isovitexin [74, 177], isoorientin It is well known that inflammatory processes with increasing
[149, 187], exerted some effects during in vitro and in vivo tests. levels of cytokines such as IL-6, TNF-α are strictly related to the
Odontuya et al., [149] observed, for example, that isoorientin inhib- oxidative stress because it was revealed that the higher production
ited the biosynthesis of thromboxane and leukotrienes. of reactive oxygen species (ROS) like superoxide anion leads to the
activation of the inflammatory pathway NF-kB and overproduction
Besides, Lin et al., [74] observed the anti-inflammatory activity
of pro-inflammatory cytokines [97, 188, 189]. Many studies re-
of isovitexin in vitro by interfering IKK phosphorylation, decreas-
vealed a relationship between inflammatory pathogenesis and oxi-
ing the NF-kB translocation from the cytoplasm to the nucleus.
dative stress, and it was shown that the overproduction of ROS
Another study showed that isovitexin diminished a few pathways,
could play a well-known role in the induction and acceleration of
such as MAPK, NF-κB, and Nrf2 [178]. However, the most inves-
inflammatory processes [189]. Recently, strong evidence showed
tigated from C-glycosylflavonoids is vitexin, which possesses anti-
that oxidative stress and ROS may occur during many inflammatory
inflammatory properties [179]. Vitexin at a dose of 10 mg/kg b.w.
diseases leading to injuries of macromolecules (proteins; lipids;
given to animals was able to decrease the expression of pro-
DNA; RNA), cells and tissues, i.e. during inflammatory bowel dis-
inflammatory cytokines (TNF-α, IL-1β, IL-6, and IL-33), which are
eases [97], type 2 diabetes [7], cardiovascular diseases [189],
responsible for hyperalgesia induced by the use of carrageenan
neuro-inflammation occurring in Alzheimer diseases [190], and
[76]. Also, in many studies, it was shown that vitexin significantly
Parkinson’s diseases [191] and many others.
inhibited the productions of the tumor necrosis factor , nitric ox-
ide, and myeloperoxidase in neutrophils [77, 175, 180], and in Recently, it was observed that extracts containing flavonoids
macrophages [180]. Vitexin (10 μM) also inhibited processes of from species of Passiflora exerted not only interesting anti-
neuroinflammation in experimental conditions after 24 h treatment inflammatory properties but also valuable antioxidant activity, i.e.
by diminishing the CDK5 expression and reduction of the release of P. subpeltata [41], P. edulis [61, 75, 89, 91, 92, 97, 192-194], P.
the proinflammatory mediators [181]. Application of vitexin in the alata [49, 73, 75], P. caerulea [49], P. incarnata [49, 56], P.
PC12 cell line showed a decline in the generation of free radicals leschenaultii [95], P. mucronata [99], P. maliformis [91], P. glan-
during oxidative stress and suppression of activation of the caspase- dulosa [195], P. ligularis [48], P. foetida [20].
3 pathway [182]. Next in vitro results showed that vitexin also in- There is strong scientific evidence that Passiflora extracts are
hibited other signaling pathways like p38, ERK1/2, and JNK, im- healthy food ingredients with antioxidant properties. Many tests for
portant in the induction of response of inflammation [181]. In cul- an estimation of this activity, using well-established in vitro meth-
tured chondrocytes from osteoarthritis patients, vitexin in various ods, mainly DPPH (2,2-diphenyl-1-picrylhydrazyl) radical assay
concentrations 24 h after its application decreased the level of nitric and ABTS (2,2'-azinobis-(3-ethylbenzothiazoline-6-sulfonic acid)
oxide, prostaglandin E2, expression of interleukins such as IL-6 and radical cation, ferric reducing/antioxidant power assay (FRAP), and
TNF-, and metalloproteinases [183]. A similar study using chon- key enzymes for inflammation, were carried out. Saravanan et al.,
drocytes revealed that vitexin also inhibited the pathway of nuclear [51] showed that the fresh pulp of fruits of P. subpeltata containing
factor kappa B and expression of cytokines of inflammation [184]. polyphenolic compounds exerted scavenging activities using DPPH
Lu et al., [185] observed that vitexin (10 mg/kg, i.p.) diminished with the value of IC50 = 5.667 μg/mL and ABTS+• (6794.96 μM
the edema of the lungs of mice, and mechanisms of its action in- Trolox equivalent/g sample). Another study by Saravanan et al.,
clude reduction of free radicals, level of IL-1β, activation of nuclear [48] revealed that the acetone extract from the leaves of P. subpel-
factor erythroid-2-related factor 2 and elevated activity of heme tata exhibited the highest DPPH (IC50 of 27.9 μg/ml) and ABTS
oxygenase. Moreover, it was shown that vitexin (0.2, 1, 1.5 mg/kg (10108.91 μM Trolox equivalent/g extract) scavenging activities.
bw p.o.) exerted beneficial effects in mice model of acute allergic The extract from leaves was more effective in comparison with an
asthma because this flavonoid reduced airway inflammation. In all extract from the pulp of fruits. These antioxidant activities may
doses, it diminished the infiltration of leukocytes such as eosino- justify anti-inflammatory activity observed in the animal model
Extracts and Flavonoids of Passiflora Species as Promising Anti-inflammatory Current Pharmaceutical Design, 2021, Vol. 27, No. 22 2599

studies carried out by authors. Furthermore, the fractionated extract CONFLICT OF INTEREST
from the leaves of P. leschenaultii also showed antiradical capacity The authors declare no conflict of interest, financial or other-
using the standard test in vitro, and it was observed that the pres- wise.
ence of phenolics, flavonoids, and tannins in extracts was responsi-
ble for this effects. Acetone extract caused maximum radical scav- ACKNOWLEDGEMENTS
enging activity using DPPH (IC50 = 29.14 μg/mL), ABTS Declared none.
(10509.69 μM TEAC/g extract), and FRAP (1511.74 mM Fe (II)
E/g extract) in comparison with petroleum ether, chloroform, REFERENCES
methanol, and hot water extracts [95]. The aqueous extract of P.
alata was investigated in an animal model of diabetes and analyses
[1] Zhong J, Shi G, Eds. Regulation of inflammation in chronic disea-
of results of the antioxidant study revealed that P. alata reduced the
se. Lausanne: Frontiers Media. 2019. doi: 10.3389/978-2-88945-
stable radical DPPH (56%), ABTS (3439 μM Trolox/g) and the 916-2
value of FRAP was 226.9 μM FeSO3/g), ORAC (302.1 μM [2] Leyva-López N, Gutierrez-Grijalva EP, Ambriz-Perez DL, Heredia
Trolox/g). Additionally, it was observed that P. alata administered JB. Flavonoids as cytokine modulators: a possible therapy for
to animals caused a higher level of glutathione, which is the en- inflammation-related diseases. Int J Mol Sci 2016; 17(6): 921.
dogenous enzyme responsible for the elimination of free radicals [3] Rubin DC, Shaker A, Levin MS. Chronic intestinal inflammation:
[73]. An evaluation of the antioxidant value carried out for hydro- inflammatory bowel disease and colitis-associated colon cancer.
alcoholic extract from the leaves of P. mucronata [99] using DPPH Front Immun 2012; 3: 107.
radicals showed a value EC50=96.05 μg/ml for extract in compari- [4] Luan YY, Yao YM. The clinical significance and potential role of
son with EC50=14.61 μg/ml for a fraction of ethyl acetate. Cazarin C-reactive protein in chronic inflammatory and neurodegenerative
et al., [97] observed that the aqueous extract of P. edulis exerted diseases. Front Immunol 2018; 9: 1302.
[5] Sommer C, Leinders M, Üçeyler N. Inflammation in the pathophy-
antioxidant activity in the serum of animals with colitis. Detailed
siology of neuropathic pain. Pain 2018; 159(3): 595-602.
results showed that the extract decreased peroxidation of lipids in [6] Tasneem S, Liu B, Li B, Choudhary MI, Wang W. Molecular
the colon, liver, and serum. However, it did not change the level of pharmacology of inflammation: Medicinal plants as anti-
reduced glutathione, activities of glutathione peroxidase and glu- inflammatory agents. Pharmacol Res 2019; 139: 126-40.
tathione reductase. It may be concluded that extract caused a bene- [7] de Candia P, Prattichizzo F, Garavelli S, et al. Type 2 diabetes:
ficial antioxidant effect together with decreasing the expression and How much of an autoimmune disease? Front Endocrinol 2019; 10:
level of pro-inflammatory cytokines. In another in vivo study, Pan- 451.
elli et al., [61] exerted that the extract from the bark of Passiflora [8] Klein A, Eliakim R. Nonsteroidal anti-inflammatory drugs and
edulis with carotenoids and phenolics was effective in improving inflammatory bowel disease. Pharmaceuticals 2010; 3: 1084-92.
antioxidant capacity in the plasma, kidney, liver and adipose tis- [9] Vonkeman HE, van de Laar MA. Nonsteroidal anti-inflammatory
sues. Lipid peroxidation and the level of malondialdehyde were drugs: adverse effects and their prevention. Semin Arthritis Rheum
also reduced. Other analyses showed that ethanol extracts from the 2010; 39(4): 294-312.
[10] Ordas I, Eckmann L, Talamini M, Baumgart DC, Sandborn WJ.
fruit peel of P. edulis administered to rats with diabetes increased
Ulcerative colitis. Lancet 2012; 380(9853): 1606-19.
the activity of superoxide dismutase and catalase and decreased the [11] Calabresi E, Petrelli F, Bonifacio AF, Puxeddu I, Alunno A. One
level of thiobarbituric acid reactive species in the heart, liver, and year in review 2018: pathogenesis of rheumatoid arthritis. Clin Exp
kidneys of animals [93]. Zeraik et al., [75] studied methanol ex- Rheumatol 2018; 36(2): 175-84.
tracts from the fruits of Passiflora edulis, pulp of Passiflora alata, [12] Baldassari LE, Fox RJ. Therapeutic advances and challenges in the
and rinds (epicarp and mesocarp) of P. edulis, using key enzymes in treatment of progressive multiple sclerosis. Drugs. 2018; 78(15):
inflammatory processes like myeloperoxidase of polymorphonu- 1549-66.
clear neutrophils. Extracts of the rind of P. edulis possessed strong [13] Zadeh AR, Ghadimi K, Ataei A, et al. Mechanism and adverse
antioxidant activity in vitro because this extract showed a 50% effects of multiple sclerosis drugs: a review article (Part 2). Int J
inhibitory effect at 0.1 mg mL-1 in comparison with the inhibitory Physiol Pathophysiol Pharmacol 2019; 11(4): 105-14.
activity of pulp extracts of P. edulis and P. alata (IC50 = 1.0 mg [14] Borg-Bartolo SP, Boyapati RK, Satsangi J, Kalla R. Precision
medicine in inflammatory bowel disease: concept, progress and
mL-1). However, isoorientin present in P. edulis, inhibited the activ-
challenges. F1000Res 2020; 9: F1000 Faculty Rev-54.
ity of myeloperoxidase with the value of IC50 = 4 µg mL -1. [15] Azab A, Nassar A, Azab AN. Anti-inflammatory activity of natural
products. Molecules 2016; 21(10): 1321.
CONCLUSION
[16] Shal B, Ding W, Ali H, Kim YS, Khan S. Anti-neuroinflammatory
In conclusion, this review provides evidence that various ex- potential of natural products in attenuation of Alzheimer’s disease.
tracts and flavonoids present in species of Passiflora spp., possess Front Pharmacol 2018; 9: 548.
anti-inflammatory and anti-oxidative activity in many models of [17] Ginwala R, Bhavsar R, Chigbu DGI, Jain P, Khan ZK. Potential
diseases tested in vitro. Moreover, the current studies indicate that role of flavonoids in treating chronic inflammatory diseases with a
these active substances obtained from various parts of Passiflora special focus on the anti-inflammatory activity of apigenin. Antio-
spp.and using various methods of extraction may differ in chemical xidants 2019; 8: 35.
composition, however, all tested substances showed pharmacologi- [18] Miroddi M, Calapai G, Navarra M, Minciullo PL, Gangemi S.
Passiflora incarnata L.: Ethnopharmacology,clinical application,
cal activity. In this context, based on the above presented results of
safety and evaluation of clinical trials. J Ethnopharmacol 2013;
many studies, it should be concluded that the extracts and flavon- 150: 791-804.
oids from Passiflora can be a valuable source of anti-inflammatory [19] Cerqueira-Silva CBM, Jesus ON, Santos ESL, Corrêa RX, Souza
and anti-oxidative medications for prevention and treatment of AP. Genetic breeding and diversity of the genus Passiflora: pro-
many diseases which occur with inflammation. However, these gress and perspectives in molecular and genetic studies. Int J Mol
challenges in drug discovery from Passiflora should be tested in Sci 2014; 15: 14122-52.
clinical trials. [20] Song Y, Wei XQ, Li MY, et al. Nutritional composition and antio-
xidant properties of the fruits of a Chinese wild Passiflora foetida.
CONSENT FOR PUBLICATION Molecules 2018; 23: 459.
Not applicable. [21] Ożarowski M, Thiem B. Progress in micropropagation of Passiflo-
ra spp. to produce medicinal plants: a mini-review. Rev Brasil
FUNDING Farmacog 2013; 23(6): 937-47.

None.
2600 Current Pharmaceutical Design, 2021, Vol. 27, No. 22 Ożarowski and Karpiński

[22] The Plant List. Passifloraceae. Available from: [43] Montanher AB, Zucolotto SM, Schenkel EP, Frode TS. Evidence
http://www.theplantlist.org/1.1/browse/A/Passifloraceae/ of anti-inflammatory effects of Passiflora edulis in an inflamma-
[23] Krosnick SE, Porter-Utley KE, Macdougal JM, Jorgensen PM, tion model. J Ethnopharmacol 2007; 109(2): 281-8.
Mcdade LA. New insights into the evolution of Passiflora subge- [44] Vargas AJ, Geremias DS, Provensi G, et al. Passiflora alata and
nus Decaloba (Passifloraceae): phylogenetic relationships and mor- Passiflora edulis spray-dried aqueous extracts inhibit inflammation
phological synapomorphies. System Bot 2013; 38(3): 692-713. in mouse model of pleurisy. Fitoterapia 2007; 78(2): 112-119.
[24] Silva EO, Milward-de-Azevedo MA, Sampaio Sá NA, de Sousa [45] Sasikala V, Saravanan S, Parimelazhagan T. Analgesic and anti-
DA, da Conceição GM. New records of Passiflora L. (Passiflora- inflammatory activities of Passiflora foetida L. Asian Pac J Trop
ceae) species from Maranhão state and northeastern Brazil. Check Med 2011; 4(8): 600-3.
List 2018; 14(2): 347-52. [46] Zucolotto SM, Fagundes C, Reginatto FH, et al. Analysis of C-
[25] EMA: Europen Medicine Agency. Assessment report on Passiflora glycosyl flavonoids from South American Passiflora species by
incarnata L., herba. Committee for Herbal Medicinal Products HPLC-DAD and HPLC-MS. Phytochem Anal 2011; 23: 232-9.
(HMPC). EMA/HMPC/669738/2013. 25 March 2014. [47] Xu F, Wang C, Yang L, et al. C-dideoxyhexosyl flavones from the
[26] Müller SD, Vasconcelos SB, Coelho M, Biavatti MW. LC and UV stems and leaves of Passiflora edulis Sims. Food Chem 2013; 136:
determination of flavonoids from Passiflora alata medicinal 94-9.
extracts and leaves. J Pharm Biomed Anal 2005; 37(2): 399-403. [48] Shanmugam S, Arunachalam K, Parimelazhagan T. Antioxidant,
[27] Thokchom R, Mandal G. Production Preference and importance of analgesic, anti-inflammatory and antipyretic effectsof polyphenols
Passion Fruit (Passiflora edulis): a review. J Agricult Engin Food from Passiflora subpeltata leaves- A promisingspecies of Passiflo-
Technol 2017; 4(1): 27-30. ra. Ind. Crops Prod. 2014; 54: 272-280.
[28] Faleiro FG, Junqueira NTV, Junghans TG, de Jesus ON, Miranda [49] Ożarowski M, Piasecka A, Paszel-Jaworska A, et al. Comparison
D, Otoni WC. Advances in passion fruit (Passiflora spp.) propaga- of bioactive compounds content in leaf extracts of Passiflora
tion. Rev Bras Frutic Jaboticabal 2019; 41(2): 1-17. incarnata, P. caerulea and P. alata and in vitro cytotoxic potential
[29] Correa RCG, Peralta RM, Haminiuk CWI, Maciel GM, Bracht A, on leukemia cell lines. Rev Brasil Farmacogn 2018; 28: 179-91.
Ferreira IC. The past decade findings related with nutritional com- [50] Ożarowski M, Pietrowiak A, Gryszczyńska A, et al. Comparison of
position, bioactive molecules and biotechnological applications of in vitro antioxidative activities of crude methanolic extracts of
Passiflora spp. (passion fruit). Trends Food Sci Technol 2016; 58: three species of Passiflora from greenhouse using DPPH, ABTS
79-95. and FRAP methods. Herba Pol 2019; 65(3):10-21.
[30] Industry Report. Passion flower extracts market size, share & [51] Shanmugam S, Thangaraj P, Trindade GG, et al. Protective effects
trends analysis report by product (organic, conventional), by appli- of flavonoid composition rich P. subpeltata Ortega. on indometha-
cation (pharmaceutical, nutraceutical, personal care, food and beve- cin induced experimental ulcerative colitis in rat models of
rages), and segment forecasts, 2019-2025. Report ID: GVR-3- inflammatory bowel diseases. J Ethnopharmacol 2020; 248:
68038-386-7. https://www.grandviewresearch.com/industry- 112350.
analysis/passion-flower-extracts-market [52] Ożarowski M, Mikołajczak P, Kujawski R, et al. Pharmacological
[31] Pan SY, Zhou SF, Gao SH, et al. New perspectives on how to effect of quercetin in hypertension and its potential application in
discover drugs from herbal medicines: CAM’s outstanding contri- pregnancy-induced hypertension. Review of in vitro, in vivo and
bution to modern therapeutics. Evid-Based Complem Altern Med clinical studies. Evid-Based Complem Altern Med 2018; 2018:
2013; 2013: ID 627375. 7421489.
[32] Atanasov AG, Waltenberger B, Pferschy-Wenzig EM, et al. Disco- [53] Cazarin CB, da Silva JK, Colomeu TC, et al. Passiflora edulis peel
very and resupply of pharmacologically active plant-derived natu- intake and ulcerative colitis: approaches for prevention and treat-
ral products: A review. Biotechnol Adv 2015; 33(8): 1582-614. ment. Exp Biol Med (Maywood) 2014; 239(5): 542-51.
[33] Katz L, Baltz RH. Natural product discovery: past, present, and [54] Cazarin CB, Rodriguez-Nogales A, Algieri F, et al. Intestinal anti-
future. J Ind Microbiol Biotechnol 2016; 43: 155-76. inflammatory effects of Passiflora edulis peel in the dextran
[34] Kole C, Ed. Wild crop relatives: genomic and breeding resources, sodium sulphate model of mouse colitis. J Funct Foods 2016; 26:
tropical and subtropical fruits. Springer-Verlag Berlin Heidelberg, 565-76.
2011. [55] Carmona-Hernandez JC, Taborda-Ocampo G, Valdez JC, Bolling
[35] Gaedcke F, Steinhoff B. Herbal medicinal products. Scientific and BW, González-Correa CH. Polyphenol extracts from three Colom-
regulatory basis for development, quality assurance and marketing bian Passifloras (passion fruits) prevent inflammation-induced bar-
authorization. CRC Press, Washington D.C. 2000. rier dysfunction of Caco-2 cells. Molecules 2019; 24(24): e4614.
[36] Domínguez-Rodríguez G, Marina ML, Plaza M. Strategies for the [56] Masteikowa R, Bernatoniene J, Bernatoniene R, Velziene S. Anti-
extraction and analysis of non-extractable polyphenols from plants. radical activities of the extract of Passiflora incarnata. Acta Pol
J Chromatogr A 2017; 1514: 1-15. Pharmaceut Drug Res 2008; 65(5): 577-83.
[37] De Santana FC, de Oliveira Torres LR, Shinagawa FB, et al. Opti- [57] Brown E, Hurd NS, McCall S, Ceremuga TE. Evaluation of the
mization of the antioxidant polyphenolic compounds extraction of anxiolytic effects of chrysin, a Passiflora incarnata extract, in the
yellow passion fruit seeds (Passiflora edulis Sims) by response sur- laboratory rat. AANA J 2007; 75(5): 333-7.
face methodology. J Food Sci Technol. 2017; 54: 3552-61. [58] Ingale AG, Hivrale AU. Pharmacological studies of Passiflora sp.
[38] Echeverry SM, Medina HI, Costa GM, Aragón DM. Optimization and their bioactive compounds. Afr J Plant Sci 2010; 4(10): 417-
of flavonoid extraction from Passiflora quadrangularis leaves with 26.
sedative activity and evaluation of its stability under stress condi- [59] Gavasheli N, Moniavo I, Eristavi L. Flavonoids from Passiflora
tions. Rev Brasil Farmacogn 2018; 28: 610-7. incarnata. Chem Nat Compounds 1974; 10(1): 99.
[39] Dhawan K, Dhawan S, Sharma A. Passiflora: a review update. J [60] Guseinov MD, Bobkova NV, Svistunov AA, et al. Flavonoids in
Ethnopharmacol 2004; 94(1): 1-23. Passiflora incarnata L. dry extract of Russian origin. Pharmacog J
[40] Da Silva J, Cazarin C, Colomeu T, et al. Antioxidant activity of 2019; 11(5): 1143-7.
aqueous extract of passion fruit (Passiflora edulis) leaves: in vitro [61] Panelli MF, Pierine DT, de Souza SLB, Ferron AJT, Garcia JL, dos
and in vivo study. Food Res Int 2013; 53: 882-90. Santos KC. Bark of Passiflora edulis treatment stimulates antioxi-
[41] Shanmugam S, Gomes IA, Denadai M, et al. UHPLC-QqQ- dant capacity, and reduces dyslipidemia and body fat in db/db
MS/MS identification, quantification of polyphenols from Passiflo- mice. Antioxidants 2018; 7: 120.
ra subpeltata fruit pulp and determination of nutritional, antioxi- [62] Chen CY, Peng WH, Tsai KD, Hsu SL. Luteolin suppresses
dant, α-amylase and α-glucosidase key enzymes inhibition proper- inflammation-associated gene expression by blocking NF-kappaB
ties. Food Res Int 2018; 108: 611-20. and AP-1 activation pathway in mouse alveolar macrophages. Life
[42] Beninca JP, Montanher AB, Zucolotto SM, Schenkel EP, Frode TS. Sci 2007; 81(23-24): 1602-14.
Evaluation of the anti-inflammatory efficacy of Passiflora edulis. [63] Park CM, Song YS. Luteolin and luteolin-7-O-glucoside inhibit
Food Chem 2007; 104: 1097-105. lipopolysaccharide-induced inflammatory responses through modu-
lation of NF-κB/AP-1/PI3K-Akt signaling cascades in RAW 264.7
cells. Nutr Res Pract 2013; 7(6): 423-9.
Extracts and Flavonoids of Passiflora Species as Promising Anti-inflammatory Current Pharmaceutical Design, 2021, Vol. 27, No. 22 2601

[64] Jia Z, Nallasamy P, Liu D, et al. Luteolin protects against vascular and opioidergic antinociceptive and behavioural mechanisms.
inflammation in mice and TNF-alpha-induced monocyte adhesion BMC Complement Altern Med 2016; 16: 77.
to endothelial cells via suppressing IΚBα/NF-κB signaling path- [85] de Lavor ÉM, Leal AEBP, Fernandes AWC, et al. Ethanolic extract
way. J Nutr Biochem 2015; 26(3): 293-302. of the aerial parts of Passiflora cincinnata Mast. (Passifloraceae)
[65] Aziz N, Kim MY, Cho JY. Anti-inflammatory effects of luteolin: A reduces nociceptive and inflammatory events in mice. Phytomedi-
review of in vitro, in vivo, and in silico studies. J Ethnopharmacol cine 2018; 47: 58-68.
2018; 225: 342-58. [86] Wasicky A, Hernandes LS, Vetore-Neto A, et al. Evaluation of
[66] Park CM, Song YS. Luteolin and luteolin-7-O-glucoside protect gastroprotective activity of Passiflora alata. Rev Bras Farmacogn
against acute liver injury through regulation of inflammatory 2015; 25(4): 407-12.
mediators and antioxidative enzymes in GalN/LPS-induced hepati- [87] Nerdy N, Ritarwan K. Hepatoprotective activity and nephroprotec-
tic ICR mice. Nutr Res Pract 2019; 13(6): 473-9. tive activity of peel extract from three varieties of the passion fruit
[67] Lodhi S, Vadnere GP, Patil KD, Patil TP. Protective effects of (Passiflora sp.) in the Albino rat. Maced J Med Sci 2019; 7(4):
luteolin on injury induced inflammation through reduction of tissue 536-42.
uric acid and pro-inflammatory cytokines in rats. J Tradit Complem [88] Tal Y, Anavi S, Reisman M, Samach A, Tirosh O, Troen AM. The
Med 2020; 10(1): 60-9. neuroprotective properties of a novel variety of passion fruit. J
[68] Zanoli P, Avallone R, Baraldi M. Behavioral characterisation of the Funct Foods 2016; 23: 359-69.
flavonoids apigenin and chrysin. Fitoterapia 2000; 71-81: S117-23. [89] Doungue HT, Kengne AP, Kuate D. Neuroprotective effect and
[69] Gazola AC, Costa GM, Castellanos L, et al. Involvement of antioxidant activity of Passiflora edulis fruit flavonoid fraction,
GABAergic pathway in the sedative activity of apigenin, the main aqueous extract, and juice in aluminum chloride-induced Alzhei-
flavonoid from Passiflora quadrangularis pericarp. Rev Brasil mer’s disease rats. Nutrire 2018; 43: 23.
Farmacogn 2015; 25: 158-63. [90] Calderon A, Salas J, Dapello G, et al. Assessment of antibacterial
[70] Foudah AI, Alam P, Kamal YT, et al. Development and validation and antifungal properties and in vivo cytotoxicity of Peruvian Pas-
of a high-performance thin-layer chromatographic method for the siflora mollisima. J Contemp Dent Pract 2019; 20(2): 145-51.
quantitative analysis of vitexin in Passiflora foetida herbal formu- [91] Ramaiya SD, Bujang JS, Zakaria MH. Assessment of total pheno-
lations. Saudi Pharmac J 2019; 27: 1157-63. lic, antioxidant, and antibacterial activities of Passiflora species.
[71] Pereira CA, Yariwake JH, Lanças FM, Wauters JN, Tits M, Ange- Sci World J 2014; 2014: ID 167309.
not L. A HPTLC densitometric determination of flavonoids from [92] da Silva JK, Cazarin CBB, Batista AG, Marostica M. Effects of
Passiflora alata, P. edulis, P. incarnata and P. caerulea and com- passion fruit (Passiflora edulis) byproduct intake in antioxidant sta-
parison with HPLC method. Phytochem Anal 2004; 15(4): 241-8. tus of Wistar rats tissues. LWT Food Sci Technol 2014; 59: 1213-
[72] Anzoise ML, Marrassini C, Bach H, Gorzalczany S. Beneficial 9.
properties of Passiflora caerulea on experimental colitis. J Ethno- [93] Kandandapani S, Balaraman AK, Ahamed HN. Extracts of passion
pharmacol 2016; 194: 137-45. fruit peel and seed of Passiflora edulis (Passifloraceae) attenuate
[73] Colomeu TC, Figueiredo D, Cazarin CB, et al. Antioxidant and oxidative stress in diabetic rats. Chinese J Nat Med 2015; 13(9):
anti-diabetic potential of Passiflora alata Curtis aqueous leaves 680-6.
extract in type 1 diabetes mellitus (NOD-mice). Int Immunophar- [94] García-Ruiz A, Girones-Vilaplana A, León P, et al. Banana passion
macol 2014; 18: 106-15. fruit (Passiflora mollissima (Kunth) L.H. Bailey): microencapsula-
[74] Lin CM, Huang ST, Liang YC, et al. Isovitexin suppresses lipo- tion, phytochemical composition and antioxidant capacity. Molecu-
polysaccharide-mediated inducible nitric oxide synthase through les 2017; 22: 85.
inhibition of NF-kappa B in mouse macrophages. Planta Med 2005; [95] Shanmugam S, Murugaiyan I, Lima BS, et al. HPLC-DAD-MS
71(8): 748-53. identification of polyphenols from Passiflora leschenaultii and
[75] Zeraik ML, Serteyn D, Deby-Dupont G, et al. Evaluation of the determination of their antioxidant, analgesic, anti-inflammatory and
antioxidant activity of passion fruit (Passiflora edulis and Passiflo- antipyretic properties. Arab J Chem 2019; 12: 760-71.
ra alata) extracts on stimulated neutrophils and myeloperoxidase [96] Aguillón-Osma J, Luzardo-Ocampo I, Cuellar-Nuñez ML, Maldo-
activity assays. Food Chem 2011; 128(2): 259-65. nado-Celis ME, Loango-Chamorro N, Campos-Vega R. Impact of
[76] Borghi SM, Carvalho TT, Staurengo-Ferrari L, et al. Vitexin inhi- in vitro gastrointestinal digestion on the bioaccessibility and antio-
bits inflammatory pain in mice by targeting TRPV1, oxidative xidant capacity of bioactive compounds from Passion fruit (Passi-
stress, and cytokines. J Nat Prod 2013; 76(6): 1141-9. flora edulis) leaves and juice extracts. J Food Biochem 2019;
[77] Bahareh AN, Farid H, Mohtaram A, Marjan NA. Anti- 43(7): e12879.
inflammatory effects of quercetin and vitexin on activated human [97] Cazarin CB, da Silva JK, Colomeu TC, et al. Intake of Passiflora
peripheral blood neutrophils. J Pharmacopuncture 2017; 20: 127- edulis leaf extract improves antioxidant and anti-inflammatory sta-
31. tus in rats with 2,4,6-trinitrobenzenesulphonic acid induced colitis.
[78] Zhang P, Jiao H, Wang C, Lin Y, You S. Chlorogenic acid amelio- J Funct Foods 2015; 17: 575-86.
rates colitis and alters colonic microbiota in a mouse model of dex- [98] Figueiredo D, Colomeu TC, Schumacher NSG, et al. Aqueous leaf
tran sulfate sodium-induced colitis. Front Physiol 2019; 10: 325. extract of Passiflora alata Curtis promotes antioxidant andanti-
[79] Adamczak A, Ożarowski M, Karpiński TM. Antibacterial activity inflammatory effects and consequently preservation of NOD mice-
of some flavonoids and organic acids widely distributed in plants. J beta cells (non-obese diabetic). Int Immunopharmacol 2016; 35:
Clin Med 2020; 9(109); 1-17. 127-36.
[80] Ayres AS, de Araújo LL, Soares TC, et al. Comparative central [99] de Araujo MH, da Silva IC, de Oliveira PF, et al. Biological activi-
effects of the aqueous leaf extract of two populations of Passiflora ties and phytochemical profile of Passiflora mucronata from the
edulis. Rev Brasil Farmacogn 2015; 25: 499-505. Brazilian resting. Rev Bras Farmacogn 2017; 27: 702-10.
[81] Jawna-Zboińska K, Blecharz-Klin K, Joniec-Maciejak I, et al. [100] Lima GC, Vuolo MM, Batista AG, Dragano NRV, Solon C,
Passiflora incarnata L. improves spatial memory, reduces stress, Marostica MR Jr. Passifloraedulis peel intake improves insulin
and affects neurotransmission in rats. Phytother Res 2016; 30(5): sensitivity, increasing incretins and hypothalamic satietogenic neu-
781-9. ropeptide in rats on a high-fat diet. Nutrition 2016; 32: 863-70.
[82] Anusooriya P, Malarvizhi D, Gopalakrishnan VK, Devaki K. [101] Aslanargun P, Cuvas O, Dikmen B, Aslan E, Yuksel MU. Passiflo-
Antioxidant and antidiabetic effect of aqueous fruit extract of Pas- ra incarnata Linneaus as an anxiolytic before spinal anaesthesia. J
siflora ligularis Juss. on streptozotocin induced diabetic rats. Int Anesth 2012; 26: 39-44.
Sch Res Notices 2014; 2014: 130342. [102] Dantas LP, de Oliveira-Ribeiro A, de Almeida-Souza LM, Groppo
[83] Salles BCC, da Silva MA, Taniguthi L, et al. Passiflora edulis leaf FC. Effects of Passiflora incarnata and midazolam for control of
extract: evidence of antidiabetic and antiplatelet effects in rats. Biol anxiety in patients undergoing dental extraction. Med Oral Patol
Pharm Bull 2020; 43(1): 169-74. Oral Cir Bucal 2017; 22(1): e95-101.
[84] Aman U, Subhan F, Shahid M, et al. Passiflora incarnata attenua- [103] Rokhtabnak F, Ghodraty MR, Kholdebarin A, et al. Comparing the
tion of neuropathic allodynia and vulvodynia apropos GABA-ergic effect of preoperative administration of melatonin and Passiflora
2602 Current Pharmaceutical Design, 2021, Vol. 27, No. 22 Ożarowski and Karpiński

incarnata on postoperative cognitive disorders in adult patients TLR4 and NLRP3 signaling pathways. Oncotarget 2016; 7(50):
undergoing elective surgery. Anesth Pain Med 2016; 7(1): e41238. 82602-8.
[104] Lee J, Jung HY, Lee SI, Choi JH, Kim SG. Effects of Passiflora [125] Chao CS, Tsai CS, Chang YP, Chen JM, Chin HK, Yang SC.
incarnata Linnaeus on polysomnographic sleep parameters in sub- Hyperin inhibits nuclear factor kappa B and activates nuclear factor
jects with insomnia disorder: a double-blind randomized placebo- E2-related factor-2 signaling pathways in cisplatin-induced acute
controlled study. Int Clin Psychopharmacol 2020; 35(1): 29-35. kidney injury in mice. Int Immunopharmacol 2016; 40: 517-23.
[105] Ngan A, Conduit R. A double-blind, placebo-controlled investiga- [126] Figueiredo DA, Pordeus LC, Paulo LL, et al. Effects of bark flour
tion of the effects of Passiflora incarnata (passionflower) herbal of Passiflora edulis on food intake, body weight and behavioral
tea on subjective sleep quality. Phytother Res 2011; 25(8): 1153-9. response of rats. Rev Brasil Farmacogn 2016; 26: 595-600.
[106] Gibbert J, Kreimendahl F, Lebert J, Rychlik R, Trompetter I. [127] Sergent T, Piront N, Meurice J, Toussaint O, Schneider Y-J. Anti-
Improvement of stress resistance and quality of life of adults with inflammatory effects of dietary phenolic compounds in an in vitro
nervous restlessness after treatment with a passion flower dry model of inflamed human intestinal epithelium. Chem-Biol Interact
extract. Complement Med Res 2017; 24: 83-9. 2010; 188: 659-67.
[107] da Silva JA, Costa MJC, Gonçalves MCR, Braga JEF, Lima CM, [128] Martin DA, Bolling BW. A review of the efficacy of dietary poly-
Pordeus LCM. Effects of the single supplementation and multiple phenols in experimental models of inflammatory bowel diseases.
doses of Passiflora incarnata L. on human anxiety: aclinical trial, Food Funct 2015; 6(6): 1773-86.
double-blind, placebo-controlled, randomized. Int Arch Med 2017; [129] Kaulmann A, Bohn T. Bioactivity of polyphenols: preventive and
10(6):1-9. adjuvant strategies toward reducing inflammatory bowel diseases-
[108] Farid R, Rezaieyazdi Z, Mirfeizi Z, et al. Oral intake of purple promises, perspectives, and pitfalls. Oxid Med Cell Longev 2016;
passion fruit peel extract reduces pain and stiffness and improves 2016: 9346470.
physical function in adult patients with knee osteoarthritis. Nutr [130] Santino A, Scarano A, De Santis S, De Benedictis M, Giovinazzo
Res 2010;30(9): 601-6. G, Chieppa M. Gut microbiota modulation and anti-inflammatory
[109] de Queiroz Mdo S, Janebro DI, da Cunha MA, et al. Effect of the properties of dietary polyphenols in IBD: new and consolidated
yellow passion fruit peel flour (Passiflora edulis f. flavicarpa deg.) perspectives. Curr Pharm Des 2017; 23(16): 2344-51.
in insulin sensitivity in type 2 diabetes mellitus patients. Nutr J [131] Nunes S, Danesi F, Del Rio D, Silva P. Resveratrol and inflamma-
2012; 11: 89. tory bowel disease: the evidence so far. Nutr Res Rev 2018; 31(1):
[110] Market Research Report: Passion flower extracts market size & 85-97.
trends, Industry Report, 2019-2025. Grand View Research, Inc. [132] Valdez JC, Bolling BW. Anthocyanins and intestinal barrier func-
San Francisco, United States. Available from: tion: A review. JFB 2019; 31: 18-30.
https://www.grandviewresearch.com/industry-analysis/passion- [133] Grotewold E. The science of flavonoids. Springer Science + Busi-
flower-extracts-market ness Media, LLC. New York, 2008.
[111] Gil-Cardoso K, Ginés I, Pinent M, Ardévol A, Blay M, Terra X. [134] Brodowska K. Natural flavonoids: classification, potential role, and
Effects of flavonoids on intestinal inflammation, barrier integrity application of flavonoid analogues. Eur J Biol Res 2017; 7(2): 108-
and changes in gut microbiota during diet-induced obesity. Nutr 23.
Res Rev 2016; 29(2): 234-48. [135] Lee HN, Shin SA, Choo GS, et al. Anti-inflammatory effect of
[112] Costa SL, Silva VD, Dos Santos-Souza C, et al. Impact of plant- quercetin and galangin in LPS-stimulated RAW264.7 macrophages
derived flavonoids on neurodegenerative diseases. Neurotox Res and DNCB-induced atopic dermatitis animal models. Int J Mol
2016; 30(1): 41-52. Med 2018; 41(2): 888-98.
[113] Maher P. The potential of flavonoids for the treatment of neurode- [136] Sanchez M, Lodi F, Vera R, et al. Quercetin and isorhamnetin
generative diseases. Int J Mol Sci 2019; 20: 3056. prevent endothelial dysfunction, superoxide production, and ove-
[114] Ayaz M, Sadiq A, Junaid M, et al. Flavonoids as prospective neu- rexpression of p47phox induced by angiotensin II in rat aorta. J
roprotectants and their therapeutic propensity in aging associated Nutr 2007; 137(4): 910-5.
neurological disorders. Front Aging Neurosci 2019; 11: 155. [137] Romero M, Jiménez R, Sánchez M, et al. Quercetin inhibits vascu-
[115] Di Paolo M, Papi L, Gori F, Turillazzi E. Natural products in neu- lar superoxide production induced by endothelin-1: role of NADPH
rodegenerative diseases: a great promise but an ethical challenge. oxidase, uncoupled eNOS and PKC. Atherosclerosis 2009; 202(1):
Int J Mol Sci 2019; 20: 5170. 58-67.
[116] Kaser A, Zeissig S, Blumberg RS. Inflammatory bowel disease. [138] Endale M, Park S-C, Kim S, et al. Quercetin disrupts tyrosine-
Annu Rev Immunol 2010; 28: 573-621. phosphorylated phosphatidylinositol 3-kinase and myeloid diffe-
[117] Dotan I, Allez M, Danese S, Keir M, Tole S, McBride J. The role rentiation factor-88 association, and inhibits MAPK/AP-1 and
of integrins in the pathogenesis of inflammatory bowel disease: IKK/NF-B-induced inflammatory mediators production in
Approved and investigational anti‐integrin therapies. Med Res Res RAW264.7 cells. Immunobiology 2013; 218(12): 1452-67.
2020; 40(1): 245-62. [139] Yang D, Liu X, Liu M, Chi H, Liu J, Han H. Protective effects of
[118] Jang SE, Jeong JJ, Kim JK, Han MJ, Kim DH. Simultaneous ame- quercetin and taraxasterol against H2O2-induced human umbilical
lioratation of colitis and liver injury in mice by Bifdobacterium vein endothelial cell injury in vitro. Exp Ther Med 2015; 10(4):
longum LC67 and Lactobacillus plantarum LC27. Sci Rep 2018; 8: 1253-60.
7500. [140] Overman A, Chuang C-C, McIntosh M. Quercetin attenuates
[119] Singh AK, Cabral C, Kumar R, et al. Beneficial effects of dietary inflammation in human macrophages and adipocytes exposed to
polyphenols on gut microbiota and strategies to improve delivery macrophage-conditioned media, Int J Obesity 2011; 35(9): 1165-
efficiency. Nutrients 2019; 11: 2216. 72.
[120] Vargas-Robles H, Castro-Ochoa KF, Citalán-Madrid AF, Schnoor [141] Lee KM, Hwang MK, Lee DE, Lee KW, Lee HJ. Protective effect
M. Beneficial effects of nutritional supplements on intestinal epi- of quercetin against arsenite-induced COX-2 expression by targe-
thelial barrier functions in experimental colitis models in vivo. ting PI3K in rat liver epithelial cells. J Agricult Food Chem 2010;
World J Gastroenterol 2019; 25(30): 4181-98. 58(9): 5815-20.
[121] Manthey JA. Biological properties of flavonoids pertaining to [142] Kim HP, Mani I, Iversen L, Ziboh VA. Effects of naturally-
inflammation. Microcirculation 2000; 7(6 Pt 2): S29-34. occurring flavonoids and biflavonoids on epidermal cyclooxygena-
[122] Lee S, Jung SH, Lee YS, et al. Antiinflammatory activity of hype- se and lipoxygenase from guinea-pigs. Prostaglandins Leukot
rin from Acanthopanax chiisanensis roots. Arch Pharm Res 2004; Essent Fatty Acids 1998; 58(1): 17-24.
27(6): 628-32. [143] Li D, Ma J, Wang L, Xin S. Apigenin prevent abdominal aortic
[123] Hämäläinen M, Nieminen R, Asmawi MZ, Vuorela P, Vapaatalo aneurysms formation by inhibiting the NF-κB signaling pathway. J
H, Moilanen E. Effects of flavonoids on prostaglandin E2 produc- Cardiovasc Pharmacol 2019. doi: 10.1097/FJC.0000000000000785
tion and on COX-2 and mPGES-1 expressions in activated macro- [144] Lim H, Hyun PK. Inhibition of mammalian collagenase, matrix
phages. Planta Med 2011; 77(13): 1504-11. metalloproteinase-1, by naturally-occurring flavonoids. Planta Med
[124] Chunzhi G, Zunfeng L, Chengwei Q, Xiangmei B, Jingui Y. Hype- 2007; 73(12): 1267-74.
rin protects against LPS-induced acute kidney injury by inhibiting
Extracts and Flavonoids of Passiflora Species as Promising Anti-inflammatory Current Pharmaceutical Design, 2021, Vol. 27, No. 22 2603

[145] Woo KJ, Jeong YJ, Park-kwon TK. Chrysin induced apoptosis is [164] Karamese M, Erol HS, Albayrak M, Findik-Guvendi G, Aydin E,
mediated through caspase activation and akt inactivation in U937 Aksak-Karamese S. Anti-oxidant and anti-inflammatory effects of
leukemia cell. Biochem Biophys Res Commun 2004; 325-334: apigenin in a rat model of sepsis: an immunological, biochemical,
1215-22. and histopathological study. Immunopharmacol Immunotoxicol
[146] Lee BK, Lee WJ, Jung YS. Chrysin attenuates VCAM-1 expression 2016; 38(3): 228-37.
and monocyte adhesion in lipopolysaccharide-stimulated brain [165] Dang Y, Li Z, Wei Q, Zhang R, Xue H, Zhang Y. Protective effect
endothelial cells by preventing NF-κB signaling. Int J Mol Sci of apigenin on acrylonitrile-induced inflammation and apoptosis in
2017; 18(7): e1424. testicular cells via the NF-κB pathway in rats. Inflammation 2018;
[147] Chylińska-Wrzos P, Lis-Sochocka M, Jodłowska-Jędrych B. Chry- 41(4): 1448-59.
sin and its potential antineoplastic effect. Eur J Biol Res 2017; 7(3): [166] Zhang J, Zhao X, Zhu H, Wang J, Ma J, Gu M. Apigenin protects
245-54. against renal tubular epithelial cell injury and oxidative stress by
[148] Zhao S, Liang M, Wang Y, et al. Chrysin suppresses vascular high glucose via regulation of NF-E2-related factor 2 (Nrf2) path-
endothelial inflammation via inhibiting the NF-κB signaling path- way. Med Sci Monit 2019; 25: 5280-8.
way. J Cardiovasc Pharmacol Ther 2019: 24(3): 278-87. [167] Zhou Q, Cheng KW, Gong J, Li ETS, Wang M. Apigenin and its
[149] Odontuya G, Hoult JR, Houghton PJ. Structure-activity relationship methylglyoxal-adduct inhibit advanced glycation end products-
for anti-inflammatory effect of luteolin and its derived glycosides. induced oxidative stress and inflammation in endothelial cells. Bio-
Phytother Res 2005; 19: 782-6. chem Pharmacol 2019; 166: 231-41.
[150] Yamagata K, Hashiguchi K, Yamamoto H, Tagami M. Dietary [168] Ai XY, Qin Y, Liu HJ, et al. Apigenin inhibits colonic inflamma-
apigenin reduces induction of LOX-1 and expression, leukocyte tion and tumorigenesis by suppressing STAT3-NF-κB signaling.
adhesion, and acetylated low-density lipoprotein uptake in human Oncotarget 2017; 8(59): 100216-26.
endothelial cells exposed to trimethylamine-n-oxide. J Cardiovasc [169] Zhao F, Dang Y, Zhang R, et al. Apigenin attenuates acrylonitrile-
Pharmacol 2019; 74(6): 558-565. induced neuro-inflammation in rats: Involved of inactivation of the
[151] Chen P, Huo X, Liu W, Li K, Sun Z, Tian J. Apigenin exhibits anti- TLR4/NF-κB signaling pathway. Int Immunopharmacol 2019; 75:
inflammatory effects in LPS-stimulated BV2 microglia through 105697.
activating GSK3β/Nrf2 signaling pathway. Immunopharmacol [170] Okla M, Al-Madani JO, Chung S, Alfayez M. Apigenin reverses
Immunotoxicol 2020; 42(1): 9-16. interleukin-1β-induced suppression of adipocyte browning via
[152] Zhang P, Mak JC, Man RY, Leung SW. Flavonoids reduces lipo- COX2/PGE2 signaling pathway in human adipocytes. Mol Nutr
polysaccharide-induced release of inflammatory mediators in Food Res 2020; 64(1): e1900925.
human bronchial epithelial cells: Structure-activity relationship. [171] Ali F, Rahul, Naz F, Jyoti S, Siddique YH. Health functionality of
Eur J Pharmacol 2019; 865: 172731. apigenin: A review. Int J Food Propert 2017; 20(6): 1197-238,
[153] Patil RH, Babu RL, Naveen Kumar M, et al. Anti-inflammatory [172] Zeng P, Liu B, Wang Q, et al. Apigenin attenuates atherogenesis
effect of apigenin on LPS-induced pro-inflammatory mediators and through inducing macrophage apoptosis via inhibition of AKT
AP-1 factors in human lung epithelial cells. Inflammation 2016; Ser473 phosphorylation and downregulation of plasminogen acti-
39(1): 138-147. vator inhibitor-2. Oxid Med Cell Longev 2015; 2015: 379538.
[154] Zhang X, Wang G, Gurley EC, Zhou H. Flavonoid apigenin inhi- [173] Calixto JB, Beirith A, Ferreira J, Santos AR, Cechinel Filho V,
bits lipopolysaccharide-induced inflammatory response through Yunes RA. Naturally occurring antinociceptive substances from
multiple mechanisms in macrophages. PLoS One 2014; 9(9): plants. Phytother Res 2000; 14: 401-18.
e107072. [174] Guardia T, Rotelli AE, Juarez AO, Pelzer LE. Anti-inflammatory
[155] Makanjuola SBL, Ogundaini AO, Ajonuma LC, Dosunmu A. Api- properties of plant flavonoids. Effects of rutin, quercetin and hespe-
genin and apigeninidin isolates from the Sorghum bicolor leaf tar- ridin on adjuvant arthritis in rat. Farmaco 2001; 56(9): 683-7.
gets inflammation via cyclo-oxygenase-2 and prostaglandin-E2 [175] Nikfarjam BA, Adineh M, Hajiali F, Nassiri-Asl M. Treatment
blockade. Int J Rheum Dis 2018; 21(8):1487-1495. with rutin - a therapeutic strategy for neutrophil-mediated inflam-
[156] Balez R, Steiner N, Engel M, et al Neuroprotective effects of api- matory and autoimmune diseases: anti-inflammatory effects of
genin against inflammation, neuronal excitability and apoptosis in rutin on neutrophils. J Pharmacopuncture 2017; 20(1): 52-6.
an induced pluripotent stem cell model of Alzheimer's disease. Sci [176] Rotelli AE, Guardia T, Juárez AO, de la Rocha NE, Pelzer LE.
Rep 2016; 6: 31450. Comparative study of flavonoids in experimental models of
[157] Pang L, Zou S, Shi Y, Mao Q, Chen Y. Apigenin attenuates inflammation. Pharmacol Res 2003; 48(6): 601-6.
PM2.5-induced airway hyperresponsiveness and inflammation by [177] Lv H, Yu Z, Zheng Y, et al. Isovitexin exerts anti-inflammatory
down-regulating NF-κB in murine model of asthma. Int J Clin Exp and anti-oxidant activities on lipopolysaccharide-induced acute
Pathol 2019; 12(10): 3700-9. lung injury by inhibiting MAPK and NF-κB and activating HO-
[158] Darabi P, Khazali H, Natanzi MM. Therapeutic potentials of the 1/Nrf2 pathways. Int J Biol Sci 2016; 12(1): 72-86.
natural plant flavonoid apigenin in polycystic ovary syndrome in [178] He M, Min JW, Kong WL, He XH, Li JX, Peng BW. A review on
rat model: via modulation of pro-inflammatory cytokines and the pharmacological effects of vitexin and isovitexin. Fitoterapia
antioxidant activity. Gynecol Endocrinol 2019; doi: 2016; 115: 74-85.
10.1080/09513590.2019.1706084. [179] Zhang S, Guo C, Chen Z, Zhang P, Li J, Li Y. Vitexin alleviates
[159] Gentile D, Fornai M, Colucci R, et al. The flavonoid compound ox-LDL-mediated endothelial injury by inducing autophagy via
apigenin prevents colonic inflammation and motor dysfunctions AMPK signaling activation. Mol Immunol 2017; 85: 214-21.
associated with high fat diet-induced obesity. PLoS One 2018; [180] Rosa SI, Rios-Santos F, Balogun SO, Martins DT. Vitexin reduces
13(4): e0195502. neutrophil migration to inflammatory focus by down-regulating
[160] Anusha C, Sumathi T, Joseph LD. Protective role of apigenin on pro-inflammatory mediators via inhibition of p38, ERK1/2 and
rotenone induced rat model of Parkinson's disease: Suppression of JNK pathway. Phytomedicine 2016; 23(1): 9-17.
neuroinflammation and oxidative stress mediated apoptosis. Chem [181] Krishnan M, Kang SC. Vitexin inhibits acrylamide-induced neu-
Biol Interact 2017; 269: 67-79. roinflammation and improves behavioral changes in zebrafish lar-
[161] Zhou RJ, Ye H, Wang F, Wang JL, Xie ML. Apigenin inhibits d- vae. Neurotoxicol Teratol 2019; 74: 106811.
galactosamine/LPS-induced liver injury through upregulation of [182] Chen L, Zhang B, Shan S, Zhao X. Neuroprotective effects of
hepatic Nrf-2 and PPARγ expressions in mice. Biochem Biophys vitexin against isoflurane-induced neurotoxicity by targeting the
Res Commun 2017; 493(1): 625-30. TRPV1 and NR2B signaling pathways. Mol Med Rep 2016; 14(6):
[162] Bougioukas I, Didilis V, Emmert A, et al. Apigenin Rreduces NF- 5607-13.
κB and subsequent cytokine production as protective effect in a [183] Yang H, Huang J, Mao Y, Wang L, Li R, Ha C. Vitexin alleviates
rodent animal model of lung ischemia-reperfusion injury. J Invest interleukin-1β-induced inflammatory responses in chondrocytes
Surg 2018; 31(2): 96-106. from osteoarthritis patients: Involvement of HIF-1α pathway.
[163] Chang X, He H, Zhu L, et al. Protective effect of apigenin on Scand J Immunol 2019; 90(2): e12773.
Freund's complete adjuvant-induced arthritis in rats via inhibiting [184] Xie CL, Li JL, Xue EX, et al. Vitexin alleviates ER-stress-activated
P2X7/NF-κB pathway. Chem Biol Interact 2015; 236: 41-6. apoptosis and the related inflammation in chondrocytes and inhibits
2604 Current Pharmaceutical Design, 2021, Vol. 27, No. 22 Ożarowski and Karpiński

the degeneration of cartilage in rats. Food Funct 2018; 9(11): 5740- [191] Adams B, Nunes JM, Page MJ, et al. Parkinson’s disease: a syste-
9. mic inflammatory disease accompanied by bacterial inflammagens.
[185] Lu Y, Yu T, Liu J, Gu L. Vitexin attenuates lipopolysaccharide- Front Aging Neurosci 2019; 11: 210.
induced acute lung injury by controlling the Nrf2 pathway. PLoS [192] Zeraik ML, Yariwake JH, Wauters J-N, Tits M, Angenot L. Analy-
One 2018; 13(4): e0196405. sis of passion fruit rinds (Passiflora edulis): Isoorientin quantifica-
[186] Venturini CL, Macho A, Arunachalam K, et al. Vitexin inhibits tion by HPTLC and evaluation of antioxidant (radical scavenging)
inflammation in murine ovalbumin-induced allergic asthma. Bio- capacity. Química Nova 2012; 35: 541-5.
med Pharmacother 2018; 97: 143-51. [193] Lourith N, Kanlayvattanakul M. Antioxidant activities and pheno-
[187] Küpeli E, Aslan M, Gürbüz I, Yesilada E. Evaluation of in vivo lics of Passiflora edulis seed recovered from juice production resi-
biological activity profile of isoorientin. Z Naturforsch C J Biosci due. J Oleo Sci 2013; 62(4): 235-40.
2004; 59(11-12): 787-90. [194] da Silva Morrone M, de Assis AM, da Rocha RF, et al. Passiflora
[188] Morgan MJ, Liu ZG. Crosstalk of reactive oxygen species and NF- manicata (Juss.) aqueous leaf extract protects against reactive oxy-
κB signaling. Cell Res 2011; 21(1): 103-15. gen species and protein glycation in vitro and ex vivo models. Food
[189] Hussain F, Mittal S, Joshee N, Parajuli P. Application of bioactive Chem Toxicol 2013; 60: 45-51.
compounds from Scutellaria in neurologic disorders. Adv Neuro- [195] Lima-Neto ABM, Marques MM, Mendes FN, Vieira IG, Diniz DB,
biol 2016; 12: 79-94. Guedes MI. Antioxidant activity and physicochemical analysis of
[190] Gross AL, Walker KA, Moghekar AR, et al. Plasma markers of passion fruit (Passiflora glandulosa Cav.) pulp native to Cariri
inflammation linked to clinical progression and decline during pre- region. Acta Sci Biol Sci 2017; 39(4): 417- 22.
clinical AD. Front Aging Neurosci 2019; 11: 229.

You might also like