Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 6941 word(s) 6941 2022-03-22 02:38:34 |
2 format correction -6 word(s) 6935 2022-03-23 02:46:14 | |
3 format correction Meta information modification 6935 2022-03-24 06:56:08 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Silva, L. Zimbro. Encyclopedia. Available online: https://encyclopedia.pub/entry/20855 (accessed on 28 April 2024).
Silva L. Zimbro. Encyclopedia. Available at: https://encyclopedia.pub/entry/20855. Accessed April 28, 2024.
Silva, Luís. "Zimbro" Encyclopedia, https://encyclopedia.pub/entry/20855 (accessed April 28, 2024).
Silva, L. (2022, March 22). Zimbro. In Encyclopedia. https://encyclopedia.pub/entry/20855
Silva, Luís. "Zimbro." Encyclopedia. Web. 22 March, 2022.
Zimbro
Edit

Zimbro or common juniper (Juniperus communis) is traditionally used to treat renal suppression, acute and chronic cystitis, bladder catarrh, albuminuria, leucorrhea, and amenorrhea. These uses are mainly attributed to its bioactive composition, which is very rich in phenolics, terpenoids, organic acids, alkaloids, and volatile compounds. In the last few years, several studies have analyzed the huge potential of this evergreen shrub, describing a wide range of activities with relevance in different biomedical discipline areas, namely antimicrobial potential against human pathogens and foodborne microorganisms, notorious antioxidant and anti-inflammatory activities, antidiabetic, antihypercholesterolemic and antihyperlipidemic effects, and neuroprotective action, as well as antiproliferative ability against cancer cells and the ability to activate inductive hepato-, renal- and gastroprotective mechanisms.

bioactive compounds phenolic compounds essential oils biological potential in vitro studies in vivo studies Juniperus communis L.

1. Introduction

Natural products have an important role in the research and development of new drugs. People have always extracted natural products from several natural sources, such as marine organisms, microorganisms, animals, and medicinal plants [1]. The main extracts from natural products come from medicinal plants. Plant-derived products and compounds have been used worldwide since ancient times in folk medicine as remedies for several diseases, such as tinctures, teas, poultices, maintaining high prevalence in public health [1][2][3][4]. Advances in clinical research and quality control have shown a greater value of herbal medicine in the treatment and overcoming of many diseases. Recent works report promising potential regarding the use of plants in the treatment and/or prevention of several hard-to-cure diseases, such as atherosclerosis [5][6], cancer [1][2][3][7][8], cardiovascular diseases [9][10][11][12], diabetes [8][13][14], and neurological disorders [4][15][16], among others.
The genus Juniperus includes roughly 68 species and 36 varieties and belongs to the Cupressaceae family [17]. The plant Juniperus communis L., named “zimbro” in Portugal, is a shrub or small evergreen tree; a perennial and long-lived coniferous, woody pioneer and colonizing plant, adapted to low nutrient availability in soil and having one the widest distribution ranges among the different plant species [18]. Its population is spread globally, being the only Juniperus species found in both hemispheres, with reports of this plant in Arctic regions of Asia and North America. In Europe, the largest population is found in some parts of the Alps, Scandinavia, Poland, northwest European lowlands, and Mediterranean mountain regions [19][20]. A significant population of “zimbro” is found in the Natural Park of Serra da Estrela, Portugal, where the var. alpina is mainly found at higher altitudes and the var. hemisphaerica at lower altitudes [21]. The wide geographical distribution is the principal reason for the remarkable variation in the morphological characteristics and secondary metabolites’ chemical composition [17].
On the other hand, J. communis seeds are too bitter due to their astringency. They are rarely consumed raw, usually being dried for use as a culinary component in different parts of the world. Together with juniper berries, they are commonly burnt in temples during religious ceremonies to purify the ambient air [22].
Relatively to their composition, “zimbro” plant parts are mostly composed of sugars, resins, organic acids, alkaloids, terpenic acids, leucoanthocyanins and flavonoids, gums, lignins, and wax. Their aromatic oils are rich in hydrocarbons of monoterpenes (α-pinene, β-pinene, sabinene, and myrcene), diterpenes, and sesquiterpenes [17][19][23][24][25][26].

2. Scientific Classification

J. communis species belongs to the Pinopsida class, Pinophyta division, Pinales order, Cupressaceae family, and Juniperus genus. Its binominal name is J. communis L.
The Juniperus genus is one of the most diverse within the conifers, being placed in the Cupressaceae family and including more than 60 species. It presents cosmopolitan distribution with a great capacity to develop in xerophytic and salinity conditions, although it is easier to find them in high-rainfall regions [27][28]. They are dioecious trees, producing seeds every 2 or 3 years that may have a globose or spherical morphology and be dispersed by zoocoria (e.g., frugivorous birds and small mammals), which allows them to colonize new territories quickly. In addition, they can be differentiated taxonomically into three well-differentiated sections, which represent different degrees of evolution within the genus, according to genetic analyses [27][29]. The Caryocedrus section, which is considered the most ancestral from an evolutionary point of view, is limited to areas of the Peloponnese, Anatolia, and Asia Minor and is only represented by the species J. drupacea, with acicular leaves, an anchor point to the stem, and woody cones [30]. The Juniperus (=Oxycedrus) section has a Holarctic distribution reaching the Mediterranean; it is represented by 14 species with acicular leaves, an anchoring point to the stem, and resiniferous cones. The third section, the Sabina section, is mainly found in the northern hemisphere and mountainous areas of the African continent. However, it also has some type of resin, and is distinguished from the other ones since it has decurrent needle-like or scale-shaped leaves and juicy cones [31]. According to these characteristics and with the fossil record, it is thought that the diffusion point of this genus occurred in the eastern Mediterranean region, first colonizing the northern regions of the Eurasian continent, and from there passing to the American continent at least 25 My ago [29].
All juniper species stand out for their high content of essential oils and phenolic compounds and are largely included in the traditional medicine of different cultures throughout the planet, exhibiting a wide range of biological activities and industrial applications [32]. Among them, it is worth highlighting the “zimbro” (J. communis) plant, since it shows the widest distribution, being practically circumboreal [29][33]. Another remarkable characteristic of this species is the ecological plasticity supported by great genetic variability, which translates into a substantially high number of varieties with phenotypes ranging from medium-sized trees (3–4 m high) to small creeping shrubs (Figure 1) [27][29]. The populations of the Iberian Peninsula are very diverse, and due to their position, their distribution is relegated to mountainous and more humid areas with a territorial occupation in islands, thus scaping from the thermophilic and xeric character of the nonmountainous lands of the Peninsula [30][33]. Indeed, it is believed that many years ago, this territory acted as a glacial refuge for many varieties that are currently found further north; even so, the populations of var. hemisphaerica show a high degree of genetic uniqueness, while the var. alpina (also known as var. nana) is mainly distributed in the upper areas of the mountains of the Iberian Peninsula, such as the Serra da Estrela mountains. These mountains are located in the middle interior of mainland Portugal and display an oromediterranean climatic island, being an isolated population from other populations of the Central System mountains or Cantabric System mountains [21][34].
Figure 1. Juniperus communis (A) main view of the plant growing in Serra da Estrela, (B) detail of leaves, and (C) details of berries. Images under Creative Commons licence, authorship: João Domingues Almeida and Paulo Ventura Araújo from www.flora-on.pt, accessed on 10 March 2022.
As well as other plants, J. communis also receives popular names. For example, Havusa or Matsyagandha (Sanskrit); Arar, Abahal oe Habbul (Assamese); Hayusha (Bengali); juniper berry, or common juniper (English); Palash (Gujrati); havuber or havubair (Hindi); zimbro (Portuguese); padma beeja (Kannada); hosh (Marathi); havulber (Punjabi); hapusha, abhal or arar (Urdu) [21][23].
For curiosity, and despite this plant not having a strong presence in ancient mythology, it is considered a symbol of fertility in Syria. On the other hand, in the Old Testament, it is described that the juniper has an angelic presence, which sheltered the prophet Elijah from Queen Jezebel’s pursuit. Moreover, a posteriorly biblical tale described that during their flight to Egypt, the infant Jesus and his parents used juniper to hide from King Herod’s soldiers [35].

3. Phytochemical Composition of Juniperus communis L.

As mentioned before, J. communis L. species are composed of a myriad of constituents, including nonessential substances, i.e., phytochemicals [36]. These compounds are secondary metabolites produced by plants to promote their normal cellular metabolism and offer protection against biotic and abiotic factors, and consequent oxidative injury [37]. Additionally, they are considered the key contributors to the organoleptic characteristics (e.g., aroma and color) and health benefits exhibited by plants [38]. They can be divided into five major categories (Figure 2). Although the plants’ genotype mainly influences their quantitative and qualitative composition, their levels also depend on the plant’s age, ripeness degree, cultivation techniques, geographical location, and meteorological conditions [39][40].
Figure 2. Main phytochemicals found in Juniperus communis L. [41].

3.1. Carotenoids and Chlorophylls

Although no studies have specifically reported the chlorophyll content of J. communis L. species, Rabska and colleagues [42] analyzed their total levels in fertilized and nonfertilized in both genders of this plant in autumn and winter (species not specified). The obtained data revealed nonfertilized plants had a lower concentration of total chlorophyll content than the fertilized ones (mean values of 5.0 versus (vs.) 7.4 mg/g in autumn and 3.6 vs. 4.8 mg/g in winter, respectively), and also lower amounts of total carotenoids (mean values of 0.64 and 0.95 mg/g for female and male, respectively, in autumn, and scores of 0.87 against 1.2 mg/g in winter). Focusing on gender, they observed that female plants had lower amounts of total chlorophyll compounds (values of 2.9 and 4.5 mg/g for female plants in autumn and winter, respectively, and 3.7 and 5.2 mg/g in autumn and winter, respectively, for the male ones) and carotenoid levels (values around 0.90 mg/g for female plants and around 1.0 mg/g for male, in autumn and winter, respectively). Without surprises, and regarding all the comparisons made, the authors also concluded that male and fertilized plants presented the highest levels of total chlorophyll and carotenoids (mean values of 4.3 and 1.3 mg/g, respectively).
This subclass of phytochemicals, highlighting carotenoids, possesses notable antioxidant potential and the ability to easily activate metabolic detoxification pathways, reducing the risk of appearance of several chronic and degenerative disorders [37][42].

3.2. Phenolic Compounds

Phenolics are the most predominant phytochemicals present in nature, and to date, about 10,000 different structures are currently described [43]. They are usually classified in (i) nonflavonoids and (ii) flavonoids [44]. The first ones can be further categorized into phenolic acids, including hydroxycinnamic and hydroxybenzoic acids, or in coumarins, lignans, or stilbenes [43]. On the other hand, flavonoids can be subdivided into isoflavones, coumestans, anthocyanidins, flavan-3-ols, flavanones, flavanonols (also called dihydroflavonols), flavones, or flavonols (Figure 2), depending on their structure [40]. This one comprises, at least, one phenol ring attached to one or more hydroxyl groups, and it is not only the main one responsible for dividing phenolics into different subclasses but also for conferring them a notable capacity to easily scavenge free radicals and reactive species; and to chelate metals, and in this way, counteract oxidative stress, diminish proinflammatory markers, and contribute to a healthy life state [43][45].
Focusing on phenolics found in J. communis L. species (Table 1), their levels depend on genotype, plant part, origin, age, gender, and solvent used to extract phenolics and perform the studies, but in a general way, they increase with latitude and plant age [42][46][47]. Additionally, male leaves and berries often present higher content in phenolic compounds than female ones [48].
Furthermore, and knowing the current interest in the biological potential of this plant, Brodowska et al. [49] conducted a study where they subjected J. communis (var. communis) L. berries to different ozone concentrations and time treatments. They verified that the treatment with ozone concentrations of 100 and 130 g/m3 for 30 min almost duplicated the phenolic content (15.47 and 12.91 mg catechin equivalent per g dry weight (dw), when compared to control (9.81 mg catechin equivalent per g dw), which consequently enhanced their antioxidant capacities positively.
Generally, the majority of phenolics reported in J. communis L. plant parts include 5-O-caffeoylquinic and quinic acids, catechin, epicatechin, amentoflavone, quercetin, luteolin, apigenin, and naringenin and their derivatives (Figure 3).
Table 1. Total phenolic, flavonoid, anthocyanin, and tannin content of different Juniperus communis L. plant parts extracts.
 

Gender

Origin

Extract

Total Phenolic Compounds a

Total Flavonoid

Content b

Total Anthocyanin Content c

Total Tannin Content b

References

Leaves

J. communis

(var. alpina)

n.s.

Serra Da Estrela, Portugal

Methanolic

(100%, v/v)

155.60

   

60.40

[50]

J. communis

(var. alpina)

n.s.

Yozgat, Turkey

Hydroethanolic (80% ethanol, v/v)

4.36

7.05

   

[47]

J. communis

(var. alpina)

n.s.

Yozgat, Turkey

Aqueous

169.27

24.30

   

J. communis

(var. communis)

Female

Rhodopes, Bulgaria

Methanolic

(80% methanol, v/v)

132.00

     

[46]

J. communis

(var. communis)

Female

Mountain Ozren, near Sarajevo, Bosnia, and Herzegovina

Methanolic

(80% methanol, v/v)

390.89

40.22 *

   

[48]

J. communis

(var. communis)

Male

Mountain Ozren, near Sarajevo, Bosnia, and Herzegovina

Methanolic

(80% methanol, v/v)

544.09

48.06 *

   

J. communis

(var. communis)

n.s.

Nainital, India

Hydroethanolic (70% ethanol, v/v)

238.78

     

[51]

J. communis

(var. communis)

n.s.

Nainital, India

Hexane

189.65

     

J. communis

(var. communis)

n.s.

Nainital, India

Ethyl acetate

315.33

     

J. communis

(var. communis)

n.s.

Nainital, India

Aqueous

205.33

     

J. communis

(var. oblonga pendula)

Male

North Carolina, USA

Methanolic

(80% methanol, v/v)

91.00

     

[46]

J. communis

(var. saxatiles)

n.s.

Turkey

Hydroethanolic

(80% ethanol, v/v)

212.10

     

[52]

Berries

J. communis

(var. alpina)

n.s.

Yozgat, Turkey

Hydroethanolic

(80% ethanol, v/v)

Ripe berry: 11.92

Unripe berry: 130.92

Ripe berry: 2.56

Unripe berry: 17.57

   

[47]

J. communis

(var. alpina)

n.s.

Yozgat, Turkey

Aqueous

Ripe berry: 4.36

Ripe berry: 7.05

   

J. communis

(var. communis)

 

North-East Slovakia

Hydroethanolic

(70% ethanol, v/v)

Ripe berry: 6.87–42.23

     

[53]

J. communis

(var. communis)

n.s.

Melbourne, Australia

Hydroethanolic

(30% ethanol, v/v)

Ripe berry: 9.08

Ripe berry: 2.25

 

Ripe berry: 3.48 *

[54]

J. communis

(var. communis)

n.s.

Quebec, Canada

Hydroethanolic

(80% ethanol, v/v)

Ripe berry: 99.20 b

 

Ripe berry: 0.47

 

[55]

J. communis

(var. communis)

n.s.

Serra Da Estrela, Portugal

Methanolic

(100%, v/v)

Ripe berry: 44.70

     

[50]

J. communis

(var. communis)

n.s.

Ağrı, Turkey

Methanolic

(100%, v/v)

Ripe berry: 59.17

     

[56]

J. communis

(var. n.s.)

n.s.

Pitesti hills, Romania

Hydroethanolic

(50% ethanol, v/v)

Ripe berry: 0.19

Ripe berry: 51.09 d

   

[57]

J. communis

(var. saxatilis)

n.s.

Yozgat, Turkey

Hydroethanolic

(80% ethanol, v/v)

Ripe berry: 21.00

     

[52]

J. communis

(var. saxatilis)

n.s.

Ankara, Turkey

Methanolic

(100%, v/v)

Ripe berry: 17.64

     

[56]

J. communis

(n.s.)

n.s.

Šara mountain in south Serbia

Chloroformic

189.82

27.11 d

   

[36]

J. communis

(var. n.s.)

n.s.

Šara mountain in south Serbia

Ethanolic

189.82

42.85 d

   

J. communis

(var. n.s.)

n.s.

Šara mountain in south Serbia

Ethyl acetate

144.21

38.40 d

   

Stems

J. communis

(var. alpina)

n.s.

Serra Da Estrela, Portugal

Methanolic

(100%, v/v)

221.30

   

79.30

[50]

Figure 3. Main phenolic compounds found in Juniperus communis L. vegetal parts.

3.3. Volatile Organic Compounds (VOC’s)

J. communis L. parts, namely their essential oils, present many volatile organic compounds (VOCs) in their composition, particularly the presence of monoterpene hydrocarbons, oxygenated monoterpenes, sesquiterpene hydrocarbons, and oxygenated sesquiterpenes [57][39]. As well as other phytochemicals, their levels also depend on genotype, origin, cultivation methods, meteorological conditions, and extraction techniques. Even so, among the species, monoterpenes such as α-pinene, β-pinene, and β-myrcene are the most commonly found, followed by some sesquiterpenes compounds, namely germacrene D (Figure 4) [58][59][60].
Figure 4. The main volatile organic compound found in Juniperus communis L. parts.
A total of 57 different VOCs were detected in leaf ethanolic extracts (50:50, v/v), with pimaric acid being the predominant one (29.74% of total VOCs), followed by α-pinene (14.86% of total VOCs), β-myrcene (6.99% of total VOCs), bicyclosesquiphellandren (6.87% of total VOCs), and β-pinene (5.29% of total VOCs) [57]. On the other hand, extracts of var. communis exhibited higher percentages of limonene (26.12%), benzene (15.62%), β-myrcene (9.08%), and β-pinene (7.30%) [61]. Focusing on var. alpina, the predominant ones in their ethanolic extracts (50% ethanol, v/v) were δ-cadinene (12.80%), α-pinene (11.0%), germacrene D (9.30%), and borneol (8.60%) [62].
Regarding essential oils of their leaves, in the var. communis, α-pinene was also the most found (34.87% of total VOCs), followed by citronellyl acetate (14.26%), limonene (10.72%), and terpinolene (10.65%). Additionally, vestigial amounts (<6.21%) of ρ-cymene, elemene, cadinene, cyclohexane, cedrol, and caryophyllene were also reported [58]. α-Pinene was also the most abundant compound detected in different leaves of var. communis from different regions of the United States of America (USA) (66.6–75.2%) [63].
On the other hand, in leaves of var. communis from Serbia, sabinene was the main one reported (39.40%), followed by α-pinene (13.3%) and β-myrcene (4.70%) [64], whereas in var. alpina from France, limonene was clearly the most predominant (30.90%), followed by α-pinene (24.40%), β-phellandrene (12.60%), β-myrcene (3.60%), and α-phellandrene (3.60%) [65]. Similar percentages of α-pinene were reported on var. saxatilis (23.60%); additionally, this species was also shown to possess considerable percentages of α-cadinene (10.71%), sabinene (9.53%), germacrene D (7.25%), α-murolene (6.58%), and γ-cadinene (5.87%) [66][64].
The combination of all of these results is evidence that local origin influences the phytochemical profile. Additionally, Gonny et al. [65] determined the VOC profile of J. communis woods and roots of var. alpina. For woods, α-terpinyl acetate (9.10%) and α-terpineol (8.4%) were the predominant ones, while for roots, a high percentage of cedrol (37.70%) and cinnamyl acetate (11.50%) were found.
VOCs have been gaining great interest owing to their remarkable antimicrobial, antioxidant, anti-inflammatory, and anticancer properties, being able to attenuate, or even mitigate, the development of cardiovascular disorders and neuropathologies, and also ameliorate the mental state of individuals [67].

4. Biological Potential of Juniperus communis Linnaeus

Since ancient times, J. communis parts have been largely used as antiseptics, contraceptives, and diuretics, and as a remedy to treat colds, chest complaints, rheumatism, headaches, dermatological and respiratory ailments, and kidney and urinary infections [68][69][67]. Given the aforementioned, it is not surprising that this plant is a focus of continuous studies to discover its full potential.
To date, several reports have highlighted its antimicrobial, antifungal, antioxidant, anti-inflammatory, and antidiabetic potential, as well as its anticarcinogenic, hepatoprotective, neuronal, and renal effects, as described in Figure 5 and Table 2, Table 3 and Table 4 [70][68][57][52][71][72][73]. Next, a summary of the main studies already published concerning the health-promoting properties of this plant will be presented.
Figure 5. Main health-promoting properties attributed to Juniperus communis L. AST: aspartate aminotransferase; ALT: alanine aminotransferase; AChE: acetylcholinesterase; BChE): butyrylcholinesterase; Bax: Bcl-2-associated X protein; AIF: apoptosis-inducing factor; Fas: cell-surface death receptor; FasL: Fas ligand; Bcl-2: B-cell lymphoma 2; ↑: increase; ↓: reduction; T: inhibition.
Table 2. Antimicrobial and antiparasitic activity of different Juniperus communis extracts.

Part of the Plant

Origin

Subspecies/Variety

Method

Inhibited Species

References

Antimicrobial activity

Essential oils

Berries

Poland

n.s.

Disc diffusion

Staphyllococcus aureus, Serratia marcenscens, Enterobacter cloace, Klebsiella pneumoniae, Pseudomonas aeruginosa, Acinetobacter baumanii, Listeria monocytogenes, and Candida albicans

[74]

Needles

Portugal

var. alpina

MIC and MLC

Epidermophyton floccosum, Microsporum canis, M. gypseum, Trichophyton mentagrophytes, T. mentagrophytes var. interdigitale, T. rubrum, and T. verrucosum

[75]

Needles and berries

Italy

var. communis

MIC

C. albicans, S. aureus, and P. aeuroginosa

[76]

Plant material (leaves and stems)

Iran

n.s.

Disc diffusion

S. aureus, P. aeruginosa, and E. coli

[22]

Berries

Slovenia

n.s.

Biofims assay

Campylobacter jejuni, L. monocytogenes

[77]

Plant material (undifferentiated)

Slovenia

n.s.

Disc diffusion

S. aureus and C. albicans

[78]

Berries

Spain

n.s.

MIC

E. coli, Proteus mirabilis, K. pneumoniae, P. aeruginosa and Morganella morganii, MRSA, and L. monocytogenes

[60]

Berries

Portugal

n.s.

MIC and MLC

B. cereus, B. subtilis, E. aerogenes, E. faecalis, E. coli, K. pneumoniae, Proteus mirabilis, P. aeruginosa, Salmonella typhimurium, S. aureus, and C. albicans

[79]

Leaves

Croatia

n.s.

Disc diffusion, MIC, and MLC

16 species of bacteria and 14 species of fungus

[80]

Berries

Serbia

n.s.

Disc diffusion, MIC, MLC, and in vivo adhesion assay

S. aureus, MRSA, E. faecalis, L. monocytogenes, E. coli, S. flexneri, S. enteritidis, P. aeruginosa, Aspergillus fumigatus, A. versicolor, A. ochraceus, A. niger, Trichoderma viride, Penicillium funiculosum, P. ochrochloron, and P. verrucosum var. cyclopium

[81]

Plant material (leaves and branches)

Egypt

n.s.

MIC

S. aureus, E. coli, and C. albicans

[82]

Plant material

Croatia

n.s.

MIC and biofilm assay

Mycobacterium avium, M. intracellulare, and M. gordonae

[83][84]

Phenolic-rich extracts

Berries

Slovenia

n.s.

Biofilms assay

C. jejuni, L. monocytogenes

[77]

Plant material

Italy

n.s.

Disc diffusion and MIC

Actinomyces viscosus, Lactobacillus casei, Streptococcus mutans, S. sobrinus, and general oral microbiota

[85]

Berries

Turkey

n.s.

Disc diffusion and MIC

S. epidermidis, S. aureus, B. subtilis, P. aeruginosa, E. coli, and C. albicans

[86]

Leaves

Turkey

var. communis and var. saxatilis

MIC

S. aureus

[87]

Leaves

Poland

n.s.

Disc diffusion

K. pneumoniae, S. enteritidis,

P. aeruginosa, A. baumannii, E. faecium, S. aureus, L. fermentum, Clostridium butyricum, L. monocytogenes, B. coagulans, C. utilis, Aspergillus spp., and Fusarium spp.

[88]

Stem (branches)

Italy

var. communis and var. saxatilis

Biofilm formation

S. aureus

[89]

Berries

Turkey

var. communis and var. saxatilis

MIC and MLC

S. aureus, S. epidermidis, E. hirae, B. subtilis, E. coli, P. mirabilis, P. aeruginosa, C. albicans, and C. parapsilosis

[56]

Leaves

India

n.s.

MIC

E. coli, S. aureus, and K. pneumoniae

[90]

Antiparasitic activity

Essential oils

Stems and leaves

France

n.s.

Radioactive micromethod

Two different strains of Plasmodium falciparum, which were chloroquine-resistant (FcBl) and chloroquine-sensitive (Nigerian) strains

[91]

n.s.: not specified; MIC: Minimal inhibitory concentration; MLC: Minimal lethal concentration.

Table 3. In vitro and in vivo antioxidant effects of Juniperus communis extracts.

Part of the Plant

Origin

Extract

Subspecies/

Variety

Experimental Model

Effect

References

In vitro assay

Berries

Romania

Ethanolic

(50% ethanol, v/v)

n.s.

Capacity to scavenge DPPH

IC50 value of 1.42 µg/mL

[24][57][36][47][53]

Serbia

Ethanolic

IC50 value of 28.55 µg/mL

Ethyl acetate

IC50 value of 106.44 µg/mL

Chloroform

IC50 value of 257.66 µg/mL

Poland

Methanolic

(70%, methanol v/v)

IC50 values from 6.86 to 13.66 µg/L

Essential oils

IC50 varying from 1.27 to 4.25 µg/L

Turkey

Methanolic

var. saxatilis

IC50 value of 1.84 mg/mL

var. communis

IC50 value of 0.63 mg/mL

Ethanolic

(80% ethanol, v/v)

var. alpina

Inhibitory percentages of 33.25, 34.27, and 36.26% at 0.5, 1, and 2 mg/mL, respectively

Aqueous

Inhibitory percentages of 48.40, 63.29, and 82.03% at 0.5, 1, and 2 mg/mL, respectively

Poland

Methanolic

(70% methanol, v/v)

n.s.

Reducing power potential

Values ranging 6.90 and 10.70 mM FeSO4 × 7H2O

[47][49][53][56]

Essential oils

Values ranging from 0.47 and 1.11 mM FeSO4 × 7H2O

Turkey

Methanolic

var. communis

IC50 value of 12.82 mg/mL

12.82 ascorbic acid equivalent/mL

var. saxatilis

IC50 value of 64.14 mg/mL

64.14 ascorbic acid equivalent//mL

Ethanolic

(80% ethanol, v/v)

var. alpina

Inhibitory percentages of 0.083, 0.095, and 0.203% at 0.5, 1, and 2 mg/mL, respectively

Aqueous

Inhibitory percentages of 0.424, 0.689, and 1.371% at 0.5, 1, and 2 mg/mL, respectively

Poland

Methanolic

(70% methanol, v/v)

n.s.

β-carotene bleaching test

β-carotene inhibitory potential varying from 24.36 to 30.63%

[49]

Essential oils

n.s.

β-carotene inhibitory potential varying from 1.19 to 2.39%

Turkey

Methanolic

var. saxatilis

Protect liposomes from lipid peroxidation

IC50 value of 120.07 µg/mL

[56]

var. communis

IC50 value of 4.44 µg/mL

Turkey

Methanolic

var. saxatilis

Ferrous ion (Fe2+)-chelating activity

Chelating ability around 30% at 2 mg/mL

[24][47][55]

var. communis

Chelating ability around 15% at 2 mg/mL

Ethanolic

(80% ethanol, v/v)

var. alpina

Inhibitory percentages of 4.88, 14.86, and 32.82% at 0.5, 1, and 2 mg/mL, respectively

Aqueous

Inhibitory percentage of 0.83% at 2 mg/mL

Canada

Ethanolic

(80% ethanol, v/v)

var. communis

Capacity to scavenge peroxyl radicals

3876 µM Trolox equivalents at 1 mg/mL

[24][52]

n.s.

Essential oil

n.s.

Capacity to scavenge ABTS•+ species

IC50 value of 10.96 µg/mL

Turkey

Ethanolic

(80% ethanol, v/v)

var. saxatilis

Capacity to scavenge ABTS•+ species

Inhibitory percentages of 42.5%, respectively at 3 mg/mL

n.s.

Essential oil

n.s.

Capacity to scavenge hydroxyl radicals

IC50 value of 0.0066 µg/mL

[24]

n.s.

Essential oil

n.s.

Capacity to scavenge superoxide anions

IC50 of 0.822 µg/mL

[24][47]

Turkey

Ethanolic

(80% ethanol, v/v)

var. alpina

Inhibitory percentages of 20.07, 21.97, and 17.80% at 0.5, 1, and 2 mg/mL, respectively

Aqueous

var. alpina

Inhibitory percentages of 5.49, 10.61, and 11.17% at 0.5, 1 and 2 mg/mL, respectively

Crushed berries

Slovakia

Ethanolic

(70% ethanol, v/v)

n.s.

Capacity to scavenge hydroxyl radicals

Inhibitory values varying from 65.59 to 88.12% (recalculated by dry matter (DM), from 3.06 to 5.75%/g DM)

[53]

Noncrushed berries

Slovakia

Ethanolic

(70% ethanol, v/v)

n.s.

Inhibitory values varying from 15.52 and 32.85% (recalculated by dry matter (DM), from 1.20 to 20.05%/g DM) for

Unripe berries

Turkey

Ethanolic

(80% ethanol, v/v)

var. alpina

Capacity to scavenge superoxide anions

Inhibitory percentages of 14.58, 10.99, and 18.37% at 0.5, 1, and 2 mg/mL, respectively

[47]

Capacity to scavenge DPPH

Inhibitory percentages of 46.21, 57.32, and 73.75% at 0.5, 1, and 2 mg/mL, respectively

[24][47]

Capacity to chelate metals

Inhibitory percentages of 6.32, 5.04, and 16.59% at 0.5, 1, and 2 mg/mL, respectively

[47]

Ferric-reducing antioxidant power

Inhibitory percentages of 0.288, 0.504, and 0.855% at 0.5, 1, and 2 mg/mL, respectively

Leaves

India

Ethanolic

(70% ethanol, v/v)

n.s.

Capacity to scavenge DPPH

IC50 value of 213 µg/mL

[46][47][51][64][92]

Aqueous

var. communis

IC50 value of 347 µg/mL

Ethyl acetate

IC50 value of 177 µg/mL

Turkey

Ethanolic

(80% ethanol, v/v)

var. alpina

Inhibitory percentages of 66.62, 83.06, and 91.40% at 0.5, 1, and 2 mg/mL, respectively

Aqueous

Inhibitory percentages of 34.92, 35.56, and 37.29% at 0.5, 1, and 2 mg/mL, respectively

Bulgaria

Methanolic

(80% methanol, v/v)

var. Oblonga Pendula

IC50 value of 258 µg/mL

Serbia

Essential oil

var. communis

IC50 value of 660 µg/mL

Serbia

Essential oil

var. saxatilis

Potential to chelate metals

IC50 value of 320 µg/mL

[47][92]

India

Ethyl acetate

var. communis

IC50 value of 261 µg/

mL

Turkey

Acetate

n.s.

Inhibitory effect of 6.05% at 1 mg/mL

Aqueous

var. alpina

Inhibitory percentages of 9.06, 12.39, and 38.40% at 0.5, 1, and 2 mg/mL, respectively

Turkey

Ethanolic

(80% ethanol, v/v)

var. alpina

Capacity to scavenge superoxide anions

Inhibitory percentages of 20.26, 25.00, and 25.38% at 0.5, 1, and 2 mg/mL, respectively

[47][66]

Turkey

Ethanolic

(80%, ethanol v/v)

var. alpina

Ferric-reducing antioxidant power

Inhibitory percentages of 0.681, 1.278, and 1.971% at 0.5, 1, and 2 mg/mL, respectively

Aqueous

Inhibitory percentages of 0.121, 0.120, and 0.154% at 0.5, 1, and 2 mg/mL, respectively

Serbia

Distilled extracts

var. saxatilis

Reduction capacity of 78.77 mg of ascorbic acid equivalents per g of dry matter

Serbia

Distilled extracts

var. saxatilis

Lipid-peroxidation inhibitory potential

IC50 value of 540 µg/mL

[66]

Essential oils

IC50 value of 2440 µg/mL

Turkey

Ethanolic

(80% ethanol, v/v)

var. saxatilis

Capacity to scavenge ABTS•+ species

Inhibitory percentage of 99.5 at 3 mg/mL

[52]

Shoots

Poland

Crude extract

n.s.

Antioxidant-enzyme activity and reactive oxygen species in vitro assays

↑↑ the activity of intracellular antioxidant enzymes superoxide dismutase and catalase

↓↓ reactive oxygen species

[42]

Turkey

Acetone

n.s.

Capacity to chelate metals

Inhibitory percentage of 6.05% at 1 mg/mL

[92]

Ethyl acetate

Inhibitory percentage of 22.59 at 1 mg/mL

Ethanolic

(75% ethanol, v/v)

Inhibitory percentage of 12.31% at 1 mg/mL

Twigs

Spain

Essential oil

n.s.

Peroxy-radical-induced oxidation inhibition

120 µmol Trolox/gram of essential oil

[39]

Hops

Australia

Ethanolic

(30% ethanol, v/v)

n.s.

Ferric ion-reducing antioxidant power

4.17 mg of ascorbic acid equivalents per g

[54]

Capacity to scavenge DPPH

9.26 mg of ascorbic acid equivalents per g

Capacity to scavenge ABTS•+ species

49.54 mg of ascorbic acid equivalents per g

Plant material (twigs, leaves, and berries)

Spain

Essential oil

n.s.

Reducing power assay

IC50 values from 135 to 970 µg/mL

[60]

Spain

Essential oil

n.s.

Inhibition of oxidation process

IC50 values from 324.76 to 1563.29 µg/mL

In vivo assay

Leaves

India

Methanolic

n.s.

Effects on Wistar rats with induced Parkinson’s disease by chlorpromazine for 21 days at a dose of 200 mg/kg

↑↑ in reduced glutathione

↓↓ levels of TBARS

[19]

Romania

Essential oil

n.s.

Effects of juniper volatile oil (1% and 3%) daily inhalation on Amyloid Beta (1–42)-induced oxidative stress in Wistar rats

↑↑ superoxide dismutase and catalase enzymes, and glutathione peroxidase activity

[67]

n.s.: not specified; IC50: half-maximal inhibitory concentration; TBARS: thiobarbituric acid-reactive substances, DPPH: 2,2-diphenyl-1-picrylhydrazyl radical; ABTS+: 2,2′-azino-bis (3-ethylbenzothiazoline-6-sulfonic acid; ↑↑: increase; ↓↓: reduction.

Table 4. In vitro and in vivo health benefits of Juniperus communis extracts.

Part of the Plant

Origin

Extract

Subspecies/

Variety

Experimental Model

Effect

References

Anti-inflammatory and antinociceptive properties

In vitro assay

Plant parts

Sweden

Aqueous

n.s.

Prostaglandin biosynthesis assay

Platelet activating factor-induced exocytosis assay

↓↓ prostaglandins by 55% at 200 µg/mL

↓↓ platelet activating factor-induced exocytosis by 78% at 250 µg/mL

[93]

Woods

Austria

Methylene chloride

n.s.

12(S)-lipoxygenase assay

↓↓ 12[S]-hydroxy-5,8,10,14-eicosatetraenoic acid by 54.0% at 100 µg/mL, 66.2 and 76.2%,

[73]

Berries

Austria

Methylene chloride

n.s.

↓↓ 12[S]-hydroxy-5,8,10,14-eicosatetraenoic acid by 66.2% at 100 µg/mL

Ethyl acetate

↓↓ 12[S]-hydroxy-5,8,10,14-eicosatetraenoic acid by 76.2% at 100 µg/mL

Plant material (twigs, leaves, and fruits)

Spain

Essential oil

n.s.

Inhibition of nitric oxide production in lipopolysaccharide-activated murine macrophage RAW 264.7 cells

IC50 values from 23.98 to 84.80 µg/mL

[60]

In vivo assays

Berries

Italia

Hydroethanolic

(80% ethanol, v/v)

var. communis

Effects on the inhibition of writhing carrageenin foot edema in male Wistar rats after 7 days of treatment at doses of 100 and 200 mg/kg

↓↓ carrageenin-foot edema by 60% and 79% at 100 and 200 mg/kg, respectively

[94]

Turkey

Aqueous

12.8% inhibition (berries)

[95]

Berries, leaves, and stems

Turkey

Methanolic

var. communis

18.5% inhibition (stems)

3.9% inhibition (berries)

18.5% inhibition (leaves)

Aqueous

var. saxatilis

9.1% inhibition (berries)

7.8% inhibition (leaves)

Methanolic

30.5% inhibition (berries)

35.2% inhibition (leaves)

Aqueous

var. communis

Effects on stimulating response latency in male Swiss albino mice using a hot plate after administration of 100 mg/kg of extract

4.27% inhibition (stems)

5.36% inhibition (berries)

4.29% inhibition (leaves)

Methanolic

4.40% inhibition (stems)

4.11% inhibition (berries)

5.16% inhibition (leaves)

Aqueous

var. saxatilis

3.26% inhibition (stems)

4.32% inhibition (berries)

5.13% inhibition (leaves)

Methanolic

3.13% inhibition (stems)

4.05% inhibition (berries)

5.31% inhibition (leaves)

Aqueous

var. communis

Effects on carrageenin-induced hind-paw edema in male Swiss albino mice after 360 min of 100 mg/kg extract administration

65.9% inhibition (stems)

65.1% inhibition (berries)

65.4% inhibition (leaves)

Methanolic

54.3% inhibition (stems)

65.8% inhibition (berries)

54.8% inhibition (leaves)

Aqueous

var. saxatilis

69.6% inhibition (stems)

51.9% inhibition (berries)

53.6% inhibition (leaves)

Methanolic

65.7% inhibition (stems)

43.4% inhibition (berries)

45.3% inhibition (leaves)

Methanolic

var. saxatilis

Effects on PGE2-induced hind-paw edema effects in male Swiss albino mice after 360 min of 100 mg/kg extract administration

17.6% inhibition (stems)

16.5% inhibition (berries)

16.8% inhibition (leaves)

Leaves

India

Methanolic

n.s.

In vivo study involving different nociceptive assays (acetic acid-induced writhing, formalin and tail-flick tests) in Swiss albino mice at 100 and 200 mg/kg

↓↓ writhing response and the late phase related with the formalin test

Act centrally since the extract and pethidine effects were blocked by naloxone in the tail-flick test

[96]

Berries

Romania

Hydroethanolic microemulsions

n.s.

Effects on paw edema in dextran-induced inflammation Wistar rats’ model

↓↓ paw edema

[57]

Berries

Romania

Hydroethanolic microemulsions

n.s.

Kaolin-induced inflammation in Wistar rats’ model

↓↓ interleukins -1β and 6 expression

↓↓ tumor necrosis factor alfa

Antidiabetic, antihypercholesterolemic and antihyperlipidemic effects

In vitro assays

Fruits

Turkey

Hydroethanolic

(80% ethanol, v/v)

var. saxatilis

Capacity to inhibit α-amylase activity

Inhibitory value of 29.8% at 3 mg/mL

[52]

Capacity to inhibit α-glucosidase activity

IC50 value of 4.4 µg/mL

Leaves

Turkey

Hydroethanolic

(80% ethanol, v/v)

var. saxatilis

Capacity to inhibit the α-amylase activity

Inhibitory value of 84.3% at 3/mg/mL

Capacity to inhibit the α-glucosidase activity

IC50 value of 53.6 µg/mL

Plant material

United Kingdom

Aqueous

n.s.

Effects on glucose movement

↓↓ glucose diffusion by 6% at 50 g/L

[97]

In vivo assays

Berries

United Kingdom

 

n.s.

Streptozotocin-induced diabetic mice models for 40 days at doses of 1 g/400 mL

↓↓ polydipsia

Prevent weight losses

[98]

Spain

Aqueous

n.s.

Effects on streptozotocin-induced diabetic rat models after 24 days of treatment at doses of 250 and 500 mg/kg

↓↓ hypoglycemia in normoglycemic rats

[99]

Effects on streptozotocin-induced diabetic rat models after 24 days of treatment at 125 mg/kg

↓↓ blood glucose levels and mortality index

Prevent weight losses

Turkey

Oil dissolved in 0.5% of sodium carboxymethyl cellulose

n.s.

Effects on albino Wistar rats after 30 days of treatment at doses of 50, 100 and 200 mg/kg

↓↓ total cholesterol, oxidized low-density lipoprotein, alanine aminotransferase, and aspartate transaminase levels

↑↑ blood urea nitrogen and creatinine levels

[100]

Plant

n.s.

Methanolic extracts

n.s.

Effects on streptozotocin-nicotinamide induced diabetic rats after 21 days of treatment at doses of 100 and 200 mg/kg

↓↓ blood glucose levels, total cholesterol, triglycerides, low-density lipoprotein, and very-low-density lipoprotein cholesterols

↑↑ high-density lipoprotein cholesterol

[101]

Herbal preparation also composed of Juniperus. communis

Croatia

Hydroethanolic

(60% ethanol, v/v)

n.s.

Effects on alloxan-induced nonobese diabetic NOD mice after 7 days of treatment at 20 mg/kg

↓↓ glucose and fructosamine levels

[102]

Antiproliferative effects

In vitro assays

Berries

Nepal

Aqueous

n.s.

Effects on OECM-1 human gingival squamous cancer cells after 24 h of exposure

Induce apoptosis, exhibiting an IC50 value of 46.20 µg/mL

[103]

Plant material

n.s

Aqueous

n.s

Effects on CE81T/VGH human esophageal squamous cell carcinoma after 24, 48 and 72 h of exposure

Induce cell cycle arrest at the G0/G1 phase by regulating the expression of p53/p21 and CDKs/cyclins, triggering cell apoptosis by activating both the extrinsic (Fas/FasL/Caspase 8) and intrinsic (Bcl-2/Bax/Caspase 9) apoptosis pathways

IC50 values of 68.41, 64.33, and 60.07 µg/mL after 24, 48, and 72 h of exposure, respectively

[69]

Effects on CE48T/VGH human esophageal epidermoid carcinoma after 24, 48, and 72 h of exposure

Induce cell-cycle arrest at the G0/G1 phase, by regulating the expression of p53/p21 and CDKs/cyclins, triggering cell apoptosis by activating both the extrinsic (Fas/FasL/Caspase 8) and intrinsic (Bcl-2/Bax/Caspase 9) apoptosis pathways

IC50 values of 69.38, 56.96, and 36.10 µg/mL after 24, 48, and 72 h of exposure, respectively

USA

Distilled extracts

Effects on B16/F10 melanoma cells after 24 and 48 h of exposure

Induced apoptosis, decreased angiogenesis and metastasis, and diminished cancer stem-cell expression

IC50 values of 27 and 44 µg/mL, after 24 and 48 h of exposure, respectively

[58]

Leaves

Turkey

Methanolic

n.s.

Effects on C6 rat brain tumor and HeLa human cervix carcinoma cells after 24 h of exposure

IC50 value of 28.43 µg/mL (C6 rat brain tumor)

IC50 value of 32.96 µg/mL (HeLa cancer cells)

[104]

Aerial parts

Egypt

Methanolic

n.s.

Effects on PC3 human prostate, HCT 116 human colon, and MCF7 breast cancer cells after 24 h of exposure

IC50 value of 23.8 µg/mL (PC3 cancer cells)

IC50 value of 37.6 µg/mL (HCT 116 cancer cells)

IC50 value of 23.8 µg/mL (MCF7 cancer cells)

[105]

Plant material

New Mexico, USA

Aqueous

n.s.

Effects on MCF-7/AZ breast cancer cells after 24 h of exposure

IC50 value of 50 µg/mL

[60][106]

Spain

Essential oil

n.s.

Effects on NCI-H460 lung, MCF-7 breast, AGS gastric, and Caco-2 cancer cells after 24 h of exposure

IC50 values varying from 41.99 to 44.87 µg/mL (NCI-H460 cancer cells)

IC50 values varying from 30.88 to 163.99 µg/mL (MCF-7 cancer cells)

IC50 values varying from 132.68 to 302.86 µg/mL (AGS cancer cells)

IC50 values varying from 107.65 to 230.79 µg/mL (Caco-2 cancer cells)

Berries

Australia

Methanolic

n.s.

Effects on Caco-2 human colorectal and HeLa cervical cancer cells after 12 h of exposure

IC50 value of 1383 µg/mL (Caco-2 cancer cells)

IC50 value of 2592 µg/mL (HeLa cancer cells)

[107]

Aqueous

n.s.

Effects on Caco-2 human colorectal and HeLa cervical cancer cells after 12 h of exposure

IC50 value of 1516 µg/mL (Caco-2 cancer cells)

IC50 value of 2157 µg/mL (HeLa cancer cells)

Serbia

Essential oil and Distilled extracts

var. saxatilis

Effects on A549 human lung adenocarcinoma epithelial cells after 24 h of treatment after 24 h of exposure

Induced apoptosis and arrested cell cycle in G2/M

IC50 value of 69.4 µg/mL (essential oil)

IC50 value 1270 µg/mL (distilled extract)

[66]

USA

Distilled extracts

n.s.

Effects on HepG2 human hepatocellular cancer cells after 24, 48, and 72 h of exposure

IC50 values of 48.9, 42.3, and 43.9 µg/mL, after 24, 48, and 72 h of exposure, respectively

[108][109]

Effects on Mahlavu human hepatocellular carcinoma cells after 24, 48, and 72 h of exposure

IC50 values of 64.9, 58.5, and 59.4 µg/mL, after 24, 48, and 72 h of exposure, respectively

Effects on J5 human hepatocellular carcinoma cells after 24, 48, and 72 h of exposure

IC50 values of 74.2, 67.2, and 53.2 µg/mL, after 24, 48, and 72 h of exposure, respectively

Effects on HT-29 colon cancer cells after 24, 48, and 72 h of exposure

Induced cell-cycle arrest at the G0/G1 phase via regulation of p53/p21 and CDK4/cyclin D1

Induced cell apoptosis via the extrinsic (FasL/Fas/caspase-8) and intrinsic (Bax/Bcl-2/caspase-9) apoptotic pathways

IC50 values of 66.71, 60.02, and 54.32 µg/mL, after 24, 48, and 72 h of exposure, respectively

Effects on CT-26 colon cancer cells after 24, 48, and 72 h of exposure

Induced cell-cycle arrest at the G0/G1 phase via regulation of p53/p21 and CDK4/cyclin D1

Induced cell apoptosis via the extrinsic (FasL/Fas/caspase-8) and intrinsic (Bax/Bcl-2/caspase-9) apoptotic pathways

IC50 values of 27.8, 22.7, and 27.3 µg/mL, after 24, 48, and 72 h of exposure, respectively

Leaves and branches

Wyoming, USA

Essential oil

n.s.

Effects on SH-SY5Y human neuroblastoma cells after 24 h of exposure

IC50 value of 53.7 µg/mL

[110]

Seed cones

Serbia

Essential oil

var. saxatilis

Effects on HT-29 and HCT116 colon cancer cells after 24 h of exposure

IC50 value 125 µg/mL (HT-29)

IC50 value of 62.5 µg/mL (HCT116)

[81]

Distilled extracts

IC50 value 625 µg/mL (HT-29)

IC50 value of 1250 µg/mL (HCT116)

Roots

China

Acetone

n.s.

Effects on N18 neuroblastoma cell lines after 24 and 48 h of exposure

Induced glioma cell-cycle arrest through intrinsic and extrinsic apoptotic pathways

IC50 values of 61.11 and 68.94 µg/mL, after 24 and 48 h of exposure, respectively

[111]

Effects on DBTRG-05MG, G5T/VGH, GBM8401, GBM8901, and RG2 glioblastoma cell lines after 24 h of exposure

Induced glioma cell-cycle arrest through intrinsic and extrinsic apoptotic pathways

IC50 value of 67.04 µg/mL (DBTRG-05MG glioblastoma cells)

IC50 value of 63.3 µg/mL (G5T/VGH glioblastoma cells)

IC50 value of 57.14 µg/mL (GBM8401glioblastoma cells)

IC50 value of 58.45 µg/mL (GBM8901 glioblastoma cells)

IC50 value of 69.97 µg/mL (RG2 glioblastoma cells)

Effects on DBTRG-05MG, G5T/VGH, GBM8401, GBM8901, and RG2 glioblastoma cell lines after 48 h of exposure

Induced glioma cell-cycle arrest through intrinsic and extrinsic apoptotic pathways

IC50 value of 49.46 µg/mL (DBTRG-05MG glioblastoma cells)

IC50 value of 67.85 µg/mL (G5T/VGH glioblastoma cells)

IC50 value of 46.68 µg/mL (GBM8401glioblastoma cells)

IC50 value of 55.49 µg/mL (GBM8901 glioblastoma cells)

IC50 value of 53.8 µg/mL (RG2 glioblastoma cells)

In vivo assays

Plant

USA

Distilled extracts

n.s.

Effects on melanoma tumor model in C57BL/6 mice after 23 days of treatment at a dose of 200 mg/kg

Cell-cycle arrest at the G0/G1 phase

↓↓ tumor size by 45.2%, B-cell lymphoma-2 (Bcl-2), procaspases 8 and 9 and higher levels of Bcl-2-associated X protein, apoptosis-inducing factor, cell-surface death receptor Fas and Fas ligand when compared to untreated control

[58]

Berries

USA

Distilled extracts

n.s.

Effects in BALB/c nude mice injected with HepG2 liver cancer cells at a dose of 200 mg/kg

↓↓ tumor size

↑↑ lifespan with no or low systemic and pathological toxicity

[108]

Effects in female BALB/c mice injected with CT-16 colon cancer cells at a dose of 200 mg/kg

Inhibited proliferation

Induced apoptosis

No obvious change in body weight or histological morphology of normal organs after treatment

[109]

Roots

China

Acetone

n.s.

Effects in male Foxn1 nu/nu mice injected with DBTRG-05MG human glioblastoma cells after 100 days of treatment at a dose of 200 mg/kg

Can penetrate the blood-brain barrier

↓↓ tumor size and the degree of neovascularization

↑↑ PCNA, VEGFR-1, and VEGFR-2 in 44.49%, 5.88%, and 5.85%, respectively, when compared to untreated control

[111]

Neuronal effects and anticataleptic activity

In vitro assays

Leaves

Turkey

Hydroethanolic

(80% ethanol, v/v)

var. alpina

Capacity to inhibit acetylcholinesterase activity

10.38% inhibition at 50 µg/mL

24.30% inhibition at 100 µg/mL

32.89% inhibition at 200 µg/mL

[47][92]

Ripe Berries

Aqueous

5.47% inhibition at 100 µg/mL

28.17% inhibition at 200 µg/mL

Shoots

Ethyl acetate, ethanolic, and acetone extracts

n.s.

21.34% inhibition at 100 µg/mL (ethyl acetate extract)

13.46% inhibition at 100 µg/mL (ethanolic extract)

28.43% inhibition at 100 µg/mL (acetone extract)

Inhibitory percentages varying from 32.34 to 41.97%% inhibition at 100 µg/mL (ethyl acetate extract)

Inhibitory percentages varying from 22.29 to 45.45% inhibition at 100 µg/mL (ethanolic extract)

Inhibitory percentages varying from 1.91 to 38.55% inhibition at 100 µg/mL (acetone extract)

Leaves

Ethyl acetate, ethanolic, and acetone extracts

n.s.

20.02% inhibition at 100 µg/mL (ethyl acetate extract)

10.56% inhibition at 100 µg/mL (ethanolic extract)

32.34% inhibition at 100 µg/mL (acetone extract)

Ripe berries and leaves

Turkey

Aqueous

var. alpina

Capacity to inhibit butyrylcholinesterase activity

25.87 (berries) and 25.33% (leaves) inhibition at 50 µg/mL

32.57 (berries) and 44.16% (leaves) inhibition at 100 µg/mL

36.97 (berries) and 62.01% (leaves) inhibition at 200 µg/mL

[47][92]

Hydroethanolic

(80% ethanol, v/v)

43.68 (berries) and 30.31% (leaves) inhibition at 50 µg/mL

45.19 (berries) and 33.17% (leaves) inhibition at 100 µg/mL

47.55 (berries) and 35.33% (leaves) inhibition at 200 µg/mL

Unripe berries

Hydroethanolic

(80% ethanol, v/v)

44.17% inhibition at 50 µg/mL

48.96% inhibition at 100 µg/mL

49.95% inhibition at 200 µg/mL

In vivo assays

Leaves

n.s.

Methanolic

n.s.

Effects on Wistar rats with induced Parkinson’s disease by chlorpromazine for 21 days at a dose of 200 mg/kg

↑↑ locomotor activity

↓↓ motor dysfunctions, including catalepsy and muscle rigidity

[19]

Plant material

India

Methanolic

Effects on Wistar rats with induced catalepsy by reserpine 4 h after juniper treatment at a dose of 200 mg/kg

↓↓ catalepsy activity

[70]

Romania

Essential oil

Effects of juniper volatile oil (1% and 3%) daily inhalation on Amyloid Beta (1–42) male Wistar rat model of Alzheimer’s disease after 21 days of treatment

↑↑ working memory and reference memory errors within radial arm maze task

↓↓ spontaneous alternations percentage within Y-maze task

[112]

Effects of juniper volatile oil (1% and 3%) daily inhalation on Amyloid Beta (1–42)-induced oxidative stress in Wistar rats

↑↑ acetylcholinesterase, superoxide dismutase and catalase activities, and malondialdehyde and protein carbonyl levels

↓↓ glutathione peroxidase-specific activity and the total content of the reduced glutathione

[67]

Hepatoprotective effects

In vivo assays

Leaves

India

Ethyl acetate

n.s.

Effects on Wistar albino rats with hepatic damage caused by paracetamol for 14 days at a dose of 200 mg/kg

↓↓ alkaline phosphatase (−57.41%), direct bilirubin (−30.33%) and total bilirubin (−38.41%), serum alanine aminotransferase (−34.17%), and serum aspartate aminotransferase (−27.58%) when compared to the untreated group

Hepatoprotective effects with rearrangement promotion of portal triads and central veins

[51]

Stems

n.s.

Petroleum ether, chloroform, and ethanol extracts

n.s.

Effects on rats with hepatic damage caused by carbon tetrachloride

Hepatoprotective activity

[113]

Co-combination of berries from juniper and Solanum xanthocarpum

India

Ethanolic

n.s.

Effects on Wistar albino rats with liver toxicity induced by paracetamol and azithromycin for 14 days at a dose of 200 mg/kg

↓↓ serum glutamate oxaloacetate transaminase (−65.4%), serum glutamate pyruvate transaminase (−59.3%), alkaline phosphatase (66.8%), total bilirubin (62.1%), and liver inflammation

Promoting liver tissue’s normal architecture

[3]

Tyrosinase inhibitory activity

In vitro assays

Berries

Republic of Korea

Methanolic

n.s.

Capacity to inhibit tyrosinase activity

about 50% inhibition at 100 µg/mL

[114]

Renal effects

In vivo assay

Berries

Croatia

Aqueous

n.s.

Daily intake of 10% aqueous infusion, 0.1% of oil (with 0.2% Tween 20 solubilizer) by healthy female Wistar rats

↑↑ diuresis and urine excretion without loss of electrolytes

[71]

Antiurolithiasis effects

In vitro assay

Berries

Iran

Hydroethanolic

(50% ethanol, v/v)

n.s.

Capacity to dissolve urinary stone brought out from human kidney at concentrations of 500, 1000, and 2000 µg/mL

Dissolve urinary stones

↓↓ dry powder weight of stones

↑↑ the ratio of calcium oxalate in normal saline aqueous solution plus stone

[115]

Gastrointestinal effects

In vivo assays

Leaves

India

Methanolic

(80% methanol, v/v)

n.s.

Effects on adult male Wistar albino rats with ulcers induced by aspirin, serotonin, indomethacin, alcohol, and stress at doses of 50 and 100 mg/kg

↓↓ aspirin, serotonin, indomethacin, alcohol, and stress-induced gastric ulcerations in rats

↑↑ healing rate of acetic acid-induced ulcers in rats

[72]

n.s.

Effects on pigs with histamine-induced duodenal lesions at doses of 50 and 100 mg/kg

↓↓ histamine-induced duodenal lesions in pigs

Vessels and trachea protective effects in passive smoking

In vitro assays

Berries

Romania

Aerosols

n.s.

Effects of 3-week juniper aerosols (40 min/day) on female Sprague-Dawley rats firstly exposed to daily passive smoking for 6 weeks

↓↓ acetylcholine endothelial-dependent relaxation

[116]

Oil

n.s.

Effects of 3-week juniper nebulization (20 min/day) on the respiratory tract of rats which firstly exposed to 2 cigarettes per day, 5 days a week for 6 weeks

Bronchodilator effects mediated by nitric oxide

[117]

Genotoxicity protective effects

In vitro assays

Berries

Romania

Hydroethanolic

(50% ethanol, v/v)

n.s.

Capacity to exhibit genoprotective effects against aberrations and abnormalities induced by ethanol on root-tip cells of Allium cepa L.

Can effectively protect chromosomes aberrations

[57]

n.s.: not specified; IC50: half-maximal inhibitory concentration ↑↑: increase; ↓↓: reduction.

References

  1. Gezici, S.; Şekeroğlu, N. Current perspectives in the application of medicinal plants against cancer: Novel therapeutic agents. Anticancer Agents Med. Chem. 2019, 19, 101–111.
  2. Benarba, B.; Pandiella, A. Colorectal cancer and medicinal plants: Principle findings from recent studies. Biomed. Pharmacother. 2018, 107, 408–423.
  3. Sen, T.; Samanta, S.K. Medicinal plants, human health and biodiversity: A broad review. In Advances in Biochemical Engineering/Biotechnology; Springer: Berlin/Heidelberg, Germany, 2015; Volume 147, pp. 59–110. ISBN 9783662450963.
  4. Dutra, R.C.; Campos, M.M.; Santos, A.R.S.; Calixto, J.B. Medicinal plants in Brazil: Pharmacological studies, drug discovery, challenges and perspectives. Pharmacol. Res. 2016, 112, 4–29.
  5. Gholipour, S.; Sewell, R.D.E.; Lorigooini, Z.; Rafieian-Kopaei, M. Medicinal plants and atherosclerosis: A review on molecular aspects. Curr. Pharm. Des. 2018, 24, 3123–3131.
  6. Kirichenko, T.V.; Sukhorukov, V.N.; Markin, A.M.; Nikiforov, N.G.; Liu, P.Y.; Sobenin, I.A.; Tarasov, V.V.; Orekhov, A.N.; Aliev, G. Medicinal plants as a potential and successful treatment option in the context of atherosclerosis. Front. Pharmacol. 2020, 11, 1–15.
  7. Akkol, K.E.; Dereli, T.G.F.; Sobarzo-Sánchez, E.; Khan, H. Roles of medicinal plants and constituents in gynecological cancer therapy: Current literature and future directions. Curr. Top. Med. Chem. 2020, 20, 1772–1790.
  8. Singh, G.; Passari, A.K.; Momin, M.D.; Ravi, S.; Singh, B.P.; Kumar, N.S. Ethnobotanical survey of medicinal plants used in the management of cancer and diabetes. J. Tradit. Chin. Med. 2020, 40, 1007–1017.
  9. Naveed, M.; Majeed, F.; Taleb, A.; Zubair, H.M.; Shumzaid, M.; Farooq, M.A.; Baig, M.M.F.A.; Abbas, M.; Saeed, M.; Changxing, L. A review of medicinal plants in cardiovascular disorders: Benefits and risks. Am. J. Chin. Med. 2020, 48, 259–286.
  10. Ajebli, M.; Eddouks, M. Phytotherapy of hypertension: An updated overview. Endocr. Metab. Immune Disord. Drug Targets 2020, 20, 812–839.
  11. Shayganni, E.; Bahmani, M.; Asgary, S.; Rafieian-Kopaei, M. Inflammaging and cardiovascular disease: Management by medicinal plants. Phytomedicine 2016, 23, 1119–1126.
  12. Rouhi-Boroujeni, H.; Heidarian, E.; Rouhi-Boroujeni, H.; Deris, F.; Rafieian-Kopaei, M. Medicinal plants with multiple effects on cardiovascular diseases: A systematic review. Curr. Pharm. Des. 2017, 23, 999–1015.
  13. Unuofin, J.O.; Lebelo, S.L. Antioxidant effects and mechanisms of medicinal plants and their bioactive compounds for the prevention and treatment of type 2 diabetes: An updated review. Oxid. Med. Cell. Longev. 2020, 1356893.
  14. Salehi, B.; Ata, A.; Kumar, N.V.A.; Sharopov, F.; Ramírez-Alarcón, K.; Ruiz-Ortega, A.; Ayatollahi, S.A.; Fokou, P.V.T.; Kobarfard, F.; Zakaria, Z.A.; et al. Antidiabetic potential of medicinal plants and their active components. Biomolecules 2019, 9, 551.
  15. Pathak- Gandhi, N.; Vaidya, A.D.B. Management of Parkinson’s disease in Ayurveda: Medicinal plants and adjuvant measures. J. Ethnopharmacol. 2017, 197, 46–51.
  16. Lemoine, P.; Bablon, J.C.; Da Silva, C. A combination of melatonin, vitamin B6 and medicinal plants in the treatment of mild-to-moderate insomnia: A prospective pilot study. Complement. Ther. Med. 2019, 45, 104–108.
  17. Fejér, J.; Gruľová, D.; Eliašová, A.; Kron, I.; De Feo, V. Influence of environmental factors on content and composition of essential oil from common Juniper ripe berry cones (Juniperus communis L.). Plant Biosyst. 2018, 152, 1227–1235.
  18. Pers-Kamczyc, E.; Tyrała-Wierucka, Ż.; Rabska, M.; Wrońska-Pilarek, D.; Kamczyc, J. The higher availability of nutrients increases the production but decreases the quality of pollen grains in Juniperus communis L. J. Plant Physiol. 2020, 248, 153156.
  19. Bais, S.; Gill, N.S.; Kumar, N. Neuroprotective effect of Juniperus communis on chlorpromazine induced Parkinson disease in animal model. Chin. J. Biol. 2015, 2015, 542542.
  20. Verheyen, K.; Adriaenssens, S.; Gruwez, R.; Michalczyk, I.M.; Ward, L.K.; Rosseel, Y.; Van den Broeck, A.; García, D. Juniperus communis: Victim of the combined action of climate warming and nitrogen deposition? Plant Biol. 2009, 11, 49–59.
  21. Meireles, C.; Pinto-Gomes, C.; Cano, E. Approach to climatophilous vegetation series of Serra da Estrela (Portugal). Acta Bot. Gall. 2012, 159, 283–287.
  22. Rezvani, S.; Rezai, M.A.; Mahmoodi, N. Analysis and antimicrobial activity of the plant Juniperus communis. Rasayan J. Chem. 2009, 2, 257–260.
  23. Bais, S.; Gill, N.S.; Rana, N.; Shandil, S. A phytopharmacological review on a medicinal plant: Juniperus communis. Int. Sch. Res. Not. 2014, 2014, 634723.
  24. Höferl, M.; Stoilova, I.; Schmidt, E.; Wanner, J.; Jirovetz, L.; Trifonova, D.; Krastev, L.; Krastanov, A. Chemical composition and antioxidant properties of juniper berry (Juniperus communis L.) essential oil. Action of the essential oil on the antioxidant protection of saccharomyces cerevisiae model organism. Antioxidants 2014, 3, 81–98.
  25. Raina, R.; Verma, P.K.; Peshin, R.; Kour, H. Potential of Juniperus communis L as a nutraceutical in human and veterinary medicine. Heliyon 2019, 5, e02376.
  26. Ochocka, J.R.; Asztemborska, M.; Zook, D.R.; Sybilska, D.; Perez, G.; Ossicini, L. Enantiomers of monoterpenic hydrocarbons in essential oils from Juniperus communis. Phytochemistry 1997, 44, 869–873.
  27. García, D.; Zamora, R.; Gómez, J.M.; Jordano, P.; Hódar, J.A. Geographical variation in seed production, predation and abortion in Juniperus communis throughout its range in Europe. J. Ecol. 2000, 88, 436–446.
  28. Castro, M.R.; Belo, A.F.; Afonso, A.; Zavattieri, M.A. Micropropagation of Juniperus navicularis, an endemic and rare species from Portugal SW coast. Plant Growth Regul. 2011, 65, 223–230.
  29. Adams, R.P.; Murata, J.; Takahashi, H.; Schwarzbach, A.E. Taxonomy and evolution of Junperus communis: Insight from DNA sequencing and SNPs. Phytologia 2011, 93, 185–197.
  30. Cano, E.; Musarella, C.M.; Cano-Ortiz, A.; Fuentes, J.C.P.; Torres, A.R.; González, S.D.R.; Gomes, C.J.P.; Quinto-Canas, R.; Spampinato, G. Geobotanical study of the microforests of Juniperus oxycedrus subsp. badia in the central and Southern Iberian Peninsula. Sustainability 2019, 11, 1111.
  31. Knyazeva, S.G.; Hantemirova, E.V. Comparative analysis of genetic and morpho-anatomical variability of common Juniper (Juniperus communis L.). Russ. J. Genet. 2020, 56, 48–58.
  32. Ložienė, K.; Rimantas Venskutonis, P. Chapter 56—Juniper (Juniperus communis L.) oils. In Essential Oils in Food Preservation, Flavor and Safety; Academic Press: London, UK, 2016.
  33. García, D.; Zamora, R.; Hódar, J.A.; Gómez, J.M. Age structure of Juniperus communis L. in the Iberian Peninsula: Conservation of remnant populations in Mediterranean mountains. Biol. Conserv. 1999, 87, 215–220.
  34. Sequeira, M.; Espírito-santo, M.D. Checklist da Flora de Portugal; ALFA: Lisboa, Portugal, 2011.
  35. Wilson, M. What Juniper Means? Available online: https://www.restaurantnorman.com/what-juniper-means/ (accessed on 22 December 2021).
  36. Živić, N.; Milošević, S.; Dekić, V.; Dekić, B.; Ristić, N.; Ristić, M.; Sretić, L. Phytochemical and antioxidant screening of some extracts of Juniperus communis L.and Juniperus oxycedrus L. Czech J. Food Sci. 2019, 37, 351–358.
  37. Bento, C.; Gonçalves, A.C.; Silva, B.; Silva, L.R. Peach (Prunus Persica): Phytochemicals and health benefits. Food Rev. Int. 2020; Ahead-of-print.
  38. Pott, D.M.; Osorio, S.; Vallarino, J.G. From central to specialized metabolism: An overview of some secondary compounds derived from the primary metabolism for their role in conferring nutritional and organoleptic characteristics to fruit. Front. Plant Sci. 2019, 10, 835.
  39. Mediavilla, I.; Guillamón, E.; Ruiz, A.; Esteban, L.S. Essential oils from residual foliage of forest tree and shrub species: Yield and antioxidant capacity. Molecules 2021, 26, 3257.
  40. Bento, C.; Gonçalves, A.C.; Jesus, F.; Simões, M.; Silva, L.R. Phenolic compounds: Sources, properties and applications. In Bioactive Compounds: Sources, Properties and Applications; Porter, R., Parker, N., Eds.; Nova Science Publishers, Inc: New York, NY, USA, 2017; pp. 271–299. ISBN 6312317269.
  41. Fatima, N.; Baqri, S.S.R.; Alsulimani, A.; Fagoonee, S.; Slama, P.; Kesari, K.K.; Roychoudhury, S.; Haque, S. Phytochemicals from Indian ethnomedicines: Promising prospects for the management of oxidative stress and cancer. Antioxidants 2021, 10, 1606.
  42. Rabska, M.; Robakowski, P.; Ratajczak, E.; Żytkowiak, R.; Iszkuło, G.; Pers-Kamczyc, E. Photochemistry differs between male and female Juniperus communis L. independently of nutritional availability. Trees Struct. Funct. 2021, 35, 27–42.
  43. Chiorcea-Paquim, A.M.; Enache, T.A.; De Souza Gil, E.; Oliveira-Brett, A.M. Natural phenolic antioxidants electrochemistry: Towards a new food science methodology. Compr. Rev. Food Sci. Food Saf. 2020, 19, 1680–1726.
  44. Singla, R.K.; Dubey, A.K.; Garg, A.; Sharma, R.K.; Fiorino, M.; Ameen, S.M.; Haddad, M.A.; Al-Hiary, M. Natural polyphenols: Chemical classification, definition of classes, subcategories, and structures. J. AOAC Int. 2019, 102, 1397–1400.
  45. Gonçalves, A.C.; Rodrigues, M.; Santos, A.O.; Alves, G.; Silva, L.R. Antioxidant status, antidiabetic properties and effects on Caco-2 cells of colored and non-colored enriched extracts of sweet cherry fruits. Nutrients 2018, 10, 1688.
  46. Olech, M.; Nowak, R.; Ivanova, D.; Tashev, A.; Boyadzhieva, S.; Kalotova, G.; Angelov, G.; Gawlik-Dziki, U. LC-ESI-MS/ MS-MRM profiling of polyphenols and antioxidant activity evaluation of Junipers of different origin. Appl. Sci. 2020, 10, 8921.
  47. Orhan, N.; Orhan, I.E.; Ergun, F. Insights into cholinesterase inhibitory and antioxidant activities of five Juniperus species. Food Chem. Toxicol. 2011, 49, 2305–2312.
  48. Mahmutović, I.; Dahija, S.; Bešta-Gajević, R.; Karalija, E. Biological activity of Juniperus communis L. extracts. In Proceedings of the 28th International Scientific-Expert Conference of Agriculture and Food Industry, Sarajevo, Bosnia and Herzegovina, 27–29 September 2017.
  49. Brodowska, A.J.; Smigielski, K.; Nowak, A.; Czyzowska, A.; Otlewska, A. The impact of ozone treatment in dynamic bed parameters on changes in biologically active substances of juniper berries. PLoS ONE 2015, 10, e0144855.
  50. Luís, Â.; Domingues, F.; Duarte, A.P. Bioactive compounds, RP-HPLC analysis of phenolics, and antioxidant activity of some Portuguese shrub species extracts. Nat. Prod. Commun. 2011, 6, 1863–1872.
  51. Ved, A.; Gupta, A.; Rawat, A. Antioxidant and hepatoprotective potential of phenol-rich fraction of Juniperus communis Linn. leaves. Pharmacogn. Mag. 2017, 13, 108–113.
  52. Orhan, N.; Hoşbaş, S.; Orhan, D.D.; Aslan, M.; Ergun, F. Enzyme inhibitory and radical scavenging effects of some antidiabetic plants of Turkey. Iran. J. Basic Med. Sci. 2014, 17, 426–432.
  53. Fejér, J.; Kron, I.; Grul’ová, D.; Eliašová, A. Seasonal variability of Juniperus communis L. berry ethanol extracts: 1. In vitro hydroxyl radical scavenging activity. Molecules 2020, 25, 4114.
  54. Tang, J.; Dunshea, F.R.; Suleria, H.A.R. LC-ESI-QTOF/MS characterization of phenolic compounds from medicinal plants (Hops and Juniper Berries) and their antioxidant activity. Foods 2020, 9, 7.
  55. Harris, C.S.; Cuerrier, A.; Lamont, E.; Haddad, P.S.; Arnason, J.T.; Bennett, S.A.L.; Johns, T. Investigating wild berries as a dietary approach to reducing the formation of advanced glycation endproducts: Chemical correlates of in vitro antiglycation activity. Plant Foods Hum. Nutr. 2014, 69, 71–77.
  56. Miceli, N.; Trovato, A.; Dugo, P.; Cacciol, F.; Donato, P.; Marino, A.; Bellinghieri, V.; La Barbera Tommaso, M.; Güvenç, A.; Taviano, M.F. Comparative analysis of flavonoid profile, antioxidant and antimicrobial activity of the berries of Juniperus communis L. var. communis and Juniperus communis L. var. saxatilis Pall. from Turkey. J. Agric. Food Chem. 2009, 57, 6570–6577.
  57. Fierascu, I.; Ungureanu, C.; Avramescu, S.M.; Cimpeanu, C.; Georgescu, M.I.; Fierascu, R.C.; Ortan, A.; Sutan, A.N.; Anuta, V.; Zanfirescu, A.; et al. Genoprotective, antioxidant, antifungal and anti-inflammatory evaluation of hydroalcoholic extract of wild-growing Juniperus communis L. (Cupressaceae) native to Romanian southern sub-Carpathian hills. BMC Complement. Altern. Med. 2018, 18, 3.
  58. Gao, H.W.; Huang, X.F.; Yang, T.P.; Chang, K.F.; Yeh, L.W.; Hsieh, M.C.; Weng, J.C.; Tsai, N.M. Juniperus communis suppresses melanoma tumorigenesis by inhibiting tumor growth and inducing apoptosis. Am. J. Chin. Med. 2019, 47, 1171–1191.
  59. Elboughdiri, N.; Ghernaout, D.; Kriaa, K.; Jamoussi, B. Enhancing the extraction of phenolic compounds from juniper berries using the box-behnken design. ACS Omega 2020, 5, 27990–28000.
  60. Xavier, V.; Finimundy, T.C.; Heleno, S.A.; Amaral, J.S.; Calhelha, R.C.; Vaz, J.; Pires, T.C.S.P.; Mediavilla, I.; Esteban, L.S.; Ferreira, I.C.F.R.; et al. Chemical and bioactive characterization of the essential oils obtained from three Mediterranean plants. Molecules 2021, 26, 7472.
  61. Kılıç, Ö.; Kocak, A. Volatile constituents of Juniperus communis L., Taxus canadensis Marshall. and Tsuga canadensis (L.) Carr. from Canada. J. Agric. Sci. Technol. B 2014, 4, 135–140.
  62. Johnson, W. Final report on the safety assessment of Juniperus communis extract, Juniperus oxycedrus extract, Juniperus oxycedrus tar, Juniperus phoenicea extract, and Juniperus virginiana extract. Int. J. Toxicol. 2001, 20, 41–56.
  63. Zheljazkov, V.D.; Astatkie, T.; Jeliazkova, E.A.; Heidel, B.; Ciampa, L. Essential oil content, composition and bioactivity of Juniper species in Wyoming, United States. Nat. Prod. Commun. 2017, 12, 201–204.
  64. Rajcevic, N.; Dodos, T.; Novakovic, J.; Janackovic, P.; Marin, P. Essential oil composition and antioxidant activity of two Juniperus communis L. varieties growing wild in Serbia. Zb. Matice Srp. Prir. Nauke 2016, 131, 197–205.
  65. Gonny, M.; Cavaleiro, C.; Salgueiro, L.; Casanova, J. Analysis of Juniperus communis subsp. alpina needle, berry, wood and root oils by combination of GC, GC/MS and 13C-NMR. Flavour Fragr. J. 2006, 21, 99–106.
  66. Vasilijević, B.; Knežević-Vukčević, J.; Mitić-Ćulafić, D.; Orčić, D.; Francišković, M.; Srdic-Rajic, T.; Jovanović, M.; Nikolić, B. Chemical characterization, antioxidant, genotoxic and in vitro cytotoxic activity assessment of Juniperus communis var. saxatilis. Food Chem. Toxicol. 2018, 112, 118–125.
  67. Cioanca, O.; Hancianu, M.; Mihasan, M.; Hritcu, L. Anti-acetylcholinesterase and antioxidant activities of inhaled Juniper oil on amyloid beta (1–42)-induced oxidative stress in the rat hippocampus. Neurochem. Res. 2015, 40, 952–960.
  68. Laouar, A.; Klibet, F.; Bourogaa, E.; Benamara, A.; Boumendjel, A.; Chefrour, A.; Messarah, M. Potential antioxidant properties and hepatoprotective effects of Juniperus phoenicea berries against CCl4 induced hepatic damage in rats. Asian Pac. J. Trop. Med. 2017, 10, 263–269.
  69. Li, C.Y.; Lee, S.C.; Lai, W.L.; Chang, K.F.; Huang, X.F.; Hung, P.Y.; Lee, C.P.; Hsieh, M.C.; Tsai, N.M. Cell cycle arrest and apoptosis induction by Juniperus communis extract in esophageal squamous cell carcinoma through activation of p53-induced apoptosis pathway. Food Sci. Nutr. 2021, 9, 1088–1098.
  70. Bais, S.; Gill, N.S.; Rana, N. Effect of Juniperus communis extract on reserpine induced catalepsy. Inventi Impact Ethnopharmacol. 2014, 2014, 1–4.
  71. Stanić, G.; Samaržija, I.; Blažević, N. Time-dependent diuretic response in rats treated with Juniper berry preparations. Phyther. Res. 1998, 12, 494–497.
  72. Pramanik, K.C.; Biswas, R.; Bandyopadhyay, D.; Mishra, M.; Ghost, C.; Chatterjee, T.K. Evaluation of anti-ulcer properties of the leaf extract of Juniperus communis L. in animals. J. Nat. Remedies 2007, 7, 207–213.
  73. Schneider, I.; Gibbons, S.; Bucar, F. Inhibitory activity of Juniperus communis on 12(S)-HETE production in human platelets. Planta Med. 2004, 70, 471–474.
  74. Filipowicz, N.; Kamiński, M.; Kurlenda, J.; Asztemborska, M.; Ochocka, J.R. Antibacterial and antifungal activity of juniper berry oil and its selected components. Phyther. Res. 2003, 17, 227–231.
  75. Cabral, C.; Francisco, V.; Cavaleiro, C.; Gonçalves, M.J.; Cruz, M.T.; Sales, F.; Batista, M.T.; Salgueiro, L. Essential oil of Juniperus communis subsp alpina (Suter) Čelak needles: Chemical composition, antifungal activity and cytotoxicity. Phyther. Res. 2012, 26, 1352–1357.
  76. Angioni, A.; Barra, A.; Russo, M.T.; Coroneo, V.; Dessí, S.; Cabras, P. Chemical composition of the essential oils of Juniperus from ripe and unripe berries and leaves and their antimicrobial activity. J. Agric. Food Chem. 2003, 51, 3073–3078.
  77. Klančnik, A.; Zorko, Š.; Toplak, N.; Kovač, M.; Bucar, F.; Jeršek, B.; Smole Možina, S. Antiadhesion activity of Juniper (Juniperus communis L.) preparations against Campylobacter jejuni evaluated with PCR-based methods. Phyther. Res. 2018, 32, 542–550.
  78. Salamon, I.; Kryvtsova, M.; Bucko, D.; Tarawneh, A.H. Chemical characterization and antimicrobial activity of some essential oils after their industrial large-scale distillation. J. Microbiol. Biotechnol. Food Sci. 2019, 8, 984–988.
  79. Falcão, S.; Bacém, I.; Igrejas, G.; Rodrigues, P.J.; Vilas-Boas, M.; Amaral, J.S. Chemical composition and antimicrobial activity of hydrodistilled oil from Juniper berries. Ind. Crops Prod. 2018, 124, 878–884.
  80. Pepeljnjak, S.; Kosalec, I.; Kalodera, Z.; Blažević, N. Antimicrobial activity of juniper berry essential oil (Juniperus communis L., Cupressaceae). Acta Pharm. 2005, 55, 417–422.
  81. Nikolić, B.; Vasilijević, B.; Ćirić, A.; Mitić-Ćulafić, D.; Cvetković, S.; Džamić, A.; Knežević-Vukčević, J. Bioactivity of Juniperus communis essential oil and post-distillation waste: Assessment of selective toxicity against food contaminants. Arch. Biol. Sci. 2019, 71, 235–244.
  82. Darwish, R.S.; Hammoda, H.M.; Ghareeb, D.A.; Abdelhamid, A.S.A.; Bellah El Naggar, E.M.; Harraz, F.M.; Shawky, E. Efficacy-directed discrimination of the essential oils of three Juniperus species based on their in-vitro antimicrobial and anti-inflammatory activities. J. Ethnopharmacol. 2020, 259, 112971.
  83. Peruč, D.; Broznić, D.; Maglica, Ž.; Marijanović, Z.; Karleuša, L.; Gobin, I. Biofilm degradation of nontuberculous mycobacteria formed on stainless steel following treatment with immortelle (Helichrysum italicum) and common juniper (Juniperus communis) essential oils. Processes 2021, 9, 362.
  84. Peruč, D.; Tićac, B.; Broznić, D.; Maglica, Ž.; Šarolić, M.; Gobin, I. Juniperus communis essential oil limit the biofilm formation of Mycobacterium avium and Mycobacterium intracellulare on polystyrene in a temperature-dependent manner. Int. J. Environ. Health Res. 2020, 32, 144–154.
  85. Ferrazzano, G.F.; Roberto, L.; Catania, M.R.; Chiaviello, A.; De Natale, A.; Roscetto, E.; Pinto, G.; Pollio, A.; Ingenito, A.; Palumbo, G. Screening and scoring of antimicrobial and biological activities of Italian vulnerary plants against major oral pathogenic bacteria. Evid. Based Complement. Altern. Med. 2013, 316280.
  86. Kose, M.; Bayraktar, O.; Balta, A. Antioxidant and Antimicrobial Activities of extracts from some selected Mediterranean plant species. Int. J. New Technol. Res. 2016, 2, 263506.
  87. Miceli, N.; Marino, A.; Köroğlu, A.; Cacciola, F.; Dugo, P.; Mondello, L.; Taviano, M.F. Comparative study of the phenolic profile, antioxidant and antimicrobial activities of leaf extracts of five Juniperus L. (Cupressaceae) taxa growing in Turkey. Nat. Prod. Res. 2020, 34, 1636–1641.
  88. Dziedzinski, M.; Kobus-Cisowska, J.; Szymanowska, D.; Stuper-Szablewska, K.; Baranowska, M. Identification of polyphenols from coniferous shoots as natural antioxidants and antimicrobial compounds. Molecules 2020, 25, 3527.
  89. Marino, A.; Bellinghieri, V.; Nostro, A.; Miceli, N.; Taviano, M.F.; Güvenç, A.Ş.; Bisignano, G. In vitro effect of branch extracts of Juniperus species from Turkey on Staphylococcus aureus biofilm. FEMS Immunol. Med. Microbiol. 2010, 59, 470–476.
  90. Rolta, R.; Kumar, V.; Sourirajan, A.; Upadhyay, N.K.; Dev, K. Phytocompounds of three medicinal plants (Juniperus communis, Urtica doica and Coleus forskohilii) of North West Himalayas increases the potency of antibacterial and antiffungal antibiotics. Plant Arch. 2020, 20, 481–489.
  91. Mllhau, G.; Valentin, A.; Benoit, F.; Mallié, M.; Bastide, J.M.; Pélissier, Y.; Bessière, J.M. In vitro antimalarial activity of eight essential oils. J. Essent. Oil Res. 1997, 9, 329–333.
  92. Senol, F.S.; Orhan, I.E.; Ustun, O. In vitro cholinesterase inhibitory and antioxidant effect of selected coniferous tree species. Asian Pac. J. Trop. Med. 2015, 8, 269–275.
  93. Tunón, H.; Olavsdotter, C.; Bohlin, L. Evaluation of anti-inflammatory activity of some Swedish medicinal plants. Inhibition of prostaglandin biosynthesis and PAF-induced exocytosis. J. Ethnopharmacol. 1995, 48, 61–76.
  94. Mascolo, N.; Autore, G.; Capasso, F.; Menghini, A.; Fasulo, M.P. Biological screening of Italian medicinal plants for anti-inflammatory activity. Phyther. Res. 1987, 1, 28–31.
  95. Akkol, E.K.; Güvenç, A.; Yesilada, E. A comparative study on the antinociceptive and anti-inflammatory activities of five Juniperus taxa. J. Ethnopharmacol. 2009, 125, 330–336.
  96. Banerjee, S.; Mukherjee, A.; Chatterjee, T.K. Evaluation of analgesic activities of methanolic extract of medicinal plant Juniperus communis Linn. Int. J. Pharm. Pharm. Sci. 2012, 4, 547–550.
  97. Gallagher, A.M.; Flatt, P.R.; Duffy, G.; Abdel-Wahab, Y.H.A. The effects of traditional antidiabetic plants on in vitro glucose diffusion. Nutr. Res. 2003, 23, 413–424.
  98. Swanston-Flatt, S.K.; Day, C.; Bailey, C.J.; Flat, P.R. Traditional plant treatments for diabetes. Studies in normal and streptozotocin diabetic mice. Diabetologia 1990, 33, 462–464.
  99. Medina, F.S.; Gamez, M.J.; Jimenez, I.; Jimenez, J.; Osuna, J.I.; Zarzuelo, A. Hypoglycemic activity of Juniper “berries. ” Planta Med. 1994, 60, 197–200.
  100. Akdogan, M.; Koyu, A.; Ciris, M.; Yildiz, K. Anti-hypercholesterolemic activity of Juniperus communis Lynn oil in rats: A biochemical and histopathological investigation. Biomed. Res. 2012, 23, 321–328.
  101. Banerjee, S.; Singh, H.; Chatterjee, T.K. Evaluation of anti-diabetic and anti-hyperlipidemic potential of methanolic extract of Juniperus communis (L.) in streptozotocinnicotinamide induced diabetic rats. Int. J. Pharma Bio Sci. 2013, 4, 10–17.
  102. Petlevski, R.; Hadžija, M.; Slijepčevič, M.; Juretič, D. Effect of “antidiabetis” herbal preparation on serum glucose and fructosamine in NOD mice. J. Ethnopharmacol. 2001, 75, 181–184.
  103. Lee, C.C.; Hsiao, C.Y.; Lee, S.C.; Huang, X.F.; Chang, K.F.; Lee, M.S.; Hsieh, M.C.; Tsai, N.M. Suppression of oral cancer by induction of cell cycle arrest and apoptosis using Juniperus communis extract. Biosci. Rep. 2020, 40, BSR20202083.
  104. Sahin Yaglioglu, A.; Eser, F. Screening of some Juniperus extracts for the phenolic compounds and their antiproliferative activities. S. Afr. J. Bot. 2017, 113, 29–33.
  105. Ghaly, N.S.; Mina, S.A.; Younis, N.A.H. In vitro cytotoxic activity and phytochemical analysis of the aerial parts of J. Communis L. cultivated in Egypt. J. Pharm. Sci. Res. 2016, 8, 128–131.
  106. Van Slambrouck, S.; Daniels, A.L.; Hooten, C.J.; Brock, S.L.; Jenkins, A.R.; Ogasawara, M.A.; Baker, J.M.; Adkins, G.; Elias, E.M.; Agustin, V.J.; et al. Effects of crude aqueous medicinal plant extracts on growth and invasion of breast cancer cells. Oncol. Rep. 2007, 17, 1487–1492.
  107. Fernandez, A.; Cock, I.E. The therapeutic properties of Juniperus communis L.: Antioxidant capacity, bacterial growth inhibition, anticancer activity and toxicity. Pharmacogn. J. 2016, 8, 273–280.
  108. Huang, N.C.; Huang, R.L.; Huang, X.F.; Chang, K.F.; Lee, C.J.; Hsiao, C.Y.; Lee, S.C.; Tsai, N.M. Evaluation of anticancer effects of Juniperus communis extract on hepatocellular carcinoma cells in vitro and in vivo. Biosci. Rep. 2021, 41, BSR20211143.
  109. Lai, W.L.; Lee, S.C.; Chang, K.F.; Huang, X.F.; Li, C.Y.; Lee, C.J.; Wu, C.Y.; Hsu, H.J.; Tsai, N.M. Juniperus communis extract induces cell cycle arrest and apoptosis of colorectal adenocarcinoma in vitro and in vivo. Braz. J. Med. Biol. Res. 2021, 54, 1–11.
  110. Elshafie, H.S.; Caputo, L.; De Martino, L.; Gruľová, D.; Zheljazkov, V.Z.; De Feo, V.; Camele, I. Biological investigations of essential oils extracted from three Juniperus species and evaluation of their antimicrobial, antioxidant and cytotoxic activities. J. Appl. Microbiol. 2020, 129, 1261–1271.
  111. Tsai, W.C.; Tsai, N.M.; Chang, K.F.; Wang, J.C. Juniperus communis extract exerts antitumor effects in human glioblastomas through blood-brain barrier. Cell. Physiol. Biochem. 2018, 49, 2443–2462.
  112. Cioanca, O.; Mircea, C.; Trifan, A.; Aprotosoaie, A.C.; Hritcu, L.; Hǎncianu, M. Improvement of amyloid-β-induced memory deficits by Juniperus communis L. volatile oil in a rat model of Alzheimer’s disease. Farmacia 2014, 62, 514–520.
  113. Mavin; Garg, G.P. Screening and evaluation of pharmacognostic, phytochemical and hepatoprotective activity of J. communis L. stems. Int. J. Pharma Bio Sci. 2010, 1.
  114. Jegal, J.; Park, S.A.; Chung, K.W.; Chung, H.Y.; Lee, J.; Jeong, E.J.; Kim, K.H.; Yang, M.H. Tyrosinase inhibitory flavonoid from Juniperus communis fruits. Biosci. Biotechnol. Biochem. 2016, 80, 2311–2317.
  115. Barzegarnejad, A.; Azadbakht, M.; Emadian, O.; Ahmadi, M. Effect of some fractions of the extract of Juniperus communis fruit on solving kidney stones in vitro. J. Maz. Univ. Med. Sci. 2013, 23, 145–152.
  116. Sturza, A.; Pleşa, C.; Ordodi, V.; Noveanu, L.; Mirica, N.; Fira-Mladinescu, O.; Lupea, A.X.; Muntean, D. Vascular responses of Juniperus communis aerosols in aortic rings isolated from rats subjected to passive smoking. Ann. Rom. Soc. Cell Biol. 2011, 16, 178–181.
  117. Pleşa, C.; Ordodi, V.L.; Noveanu, L.; Vasile, L.; Ardelean, A.; Lupea, A.X. Effects of Juniperus communis aerosols on trachea and lung from rat subjected to passive smoking. Stud. Univ. Vasile Goldis Ser. Stiint. Vietii Life Sci. Ser. 2011, 21, 319–328.
More
Information
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 1.1K
Revisions: 3 times (View History)
Update Date: 24 Mar 2022
1000/1000