Next Article in Journal
Burnout, Goal Orientation and Academic Performance in Adolescent Students
Next Article in Special Issue
Comparative Study of the Restorative Effects of Forest and Urban Videos during COVID-19 Lockdown: Intrinsic and Benchmark Values
Previous Article in Journal
Female Human Papillomavirus Infection Associated with Increased Risk of Infertility: A Nationwide Population-Based Cohort Study
Previous Article in Special Issue
Effects of a Forest Walk on Urinary Dityrosine and Hexanoyl-Lysine in Young People: A Pilot Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Forest Volatile Organic Compounds and Their Effects on Human Health: A State-of-the-Art Review

1
Terme di Monticelli, 43022 Monticelli Terme PR, Italy
2
Institute of Public Health, University of Parma, 43125 Parma PR, Italy
3
CERFIT, Careggi University Hospital, 50139 Firenze FI, Italy
4
AUSL-IRCCS Reggio Emilia, 42122 Reggio Emilia RE, Italy
5
Binini Partners S.r.l. Engineering and Architecture, 42121 Reggio Emilia RE, Italy
6
European Herbal and Traditional Medicine Practitioners Association (EHTPA), Norwich NR3 1HG, UK
*
Author to whom correspondence should be addressed.
Int. J. Environ. Res. Public Health 2020, 17(18), 6506; https://0-doi-org.brum.beds.ac.uk/10.3390/ijerph17186506
Submission received: 9 August 2020 / Revised: 3 September 2020 / Accepted: 4 September 2020 / Published: 7 September 2020

Abstract

:
The aim of this research work is to analyze the chemistry and diversity of forest VOCs (volatile organic compounds) and to outline their evidence-based effects on health. This research work was designed as a narrative overview of the scientific literature. Inhaling forest VOCs like limonene and pinene can result in useful antioxidant and anti-inflammatory effects on the airways, and the pharmacological activity of some terpenes absorbed through inhalation may be also beneficial to promote brain functions by decreasing mental fatigue, inducing relaxation, and improving cognitive performance and mood. The tree composition can markedly influence the concentration of specific VOCs in the forest air, which also exhibits cyclic diurnal variations. Moreover, beneficial psychological and physiological effects of visiting a forest cannot be solely attributed to VOC inhalation but are due to a global and integrated stimulation of the five senses, induced by all specific characteristics of the natural environment, with the visual component probably playing a fundamental role in the overall effect. Globally, these findings can have useful implications for individual wellbeing, public health, and landscape design. Further clinical and environmental studies are advised, since the majority of the existing evidence is derived from laboratory findings.

Graphical Abstract

1. Introduction

1.1. Forest Volatile Organic Compounds (VOCs): Background and Natural Functions

The first systematic analysis of the importance and role of volatile organic compounds (VOCs) emitted by plants in the atmosphere above forests was probably performed from the perspective of atmospheric chemists who were interested in the effects of VOCs on atmospheric composition and climate. In two seminal articles [1,2], the Dutch scientist Fritz Went and his colleagues hypothesized that the blue haze observed above many forests was caused at least partially by volatile organic compounds emitted by plants (forest VOCs) and went on to estimate the impact that biogenic emissions have on the global balance of organic compounds released into the atmosphere, compared to anthropogenic production. The first estimate of global terpene emissions was 175 millions of tons C/year [1], but more recent evaluations state that worldwide biogenic VOC (BVOC) emissions into the atmosphere could amount to around 1 billion of tons C/year [3]. Forests seem to be the greatest BVOC emitters. Specifically, tropical trees appear to be responsible for around 80% of terpenoid emissions and for around 50% of other BVOC emissions, while other tree species seem to contribute to around 10% of total BVOC emissions [3]. BVOCs form 80% to 90% of total VOC emissions in the atmosphere each year [4,5].
Some forest VOCs are also defined as “phytoncides” and this term, first introduced in the last century by the Russian biologist Boris Petrovich Tokin, derives from the combination of two words: “phyton”, which refers to plants and botany in ancient Greek, plus the suffix “cide”, which indicates “killing” or “exterminating”, thus underscoring the antimicrobial and insecticidal activity of these substances [6,7]. However, the term “phytoncide” may be misleading because, due its broad definition and etymology, it can indicate either plant-derived volatile substances with antiparasitic properties or any (volatile and nonvolatile) antimicrobial/insecticidal compound released by plants, any volatile VOC, or, in some cases, essential oils obtained from aromatic woods. In order to avoid potential misunderstandings, in this research work, which is focused on biogenic volatile substances found in forest air, the term “forest VOCs” will be preferred over the less specific “phytoncide”, and BVOCs will be defined as any organic compound, except carbon dioxide and monoxide, emitted by plants and having vapor pressure high enough to be vaporized in relevant amounts. Usually, the definition excludes dimethyl sulfide and methane because it is still debated as to whether they are produced by terrestrial plants [8].
More than 1000 different BVOCs are released from plant flowers, vegetative parts, or roots [9]. These substances are largely lipophilic products with molecular masses under 300 Da, and the vast majority are isoprenoids, including hemiterpenes (C5H8) such as isoprene, monoterpenes (C10H16), irregular acyclic homoterpenes (C11H18 or C16H26), and sesquiterpenes (C15H24), both as hydrocarbons and as oxygenated compounds; there is also a number of low-molecular-weight molecules, especially C1 and C2 oxygenated compounds, such as methanol and acetaldehyde, and C6H10 fatty acid derivatives, including lipoxygenase pathway products such as green leaf volatiles (henceforth GLVs), plus benzenoids and phenylpropanoids [5,9,10,11,12,13].
Many BVOCs are common to phylogenetically distant plant species [14]. For instance, only some plant species, even belonging to taxonomically distant taxa (like Bryophyta and Quercus), emit isoprene, while in close taxa, there can be both emitters (Quercus spp.) and non-emitters (Acer spp.) [11], thus suggesting that metabolic pathways related to their production have been highly preserved during evolution [14].
From the perspective of plant physiology, BVOCs can be divided into constitutive and inducible compounds. All plants can potentially synthesize volatile isoprenoids if triggered by abiotic and biotic stress factors, but only some species can do this constitutively [15].
  • Constitutive forest VOCs are already synthesized in the organism and either stored in specialized structures or constantly emitted without any form of storage [16]. They are emitted at baseline levels and are mainly made of terpenoids, plus shikimate derivatives and polyketides [5].
  • Inducible forest VOCs (herbivore-induced plant volatiles; henceforth HIPVs) are compounds whose synthesis is increased or initiated de-novo after herbivore attacks but also after stimulation by abiotic stressors. They have some metabolic costs but they make the plant phenotypically plastic and herbivore adaptation more unlikely to occur [5].
HIPVs comprise isoprenoids, products of the lipoxygenase (LOX) pathway, such as GLVs, some carotenoid derivatives, indoles and phenolics, and phytohormones such as ethylene, jasmonic acid, and others [17,18]. It is hypothesized that forest VOCs are not only involved in some plant-related physiological functions (biogenic stress response, adaptation to climate changes), but they also have a central role in forest ecosystems (interplant communication, antimicrobial and insecticidal activity against parasites, influence on animals’ feeding behaviors as well as on underwood environmental conditions) [19]. Thus, forest VOCs can have several functions for plant physiology [18]; the most important of them are summarized in Table 1 and Figure 1.

1.2. Non-Tree-Derived Forest BVOCs

In temperate forests, the canopy layer of trees acts as the largest emission source of BVOCs [19] and, in general, from a quantitative point of view, plant leaves with their canals, oil glands, and glandular trichomes are the major emitters of these volatile compounds [8,10]. However, all plant organs and tissues produce BVOCs: leaves, flowers, and fruits release them into the atmosphere, whereas roots secrete them into the soil. Moreover, wood, phloem, and bark of trunks can serve as pools of stored BVOCs [19].
Nevertheless, BVOCs are not exclusively tree-derived: soil bacteria, mycorrhizal fungi, and other rhizosphere microbes can also emit BVOCs, but it is not easy to evaluate the exact contribution of these non-plant sources of BVOCs in the soil or their role in the rhizosphere. This is because their emissions cannot be easily separated from those of the roots [19] and because of experimental difficulties [8]. These limits notwithstanding, one study showed that the emission of sesquiterpenes (in particular, (–)-thujopsene) by fungi (Laccaria bicolor) can interact with the ability of Populus trees to develop their root system (in particular, with the proliferation of their lateral roots) [20]. One research article reported that, in the Amazonian rainforest, soil microorganisms can emit large amounts of sesquiterpenes, and these emissions strongly influence atmospheric chemistry near the soil surface and under the canopy, to an extent comparable with canopy emissions themselves [21]. Šimpraga and colleagues reported that there is no similar assessment for temperate and boreal forests, but there are limited data on bacterial VOC profiles (rich in alkenes, alcohols, ketones, and terpenes) and fungal VOC profiles (dominated by alcohols, benzenoids, aldehydes, and ketones) [19]. Apart from soil-derived emission, other, quantitatively less important, BVOC emissions in the forest can originate from understorey vegetation. In an article relative to subarctic forests of Betula pubescens ssp. czerepanovii (mountain birch), the authors found that BVOCs emitted by the understorey vegetation of Rhododendron tomentosum are adsorbed by tree stands growing above them, thus affecting the volatile emission profile of mountain birches [22].
Overall, as suggested by these limited data, trees are considered as the major emitters of BVOCs in temperate forests and the contribution of non-tree emitters is still difficult to properly characterize in sufficient detail, thus probably having a more indirect effect on the composition of the forest air. For these reasons, it was decided to focus this review on tree-derived forest VOCs.

1.3. Research Aim

The primary aim of this research work is to outline evidence-based physiological effects of forest VOCs in order to evaluate their potential integrated action on health. A secondary aim is to propose strategies to effectively and sustainably harness any of their beneficial effects in real-life contexts. Among all forest VOCs, it has been decided to focus our attention on pinene and limonene due to their wide uses and applications, as well as to the fact that their effects on health have been better studied [23,24]. The chemistry and diversity of forest VOCs (volatile organic compounds) is also analyzed for better comprehensiveness.

2. Materials and Methods

This research work was designed as an overview of the scientific literature with a critical discussion of retrieved evidence [25]. General recommendations to improve the quality of narrative reviews were taken into account for better structuring this research [26,27]. Scientific databases like Medline, PsycINFO, Cochrane Library, and Google Scholar were searched for articles on the topic with relevant keywords, including “volatile organic compound*”, “forest air”, “forest atmosphere”, “forest bathing”, “phytoncide*”, “pinene”, “limonene”, “psycho*”, “neuro*”, “endocrine*”, “hormone*” “immune*”, “prevention”, “inflammation”, “stress”, and “health”. The search yielded 4159 results (1211 from Medline, 200 from PsycINFO, 158 from Cochrane Library, and 2590 from Google Scholar) and, after article screening, 147 studies were considered eligible to be included and discussed in this review. Considering that evidence on the topic is limited, cross-disciplinary, and highly heterogeneous, it was decided to structure the article as a narrative qualitative synthesis of the scientific literature in order to provide the reader with a state-of-the art dynamic description of the topic, stemming from a combination of retrieved evidence with the specific professional background and expertise of each author. Beyond the basis of available evidence, it was decided to collect and discuss ideas, hypotheses, current examples, and future perspectives for research and practice. In particular, the focus on health effects of forest VOCs (especially limonene and pinene) led to highlighting their potential activity on stress and inflammation. Finally, three levels of discussion were considered, with a wide range of implications varying from clinical activities to policymaking: individual wellbeing, public health, and landscape design.

3. Results

3.1. Biochemistry of Forest VOCs

Forest VOCs are the product of many different metabolic pathways: the isoprenoids are the most important group of metabolites in terms of diversity of compounds and quantities of emissions, but the products of the LOX pathway as well as shikimate derivatives are also highly relevant. Main biosynthetic pathways for BVOCs are graphically displayed in Figure 2.

3.1.1. Isoprenoids

The volatile isoprenoids include isoprene (C5), monoterpenes (C10), homoterpenes (C11 or C16), and sesquiterpenes (C15), and they all derive from the precursor dimethylallyl diphosphate (DMAPP) and its isomer isopentenyl diphosphate (IPP). These precursors are synthesized by two separate pathways, one active in plastids (the deoxyxylulose-5-phosphate (DXP), also known as the methylerythritol phosphate (MEP) pathway), and the second in the cytosol of the secretory cells themselves (the mevalonate (MVA) pathway) [17]. In fact, IPP and DMAPP seem to originate in the plastids from the MEP pathway, while leucoplasts contain the enzymes for the first steps of monoterpene biosynthesis. In the MVA pathway, DMAPP accepts one IPP (by prenyltransferase—PT), leading to the production of farnesyl diphosphate (FPP) and, consequently, to sesquiterpenoids (although, in some instances, there is a very small production of monoterpenes) and to the homoterpene DMTT. In the MEP pathway, the hemiterpene isoprene is synthesized from DMAPP alone by terpene synthase or cyclase (TPS), and the fusion of DMAPP to one or more IPPs leads to the production of geranyl diphosphate (GPP) (precursor of the monoterpenoids) and geranyl geranyl diphosphate (GGPP), which leads to the synthesis of diterpenes from which the homoterpene TMTT derives. Although the MEP pathway is plastid-located, the enzymes necessary for sesquiterpene synthesis are cytosolic. Hence, MEP-derived IPP or DMAPP need to travel from plastids to the cytosol [17,18]. The biosynthesis of the homoterpenes DMNT and TMTT follows a less direct pathway. DMNT derives from the oxidative degradation of (E)-nerolidol (a C15-alcohol), and TMTT from the oxidative degradation of (E,E)-geranyl linalool (a C20-alcohol) [28].

3.1.2. Oxylipins

Oxylipins are products of the lipoxygenase (LOX) pathway. They derive from the polyunsaturated C18 fatty acids of the chloroplast membrane (such as 13-hydroperoxylinolenic acid), which, in times of stress or damage, are cleaved by 9-LOX and 13-LOX pathways to give highly reactive products like 6-carbon aldehydes and alcohols (GLVs) and derivatives of jasmonic acid such as methyl jasmonate [9].

3.1.3. Shikimate Pathway

Another important group of BVOCs consists of compounds containing an aromatic ring, such as indole (derived from reactions of modified amino acids or their precursors) or the derivatives of phenylalanine, either via the phenylpropanoid pathway (eugenol and methyl chavicol) or the benzoate pathway (methyl salicylate) [17] (Figure 2).

3.2. A Qualitative and Quantitative Analysis of Forest VOC Emissions

The composition of forest VOCs (probably the key VOC source [11]) is expected to consist mainly of constitutive compounds, but, since it cannot be excluded that, at any given moment, some individual plants in the forest may be under herbivore attack or abiotic stress, and provided that the rate of VOC emissions can be many times higher than baseline after a herbivore attack [29], a part of the total forest emissions can consist of biotic and abiotic-induced VOCs. This can be important for the evaluation of biological activities of forest VOCs because the range of biotic-induced compounds is often highly different from that of constitutive ones and can be also different from the abiotic-induced ones [29,30]. If seen from the plants’ functional point of view, VOCs thus comprise all volatile constitutive defense compounds (volatile phytoanticipins, or phytoncides, as defined by Tokin), plus volatile inducible defense compounds (volatile phytoalexins or HIPVs) and non-defense compounds (attractants of pollinator and of seed dispersals).
The biochemical composition of forest air tends to follow diurnal variations, with BVOCs concentration usually peaking early in the afternoon and, especially in summertime, even in the morning (around 2 h after dawn), and it can be markedly influenced by several factors, including meteorological conditions, altitude, seasons, sunlight exposure, and tree types [31,32]. For example, it has been demonstrated that the air of forests dominated by conifer trees has a higher concentration of VOCs if compared to that of forests with more deciduous trees [31]. However, beyond these differences, it is estimated that, from a biochemical point of view, the majority of forest VOCs are isoprene and terpenes, especially monoterpenes, including limonene or pinene [33]. Specifically, according to Guenther and colleagues [3], isoprene dominates the emissions, with around 50% of the total BVOC emissions of 1 billion tons C/year. C1 and C2 oxygenated compounds such as methanol and acetaldehyde and C10 hydrocarbons such as α-pinene, β-pinene, (E)-β-ocimene, and d-limonene contribute to another 30%, while the remaining 20% is dominated by 20 compounds (mostly terpenoids), with the other 100 compounds accounting for a mere 3%.
Evidence from research done on open-field emissions of both conifer and deciduous forests [34,35,36], and on un-detached leaves or branchlets [30,37], supports these estimates, indicating that, apart from isoprene, the most common and dominant forest VOCs in terms of percentage of the total emissions are monoterpenes such as α-pinene, d-limonene, and β-pinene, followed by sabinene, myrcene, and camphene. Monoterpenes’ global annual emissions amount to 330–480 million tons, with an average atmospheric concentration above forests from some pmol/mol to several nmol/mol. Other quantitatively important forests VOCs such as GLVs and acetaldehyde are emitted in quantities of around 260 million tons and show atmospheric concentrations of around 1–3 nmol/mol [11,16].
From a quantitative point of view, studies which aimed to determine the atmospheric concentration of some categories of BVOCs under forest canopy reported heterogeneous findings, probably due to a significant influence of the abovementioned factors on the forest air composition: approximately, on the basis of available evidence, the total amount of terpenes can vary from a minimum of 3.5 to a maximum of 56.0 μg/m3, isoprene can range from 0.1 to 28.0 μg/m3, and monoterpenes may have an average concentration of 4.5 μg/m3, whereas the amount of LOX pathway products is usually below the upper threshold of 3.4 μg/m3 [16,31,32,34,38,39,40,41,42,43,44] (Figure 3). In particular, if we only consider monoterpenes, the concentration range of seven of them under the canopy of a Scots pine forest was reported in a study [44] (Figure 4). For example, limonene concentration can vary from 0.81 to 8.10 μg/m3, while α-pinene can reach higher concentrations in pine forest air, up to 13.05 μg/m3 [44]. However, as reported in a review endorsed by the World Health Organization (WHO), measured concentrations of limonene in the air of different forest areas across the world (Europe, North America, and Asia) can largely vary and even reach values of 12.2 g/m3 [45]. Therefore, it is important to consider this high degree of variability when studying health and environmental effects of forest VOCs.
Studies on emissions from plants after artificial lesions [46,47], or experiments performed using techniques such as branch enclosures, which are likely to produce stress in the plant [48], show the presence of GLVs like cis-3-hexen-1-ol, cis-3-hexenyl acetate, cis-3-hexenyl butyrate, and cis-3-hexenyl valerate, although there are contradictory examples [49]. This fits well with the fact that C6 volatiles (aldehydes, alcohols, and their derivatives) such as GLVs are derived from cell membrane denaturation [12]. Not only direct physical stress but also temperature variations can affect the release of BVOCs. GLVs, as well as C1 and C2 oxygenated compound emissions, can increase during heat stress and might continue even for a long time after the temperature has returned to normal levels. Trees with specific storage structures (such as resin ducts in most of the conifers) are less affected by changes in temperature and are moderate isoprenoid emitters, unless these structures are damaged by insects, fires, strong wind, or high humidity. Most deciduous species do not have such structures and release BVOCs directly from mesophyll cells in a more light- and temperature-dependent manner (in particular, Fagaceae and Salicaceae) [12]. However, the most typical compounds emitted after biotic stress are the homoterpenes TMTT and DMNT [17].
Overall, the diversity of BVOCs is certainly much higher, and this is represented in Table 2, in descending order of quantity of emissions [4,16,30,34,35,37,42,46,47,48,49,50,51].

3.3. BVOCs and Plant-Derived Essential Oils

Apart from being plant-emitted components of the atmosphere, forest VOCs can be found in plant-derived products and, in particular, in distilled essential oils, the lipophilic result of steam distillation of aromatic plants [52]. Although essential oils do contain BVOCs, not all BVOCs are present in essential oils, and some molecules included in essential oils are not part of the BVOC molecular suite but are rather artefacts of distillation. Essential oils are dominated by volatile terpenes and terpenoids (C10 and C15) and, in specific cases, they contain phenylpropanoids, oxylipins, and, more rarely, N- and S-containing molecules. They do not contain appreciable quantities of isoprene, highly reactive chemical species such as GLVs (although occasionally found in essential oils from Camellia sinensis, Viola odorata, Carpinus betulus, Fragaria vesca, Rubus idaeus), or any of the volatile but hydrophilic compounds found in hydrosols [53]. Essential oils have a higher percentage of heavier compounds (sesquiterpenes and sesquiterpenoids) compared to atmospheric BVOCs, due to higher temperatures involved in distillation that allow vaporization of the less volatile compounds (the high-boiling ones), and are mainly obtained from plant species with storage structures (glandular trichomes, resiniferous ducts, etc.). Many essential oils seem capable of exerting interesting pharmacological activities, even if not all essential oils are active and no single essential oil exerts all these activities. Moreover, given that essential oils are inherently perceptively salient, they can have mood-altering effects, which are mediated not by classical pharmacological mechanisms but by olfactory images.
A simplified list of the most common and noteworthy activities of essential oils upon inhalation includes general antimicrobial ones and specific effects [54,55,56,57]:
  • antimicrobial effects on antibiotic-resistant bacterial strains;
  • antitussive, mucoactive, bronchodilation, and antispasmodic activities on the respiratory system;
  • non-olfactory-mediated psychopharmacological effects on arousal, activation, memory loss, dementia, cognitive performance, anxiety, quality of life, quality of sleep;
  • antioxidant effect;
  • antinociceptive, anti-inflammatory, and cytotoxic activity;
  • anti-nausea and spasmolytic effects on the intestine.

3.4. Evidence about the General Effects of Forest VOCs on Health

From a pharmacokinetic point of view, when visiting a forest, monoterpene VOCs such as limonene and pinene are mainly absorbed through inhalation, their blood levels rapidly rise after exposure, and they are mostly eliminated unchanged both in exhaled air and in the urine [58]. Accumulation in the adipose tissue has been demonstrated for some BVOCs like limonene [59], as well as some degree of metabolization for terpenes like pinene [60]. Their general activity on inflammation, stress, sleep, and psychological behavior is described below. In particular, among monoterpenes, limonene and pinene are important for their biological activities even on human subjects [36,58] and, therefore, their health properties have been described in dedicated paragraphs.

3.4.1. Immune System and Inflammation

A recent review synthesized available evidence on the immune function enhancing effects of essential oils and, in particular, results of studies about forest bathing were discussed, thus suggesting that even 2-h-long forest walks can be associated with a significant increase in Natural Killer (NK) cell count and activity and can augment the number of cytolytic molecules expressing cells, with such variations lasting for many days after the experience, both in healthy subjects and in individuals with cancer-related pain [61]. Some studies included in this review also showed an effect of forest bathing on serum levels of IL-8, IL-6, and TNFα [61]. These findings confirmed the main results of preclinical laboratory experiments [62]. As with all studies about forest bathing, the suggestion that the mechanism for the observed health benefits is due to the inhalation of forest BVOCs must be balanced by the possible role of mediators like other sensory or non-sensory stimulators [63].
A review of preclinical studies analyzed available evidence regarding the effects of the most common volatile terpenoids found among BVOCs on inflammation and concluded that these compounds might act as inhibitors of pro-inflammatory mediators such as NO, TNF-α, and PGE2, thus modulating signal transduction pathways involving transcription factors like NF-κB and the mitogen-activated protein kinase (MAPK) [4]. Moreover, some terpenoids can directly interact with TRP receptors (implicated in nociception and inflammatory responses), reduce oxidative stress, and stimulate cell autophagy. The authors reported that, although these activities are not limited to forest bathing and can be exploited through skin applications or oral intake of essential oils, the contact during forest bathing may be safer, although less beneficial [4]. In any case, only a small number of preclinical studies have investigated the role of total extracts of forest BVOCs: volatile compounds extracted from hinoki wood essential oil have been reported to increase in vitro the natural killer (NK) cell activity by enhancing the expression of cytolytic enzymes [64]; also, an extract obtained from pinecones has been found to protect bovine mammary epithelial cells, stimulated with lipopolysaccharide (LPS), from oxidative stress after downregulating cyclooxygenase-2 (COX-2) expression [65]. On the other hand, single terpenes (α-phellandrene, 1,8 cineole, linalool, and β-caryophyllene) have been successfully used as pre-treatment to reduce the levels of interleukins, TNF-α, and iNOS in cellular and animal inflammatory-induced models [66,67,68,69]. Furthermore, mitogen-activated protein kinase (MAPK) signal transduction pathways (e.g., JNK and p38) can be inhibited by myrcene, limonene, and borneol [70,71]. Interestingly, β-caryophyllene has been shown to bind the peripheral receptor for cannabinoids (CB2R) and modulate the immune function [72,73]. Additionally, terpene treatment is able to reduce nucleus translocation, phosphorylation, and expression levels of NF-kB [4] and Nrf2 [74,75,76], the main transcriptional factors regulating levels of pro-inflammatory mediators. Moreover, many studies have demonstrated some BVOCs’ (i.e., myrcene, linalool, 1,8 cineole, α- and γ-terpinene) antioxidant properties such as the decrease of Reactive Oxygen Species (ROS), Matrix Metallo-Proteinase (MMP), and Nitric Oxide (NO) production [77,78,79,80]. Finally, inhibition of neuro-inflammation has been shown in vitro for 1,8-cineole [81] and β-caryophyllene [82,83] and in in vivo animal models for d-limonene [84] and linalool [85].

3.4.2. Nervous System and Psychological Behavior

A systematic review on the general effects of forest bathing found that this meditative practice can be associated with positive health benefits, like a reduction in heart rate and blood pressure, increased relaxation, ameliorated general wellbeing, and improvement of depression scores [86], although available evidence is still limited and observed effects are likely to be multifactorial, thus probably depending not only on the presence of BVOCs in the atmosphere but also on the whole natural, green context [87]. These results are in line with preclinical studies on animal behavioral models investigating BVOCs’ neurological role in central nervous system (CNS) depression. In fact, BVOCs have been able to reduce locomotor activity and increase muscle relaxation, with consequent improvement of sleep, pain, and anxiety in mice [88,89,90]. Recently, it has been reported that substances derived from pine trees, such as α-pinene and 3-carene, can enhance sleep by acting as a positive modulator for GABA-A-BZD receptors [91]. Similar effects are reported for borneol, verbenol, isopulegol, and pinocarveol [92,93]. Moreover, inhaled linalool and β-pinene have shown anxiolytic [94,95] and antidepressive [96,97] properties in different animal models.

3.4.3. Endocrine System and Stress

A systematic review concluded that forest bathing can induce a state of physiological relaxation, with a concomitant reduction in stress hormone levels, and it can also exert beneficial effects on general stress-related health outcomes, such as short-term improvements of cardiovascular and metabolic parameters, including reduced glucose levels [98]. A very recent systematic review of 22 clinical studies analyzing the effects of forest bathing on stress discovered that salivary cortisol levels were significantly lower in forest groups compared to control groups both before and after the intervention and pointed out that, although further research is needed, forest bathing can significantly influence cortisol levels in the short term in such a way as to reduce stress, with anticipated placebo effects playing an important role in this [99]. In animal models, forest VOCs have shown a positive effect on stress response [100,101], whilst levels of stress hormones (i.e., plasma corticosterone and adrenaline) have not been influenced by limonene and α-pinene [102,103]. On the other hand, limonene may protect PC12 cells against corticosterone-induced neurotoxicity via the modulation of AMP-activated protein kinase cascade [104].

3.5. Limonene

Limonene (1-methyl-4-prop-1-en-2-ylcyclohexene) is a monocyclic monoterpene hydrocarbon, a cyclohex-1-ene substituted by a methyl group at position 1 and a prop-1-en-2-yl group at position 4, respectively. Given that limonene has a carbon chiral center at position 4, it occurs as two optical isomers: D-limonene or [R-(+)-isomer] and L-limonene or [S-(+)-isomer]. Of the two, D-limonene (also called “citrene” or “carvene”) is the most abundant in plants and it is typical of Rutaceae such as orange, lemon, mandarin, etc. On the other hand, L-limonene is far less common and it can be found in essential oils obtained from Pinus spp., Illicium verum, and Mentha spp., while the D-L mixture (dipentene) is even less common [53,105]. Table 3 reports average contents in D-limonene of some essential oils derived from trees [106].

3.5.1. Anti-Inflammatory Activity

In a systematic review investigating the anti-inflammatory potential of forest BVOCs, D-limonene was shown to be able to modulate many pro-inflammatory mediators such as TNF-α, NO, IL-1β, IL-6, enzymes involved in the inflammatory response (5-LOX, COX-2, iNOS), transcription factors (NF-κB), and signal transduction kinases (p38, JNK, ERK) [4]. In particular, the modulation of inflammatory substances and transcription factors was demonstrated in various animal models (Wistar rats, BALB/c mice, fruit flies) and in some cell cultures (raw 264.7 cell lines, human chondrocytes) [4]. These activities seemed responsible both for preventative action and for a direct effect on inflammation [24]. It was also noted that the (l)-(+)-limonene enantiomer was approximately three-fold less active than the (S)-(–)-limonene one in 5-lipoxygenase inhibitory activity [107].
On the specific role of limonene in respiratory tract inflammation, the review by Santana and colleagues concluded that the monoterpene has effective anti-inflammatory activity in both preventing and controlling respiratory system injuries [108], and this points towards a possible role for limonene in the management of allergic airway diseases and asthma [24].
In a clinical trial on “inflamm-ageing” and oxidative stress in healthy elderly subjects, the intake of 10 mg/kg of limonene as a food supplement in the context of a specific diet resulted in a decrease in fibrinogen, glucose, and insulin levels and in an amelioration of the HOMA-IR index, a homeostatic model assessment used to quantify insulin resistance and beta-cell function [109].

3.5.2. Antioxidant Activity

In a review conducted by Vieira and colleagues, limonene was described as an antioxidant compound with free radical scavenging properties, capable of reducing oxidative stress in diabetic rats and of attenuating oxidative stress not only in murine lymphocytes but also in human epithelial cells and fibroblasts, with cell proliferating, anti-apoptotic, and DNA-protecting effects [24]. Specifically, limonene may act by inhibiting caspase-3/caspase-9 activation, p38 MAPK phosphorylation, and LOX activity and by increasing the activities of cell antioxidant enzymes and the Bcl-2/Bax ratio [4].

3.5.3. Antiproliferative Activity

D-Limonene has been studied as an antiproliferative agent, and in vitro experiments have suggested a series of possible mechanisms, such as the induction of phase II carcinogen-metabolizing enzymes (a); the reduction of cancer cell proliferation via prenyl-transferase inhibiting activity (possibly diminishing the addition of FPP to the tail of RAS proteins) (b); increased tumor cell apoptosis, autophagy (via the mitochondrial death pathway, the PI3k/Akt pathway, and the caspase-3 and -9 activity), and differentiation (c); growth suppression (through a reduction in cyclin-D1 and an increase in TGF-β signaling leading to a more benign phenotype) (d); decreased tumor-induced immunosuppression, tumor-promoting inflammation, angiogenesis, and metastasis (through a reduction in circulating VEGF and a blockage of the receptor VEGFR1) (e); improved genome stability (increased DNA damage repair and PARP cleavage) (f) [4,17,24,110,111]. In spite of the many mechanistic studies, there is a dearth of preclinical studies with proper animal models and of human clinical trials. The most relevant ones are summarized in Table 4 and mostly involve the oral administration of limonene.

3.5.4. Antinociceptive Activity

Data about the activity of D-limonene on nociception are scant. Available evidence with laboratory animals points towards a bimodal action of this monoterpene, which, when administered orally, appears to inhibit nociception and to prevent artificially-induced hyperalgesia (probably by reducing the synthesis of cytokines), while, when topically applied, it seems capable of eliciting pain, probably after an interaction with TRPA1 ion channels [24,116].

3.5.5. Other Pharmacological Activities

According to a review by Vieira and colleagues analyzing data from experiments with animal models, limonene may be a potential metabolic agent, capable of improving hyperglycemia, pancreatic β-cell mass, lipid blood levels, and liver fat accumulation [24]. Limonene could also be a solvent for cholesterol, and it has been used to dissolve gallstones after direct infusion into the gallbladder [117]. Limonene may even exert protective and healing effects in experimental models of gastric ulcer and colitis, possibly by promoting mucus secretion and PGE2 production [24]. In in vitro settings with Rolf B1.T cells, limonene can decrease levels of neuropeptide Y, the most important appetite regulator [118]. It finally shows moderate antifungal and antibacterial activities, more pronounced in Gram-positive strains [116]. A summary of preclinical evidence of limonene biological activity has been reported in Table 5.

3.6. Pinenes

Pinene is a bicyclic monoterpene hydrocarbon with two structural isoforms, α- and β-pinenes. Both L- and/or D-forms, as well as racemic forms, may occur. These two isoforms are important constituents of conifer oleoresins and are widely distributed in plants, although α-pinene is generally more abundant.
α-pinene (2,6,6-trimethylbicyclo[3.1.1]hept-2-ene) is a bicyclo[3.1.1]hept-2-ene substituted by methyl groups at positions 2, 6, and 6, respectively. Both optical forms are present in conifer essential oils [138]. Table 6 reports the most common tree species whose total essential oil contains a quantity of α-pinene at a percentage of more than 20% [70].
β-pinene (6,6-dimethyl-2-methylidenebicyclo[3.1.1]heptane) is a bicyclo[3.1.1]heptane substituted by methyl groups at positions 6 and 6, respectively, and an exocyclic double bond [139]. It usually accompanies α-pinene in turpentine and other essential oils and is present with both optical forms. It is the main compound of essential oil derived from Piper capense [53]. Table 7 reports the most common tree species whose total essential oil contains a quantity of β-pinene at a percentage of more than 20% [70].

Pharmacological and Clinical Activities of Pinenes

In a recent review of the scientific literature, it has been reported that, although many in vitro studies exist about the pharmacological activities of pinene and pinene-containing essential oils, few in vivo studies and even less clinical trials on the topic have been conducted so far [23]. In fact, none of the reviewed clinical studies tested isolated pinene, but mixtures of different molecules or whole essential oils have been often tested.
A clinical trial involving patients with bronchitis tested a product containing α-pinene, D-limonene, and 1,8-cineole, and, although study results were positive [140], if we consider the clinically recognized effects of 1,8-cineole on bronchitis, it is impossible to discern whether α-pinene was important for the results.
A single-blind clinical trial on pediculosis tested two essential oils, one of Eucalyptus globulus and one of Cinnamomum zeylanicum, both containing α-pinene, and found eucalyptus oil to be superior to permethrin [141], while a second study evaluated the effects of essential oil obtained from the leaves of Lippia multiflora and found it to be superior to benzyl benzoate [142]. In both cases, however, given the recognized significant activity of 1,8-cineole and terpineol, respectively, on pediculosis, it is impossible to declare pinene as an active and useful compound per se.
The reviewed trials on memory and Alzheimer’s disease tested only whole essential oils belonging to the genus Salvia (Salvia officinalis and Salvia lavandulaefolia) for their inhibitory activity on cholinesterases [143,144,145]. Again, as previously noted, these essential oils contain, apart from pinene, several compounds, and, in fact, 1,8-cineole is likely to be the most active molecule, although a synergistic interaction with other monoterpenes has been suggested.
Amongst pharmacological effects of pinenes demonstrated on the basis of preclinical evidence, authors of the previously mentioned review specifically analyzed those relevant to cancer growth, allergies, inflammation, infections, and anxiety [23]. Data about cancer cells suggest that α-pinene might have effects on NF-κB and IκBα, and, therefore, on inflammation, mitochondrial functions, ROS production, caspase-3 activity, and DNA fragmentation [146], but also on efflux pumps responsible for multidrug-resistant tumors [147], as well as on cell cycle arrest via the cyclin-B protein [148]. A pre-treatment of allergic rhinitis models with α-pinene resulted in a significant reduction in clinical symptoms and inflammatory cytokine levels [149]. α-pinene also appears to exert moderate antimicrobial [150,151] and immune-modulating effects [61,62]. Finally, it has been shown that α-pinene binds to the GABA-A receptor, at the benzodiazepine binding site, thus prolonging GABAergic inhibitory signaling and suggesting an anxiolytic and sleep-enhancing effect [91]. A summary of preclinical evidence of α-pinene biological activity has been reported in Table 8.

4. Discussion

4.1. Implications for Individual Health

Inhaling forest VOCs can result in useful antioxidant and anti-inflammatory effects on the airways, and the pharmacological activity of some terpenes absorbed through inhalation may be also beneficial to promote brain function by decreasing mental fatigue, inducing relaxation, and improving cognitive performance and mood [164,165]. The effect of these volatile compounds found in forest air is not limited to an action on the respiratory system, but, after absorption through inhalation, they are supposed to exert systemic effects, such as those on the nervous system (relaxing, anxiolytic, and antidepressant action). Evidence from laboratory studies seems to confirm these effects, thus underscoring that, in particular, limonene and pinene can modulate the release of various cytokines (for example, TNF-α, IL-1, IL-6), inflammatory mediators (for example, NF-κB signal transduction pathway, MAPK, COX-2 activity), and neurotransmitters (for example, dopamine levels, action on GABA receptors) in such a way as to reduce inflammation and pain and to improve anxiety, mood, and sleep quality (Table 5, Table 8, and Table 9).
Although the magnitude of some of these pharmacological effects may be limited when inhaling such compounds during a short forest trip due to their relative atmospheric concentration (generally lower than that one prepared in experimental settings) and pharmacokinetics, even a mild action associated with psychophysical relaxation may be already beneficial for individual wellbeing. In fact, large studies aimed at thoroughly assessing pharmacokinetic and pharmacodynamic characteristics of forest VOCs in real-life situations are, to date, actually lacking, and it is only possible to formulate some plausible hypotheses. For example, with regard to limonene, human studies discussed in this review mostly investigated pharmacological effects of its oral intake when taken in relatively high quantities, up to several grams a day (Table 4). In a pharmacokinetic study coupled with a phase-I trial, the administration of limonene to cancer patients was observed to be followed within a few hours by a peak in limonene plasma concentrations of 10.8 μM (mean Cmax) when the oral daily dose amounted to 8 g/m2 and of 20.5 μM (mean Cmax) when the dose was 12 g/m2 [114]. However, inhaling limonene in a forest setting can be quite different, since many environmental and individual characteristics along with inhalation-related pharmacokinetic mechanisms can introduce a high degree of variability, which makes it hard (to date) to fully predict the exact short- and long-term health effects of a natural and real-life exposure to specific BVOCs. In fact, as reported in a review, measured concentrations of limonene in the air of different forest areas can largely vary, ranging from as little as 0.9 ng/m3 to as much as 12.2 g/m3 [45]. In a toxicokinetic study with healthy volunteers, participants were exposed for 2 h in a chamber with different concentrations of limonene (10, 225, and 450 mg/m3) and the pulmonary uptake was estimated to be up to 70% of the amount supplied, with plasma concentrations of this substance rapidly rising within 2 h after exposure and peaking at different levels (ranging, on average, from less than 2 to almost 25 μM), depending on the quantity of limonene diffused in the air [166]. No major short-term clinically symptomatic or toxic/irritative effects were observed, and only minor changes in respiratory function were reported when limonene concentration in the chamber was set at the highest experimental level (450 mg/m3) [166]. However, the pulmonary absorption of terpenes may be even less efficient, since more recent studies state that it could be 54–76% of the dose supplied with inhaled air [167]. In an interesting study involving a small cohort of healthy subjects, blood concentrations of some conifer-derived monoterpenes were measured before and after a 60-min trip in a Japanese forest [58]. The authors noticed that blood concentrations of some BVOCs exhibited a marked increase after visiting the forest, and, in particular, average plasma levels of α-pinene changed from 2.6 (baseline) to 19.4 nM (post-intervention) [58]. A pharmacokinetic study about metabolites of α-pinene showed that their blood levels tend to markedly and rapidly peak after the administration of a single oral dose of 10 mg to four volunteers, with trans-verbenol (one of the most relevant pinene metabolites) Cmax quickly reaching a mean value of 26 nM and α-pinene being already undetectable 1 h after the exposure [60]. In brief, blood concentrations of BVOCs after indoor and, even more, outdoor inhalation show a higher degree of variability than after their oral intake in more controlled experimental settings. With available data, we can only affirm that plasma concentrations of BVOCs tend to rise whenever a subject is exposed to a forest, but, to date, due to limitations of existing studies, it is not possible to exactly quantify the actual magnitude of their specific beneficial effects for human health, especially with regard to their action at a systemic level.
Taking into account all these findings, potential moderators of the effect and confounding factors to be considered by researchers when planning future studies can include the following:
  • the exact concentration of any BVOC in the forest air when exposure occurs;
  • the duration of exposure;
  • the intensity of physical activities performed in the green environment;
  • specific modalities of biological sample collection, transport, and analysis;
  • individual health-related characteristics, which can influence the uptake, metabolism, accumulation, and excretion of inhaled BVOCs;
  • lifestyle habits, which can determine baseline blood levels of compounds like limonene and pinene absorbed from food, medicines, and perfumes [58].
Further rigorous pharmacokinetic studies ought to be performed to thoroughly estimate the actual absorbed quantity of BVOCs during a forest trip. Then, it would be possible to precisely assess short- and long-term effects on health of the inhalation of specific BVOCs and to correlate such effects with specific thresholds of BVOCs concentrations in the forest air. It is known that forest VOCs like limonene and, even more, pinene, are mostly released by conifers (pines, fir trees, cypresses) (Table 3, Table 6, and Table 7), and, therefore, the exposure to these volatile substances (with more documented effects on health) can be maximized by visiting a forest whose composition is rich in these trees. In general, beyond differences in tree composition, levels of BVOCs in the forest air exhibit diurnal quali-quantitative variations, with two daily peaks (early in the morning and in the afternoon) [31,32], and these cyclic changes may influence the magnitude and type of potential beneficial effects of their inhalation depending on the time of the day when the exposure occurs. For this reason, it would be useful to carry out additional environmental studies aimed at quantifying all concentration changes in forest VOCs in order to maximize the visitors’ exposure and, therefore, the impact of such compounds on health after inhalation.
Moreover, beneficial psychological and physiological effects of visiting a forest (such as in the meditative practice called “forest bathing”) cannot be solely attributed to VOC inhalation but are due to a global and integrated stimulation of the five senses [63], induced by all specific characteristics of the natural environment, with the visual component probably playing a fundamental role in the overall effect [99]. Therefore, even if inhaling BVOCs can exert useful effects on health while visiting a forest, absorbing these volatile compounds does not cover all benefits of forest bathing, which, to date, cannot be artificially replaced with a virtual experience and actually requires a full real-life interaction with the natural environment. However, further investigating all medicinal properties of specific forest VOCs with properly designed clinical studies can be useful to discover their potential uses in medical practice as drugs for some health conditions and to possibly discover new therapeutic agents derived from them.
Table 9. Summary of preclinical evidence relative to biological activities of the five most common forest VOCs [3,30,34,35,36,37].
Table 9. Summary of preclinical evidence relative to biological activities of the five most common forest VOCs [3,30,34,35,36,37].
MoleculeEffectsMechanismsReferences
D-LimoneneAnti-inflammatoryIt inhibits the synthesis or release of pro-inflammatory mediators (TNF-α, NO, IL-1β, IL-6, IL-5, IL-13, TGF-β), enzymes (5-LOX, COX-2, iNOS), transcription factors (NF-κB), and mitogen-activated protein (MAP) kinase family members (p38, JNK, ERK).[4,70,71,124]
AntioxidantIt inhibits caspase-3/caspase-9 activation, increases the activities of cell antioxidant enzymes (catalase, peroxidase) and the Bcl-2/Bax ratio.[4,70,123]
AntiproliferativeIt induces phase II carcinogen-metabolizing enzymes, inhibits prenyl-transferase activity, increases cell autophagy (via MAP1LC3B, mitochondrial death pathway, the PI3k/Akt pathway, and caspase-3 and -9 activity) and differentiation, reduces cyclin-D1 and increases TGF-β signaling, decreases tumor-induced immunosuppression, reduces circulating Vascular Endothelial Growth Factor (VEGF) and blocks the receptor VEGF-R1, increases DNA damage repair and PARP cleavage. It modulates the expression of the chemotactic protein MCP-1 and of the proteolytic enzymes MMP-2, MMP-9.[4,17,24,110,111,121,124,128,129]
AntinociceptiveBimodal activity: topically applied, it seems capable of eliciting pain, via interaction with TRPA1 ion channels, while in other modes of administration, it has shown antinociceptive effects in different experimental models.[24,90,116,127,133,136,137]
AnxiolyticIt shows some degree of efficacy in mice and rat models.[90,127,135]
AntidepressantIt shows some degree of efficacy in mice and rat models.[132,133,134]
α-PineneAnxiolytic, sedativeIn several animal models, it enhances sleep by acting as a positive modulator for GABA-A-BZD receptors, prolonging GABAergic inhibitory signaling.[91,155,157,158,159,160,161]
Anti-inflammatoryIt modulates NF-κB and IκBα, ERK, JNK, IL-1β, IL-6, iNOS (and NO secretions), MMP-1, MMP-13, COX-2.[146,152,153,155,156]
AntioxidantIt reduces ROS production, caspase-3 activity, modulates superoxide dismutase, catalase, peroxidase activity, NO and IL-6 secretions.[146,155,168]
AntiproliferativeIt acts on efflux pumps responsible for multidrug-resistant tumors and on cell cycle arrest via the cyclin-B protein.[147,148]
AnalgesicIt shows some degree of efficacy in animal models.[156,162,163]
β-PineneAnxiolytic,
antidepressant
It binds to the GABA-A receptor, prolonging GABAergic inhibitory signaling. It showed efficacy in animal models.[91,94,95,96,97]
Anti-inflammatoryIt modulates NF-κB and IκBα.[146]
AntioxidantIt reduces ROS production, caspase-3 activity, MMP and NO activities.[77,78,79,80,146]
AntiproliferativeIt acts on efflux pumps responsible for multidrug-resistant tumors and on cell cycle arrest via the cyclin-B protein.[147,148]
β-MyrceneAnti-inflammatoryIt modulates MAP kinases such as JNK, p38, and it inhibits the synthesis and release of PGE-2.[70,71,169]
AntiproliferativeIt blocks hepatic carcinogenesis caused by aflatoxin.[170]
AnalgesicIt is analgesic in mice, and its action is blocked by naloxone, perhaps via the α-2 adrenoreceptor.[171]
Sedative, myorelaxantIt is a muscle relaxant in mice, and it potentiates barbiturate sleep time at high doses.[88]
GastroprotectiveIt contributes to the integrity of the gastric mucosa, decreasing ulcerative lesions, attenuating lipid peroxidative damage, and preventing depletion of GSH, GR, and GPx.[172]
CampheneMetabolismAs a food supplement, it reduces animal models’ body weight and increases adiponectin levels and receptor mRNA expression in the liver.[173]
AntiproliferativeIt induces apoptosis in cancer cell lines (B16F10-Nex2 melanoma), chromatin condensation, cell shrinkage, apoptotic body formation, fragmentation of nucleus, and caspase-3 activation.[174]
AntioxidantIt prevents AAPH-induced lipoperoxidation and inhibits the superoxide radical.[175]
AntinociceptiveWeak effects on acetic acid-induced writhing in mice models.[175]
AntihyperlipidemicIt reduces total and LDL-cholesterol and triglycerides in hyperlipidemic rats and in HepG2 cells, not by inhibiting HMG-CoA reductase but by increasing apolipoprotein AI expression, possibly via SREBP-1 upregulation and MTP inhibition.[176]

4.2. Implications for Preventive Medicine and Public Health

Useful effects of forests and green areas on psychophysical wellbeing have been underscored by several studies, with all sensory components, ranging from visual to olfactory stimulations, playing an integrated role in the overall beneficial action on individual health [63,86,99,177]. This may also explain the tight link between shamanism and wild environments, and why folkloristic and ritual practices have spread worldwide from the Amazon to the Siberian forest [178,179]. Evidence from research works analyzed in this review suggests that forest bathing can be beneficial for general health, thus exerting a general anti-stress and immune boosting activity. At a public health level, epidemiological studies have highlighted that there is evidence for a significant association between the extension of green areas within a given community and specific health outcomes like perceived mental and general health, as well as all-cause mortality [177]. Similar research works have confirmed a positive association between living closer to green areas and improvements in physician-assessed morbidity determined by various cardiovascular, muskuloskeletal, respiratory, mental, neurological, digestive, and other health conditions [180]. Indeed, the type of activities performed outside, possibly ranging from static relaxation to intense physical exercises, along with the subjective perception of safety and connectedness to nature while visiting the natural environment, can influence the magnitude of beneficial effects exerted by the exposure to green areas [181]. In particular, forests can be perceived as dangerous for several reasons (wild and unknown environment, potentially aggressive fauna, parasites, poisonous plants, limited supporting facilities, human activities like hunting, cutting trees, practicing extreme sports, etc.), and all necessary precautions have to be taken to prevent any risk of being harmed in order to fully and safely benefit from forest exposure. Additionally, when subjects suffer from specific diseases, a medical consultation is advised before planning to visit a forest.
Recently, research efforts of many expert scientists from different countries all around the world have converged into the definition of new concepts like “forest therapy” and “forest medicine”, which both refer to the scientific study and evidence-based applications of visiting a forest for therapeutic or preventive purposes [182]. In particular, from the foundation of the International Society of Nature and Forest Medicine (INFOM), researchers of this field have advocated for studying the topic in more depth, as well as to disseminate relevant findings worldwide in order to “establish Forest Therapy as an effective, evidence-based and low-cost public health treatment for selected lifestyle-related diseases” [182,183]. Beneficial effects of this practice are (although not exclusively) determined by the inhalation of forest VOCs, which are responsible for the olfactory perception of pleasing fragrances from trees and flowers [182]. Beyond these chemical factors, useful health effects of visiting a forest can be also attributed to physical (such as environmental characteristics, climate conditions, visual impressions, sounds) and psychological factors (to what extent the environment is perceived as pleasant, relaxing, and safe by an individual, i.e., placebo effects) [182]. Visiting forests on a relatively regular basis can be a good health-promoting practice, since, by reducing stress levels and boosting immune function, it seems capable of diminishing the incidence of stress-related and lifestyle-induced illnesses, varying from cardiovascular, respiratory, or metabolic diseases to neuropsychiatric conditions and, possibly, even cancer [86]. Some practical recommendations to optimally benefit from forest bathing for preventive purposes include performing only light physical activity in the natural environment (like walking at a regular pace without getting tired), stay well hydrated, avoiding the use of technological devices for recreation (thus preferring meditative contemplation as a relaxing practice during the forest visit and a bath in hot spring waters after the trip), spending in the forest 2 to 4 h and walking 2.5 to 5 km in a day along clean paths with some stopovers [182].
In general, on the basis of retrieved evidence and experts’ proposals, visiting a forest can be a useful practice, with the olfactory component playing a role in it, and it can be a sustainable public health practice to promote both individual wellbeing and community health, thus contributing to the prevention of many stress- and lifestyle-related diseases.

4.3. Forest VOCs and Natural Landscape Design

Today, around half of the world’s population lives in metropolitan areas and, over the years, technology, commuting, working, and some leisure activities have made us more and more inhabitants of an “indoor society”, in which natural light has been almost completely supplanted by artificial light, and the sounds and smells of nature are often a distant memory. Even if we do not notice it immediately, this can keep us under constant stress, not only because of the constant exposure to urban environments and their stressful stimuli but also because of the lack of contact with natural environments, which can promote health and wellbeing. Another equally important aspect is that green areas, even within an urban space, offer the possibility to organize open-air recreational activities, thus increasing socialization and residents’ quality of life [184,185,186].
In the last few decades, the concept of “biophilic design” has been introduced to indicate any sustainable design aimed at better connecting people with the natural world and with vital processes [187]. In particular, the attention of some outdoor designers has been focused on “healing gardens”, namely green areas purportedly planned to induce a psycho-physical state of wellbeing in their visitors/users [188]. Elements involved in the design of a healing garden are closely related to the therapy of the senses: perfumes (aromatherapy), colors (chromotherapy), sounds (music therapy), sometimes tactile or gustatory experiences, and symbols. Provided that, today, the relationship between man and architecture is mainly governed by sight, but, as underscored in this review, some natural volatile compounds can influence health, designing outdoor environments that have an impact on both sight and smell should be the next step. In particular, it is deemed necessary to create green areas and paths surrounded by fragrant shrubs, where it is possible to talk or relax, while walking immersed in nature with positive olfactory stimulations to guarantee an inclusive experience for all, both sighted and visually impaired or blind people. From this point of view, a precaution to take in order not to create confusion in passers-by is to group aromatic plants by species and arrange them at a certain distance, thus avoiding creating chaotic green spots. In fact, the relationship between space and smell is complex and depends on many factors, including the type of building materials, specific activities that take place in a given environment, the time of the day, air humidity, but also plants, animals, and people who pass through it. All these factors should be taken into account by landscape and outdoor designers, who must be encouraged to cooperate with a multidisciplinary team in order to achieve the best results.
A virtuous example of how to harness beneficial properties of forests in order to promote health is the landscape project involving the natural park of the Oasi Zegna, in Alta Val Sessera (Piedmont region, Italy). A study carried out in 2012 has scientifically shown that the beech wood of the Oasi Zegna can release volatile substances from the foliage, like monoterpenes, capable of bringing benefits to the organism and of stimulating the immune system in a positive way [189]. For this reason, three freely accessible paths were created from June to September, which corresponds to the period of maximum foliation of beech trees.
Another project designed by the CERFIT team (Research and Innovation Center in Phytotherapy and Integrated Medicine, Careggi University Hospital) is the so called “Giardino della Salute”, a healing garden which will be located in an outdoor green area (a pine forest) within the Careggi Hospital district in Florence (Italy) [190]. The ultimate aim of the project is to revitalize and redevelop a public park, as well as to improve the wellbeing and quality of life of all its users, with keen attention to healthcare personnel, hospital patients, and their relatives or visitors. In order to offer a personalized experience which is expected to meet individual needs, some specific footpaths will be laid: the “nutrition” path, where there will be plants used for food; the path of “the five senses”, characterized by an immersive sensory experience with various impressions; the “mind” path, for relaxing or stimulating the nervous system; the path of “natural hazards”, in which poisonous plants will be described for educational purposes; a path for generic users, another one dedicated to patients, and the last one to students. Specific characteristics of plants and flowers displayed along each path, including their appearance, colors, and phytochemical substances released into the surrounding environment, have been chosen to be synergically harnessed for beneficial purposes. The project also involves a herbalist and an educator who can guide visitors along their journey across the healing garden and help them to understand the most important uses and properties of medicinal plants. This project is a valuable example of how to make the most of beneficial properties of an ad hoc designed urban green area, taking into consideration various aspects of the effects of plants on human health, including the activity of biogenic VOCs released in the air.

5. Conclusions

Inhaling forest VOCs like limonene and pinene can result in useful antioxidant and anti-inflammatory effects on the airways, and the pharmacological activity of some terpenes absorbed through inhalation may be also beneficial to promote brain function by decreasing mental fatigue, inducing relaxation, and improving cognitive performance and mood. The tree composition can markedly influence the concentration of specific VOCs in the forest air. Moreover, beneficial psychological and physiological effects of visiting a forest (such as in the meditative practice called “forest bathing”) cannot be solely attributed to VOC inhalation but are due to a global and integrated stimulation of the five senses, induced by all specific characteristics of the natural environment, with the visual component probably playing a fundamental role in the overall effect. Globally, these findings can have useful implications for individual wellbeing, public health, and landscape design. Further clinical and environmental studies are advised, since the majority of the existing evidence is derived from laboratory findings.

Author Contributions

Conceptualization, M.A. and D.D.; Methodology, M.A., D.D., G.B., M.V., V.M., and F.F.; Validation, M.A., D.D., G.B., M.V., V.M., and F.F.; Investigation, M.A., D.D., G.B., M.V., V.M., and F.F.; Resources, M.A., D.D., G.B., M.V., V.M., and F.F.; Data Curation, M.A., D.D., G.B., M.V., V.M., and F.F.; Writing—Original Draft Preparation, M.A., D.D., G.B., M.V., V.M., and F.F.; Writing—Review and Editing, M.A., D.D., G.B., M.V., V.M., and F.F.; Visualization, M.A., D.D., G.B., M.V., V.M., and F.F.; Supervision, M.A., D.D., and F.F.; Project Administration, M.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research work was not funded.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Went, F.W. Blue hazes in the atmosphere. Nature 1960, 187, 641–643. [Google Scholar] [CrossRef]
  2. Rasmussen, R.A.; Went, F.W. Volatile organic material of plant origin in the atmosphere. Proc. Natl. Acad. Sci. USA 1965, 53, 215–220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Guenther, A.B.; Jiang, X.; Heald, C.L.; Sakulyanontvittaya, T.; Duhl, T.; Emmons, L.K.; Wang, X. The Model of Emissions of Gases and Aerosols from Nature version 2.1 (MEGAN2.1): An extended and updated framework for modeling biogenic emissions. Geosci. Model Dev. 2012, 5, 1471. [Google Scholar] [CrossRef] [Green Version]
  4. Kim, T.; Song, B.; Cho, K.S.; Lee, I.-S. Therapeutic Potential of Volatile Terpenes and Terpenoids from Forests for Inflammatory Diseases. Int. J. Mol. Sci. 2020, 21, 2187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Niinemets, Ü.; Monson, R.K. Biology, Controls and Models of Tree Volatile Organic Compound Emissions; Springer: Dordrecht, The Netherlands, 2013; ISBN 9789400766051. [Google Scholar]
  6. Dyakov, Y.T.; Dzhavakhiya, V.G.; Korpela, T. Comprehensive and Molecular Phytopathology; Elsevier: Amsterdam, The Netherlands, 2007; ISBN 9780444521323. [Google Scholar]
  7. Nomura, M. Phytoncide—Its Properties and Applications in Practical Use. In Gas Biology Research in Clinical Practice; Karger Publishers: Basel, Switzerland, 2011; pp. 133–143. [Google Scholar]
  8. Laothawornkitkul, J.; Taylor, J.E.; Paul, N.D.; Hewitt, C.N. Biogenic volatile organic compounds in the Earth system. New Phytol. 2009, 183, 27–51. [Google Scholar] [CrossRef] [PubMed]
  9. War, A.R.; Paulraj, M.G.; Ahmad, T.; Buhroo, A.A.; Hussain, B.; Ignacimuthu, S.; Sharma, H.C. Mechanisms of plant defense against insect herbivores. Plant Signal. Behav. 2012, 7, 1306–1320. [Google Scholar] [CrossRef] [Green Version]
  10. Materić, D.; Bruhn, D.; Turner, C.; Morgan, G.; Mason, N.; Gauci, V. Methods in plant foliar volatile organic compounds research. Appl. Plant Sci. 2015, 3. [Google Scholar] [CrossRef]
  11. Sonwani, S.; Saxena, P.; Kulshrestha, U. Role of Global Warming and Plant Signaling in BVOC Emissions. In Plant Responses to Air Pollution; Kulshrestha, U., Saxena, P., Eds.; Springer: Singapore, 2016; pp. 45–57. ISBN 9789811012013. [Google Scholar]
  12. Loreto, F.; Schnitzler, J.-P. Abiotic stresses and induced BVOCs. Trends Plant Sci. 2010, 15, 154–166. [Google Scholar] [CrossRef]
  13. Dudareva, N.; Pichersky, E.; Gershenzon, J. Biochemistry of plant volatiles. Plant Physiol. 2004, 135, 1893–1902. [Google Scholar] [CrossRef] [Green Version]
  14. Vivaldo, G.; Masi, E.; Taiti, C.; Caldarelli, G.; Mancuso, S. The network of plants volatile organic compounds. Sci. Rep. 2017, 7, 11050. [Google Scholar] [CrossRef] [Green Version]
  15. Memari, H.R.; Pazouki, L.; Niinemets, Ü. The Biochemistry and Molecular Biology of Volatile Messengers in Trees. In Biology, Controls and Models of Tree Volatile Organic Compound Emissions; Niinemets, Ü., Monson, R.K., Eds.; Springer: Dordrecht, The Netherlands, 2013; pp. 47–93. ISBN 9789400766068. [Google Scholar]
  16. Peñuelas, J.; Llusià, J. BVOCs: Plant defense against climate warming? Trends Plant Sci. 2003, 8, 105–109. [Google Scholar] [CrossRef]
  17. Maffei, M.E.; Gertsch, J.; Appendino, G. Plant volatiles: Production, function and pharmacology. Nat. Prod. Rep. 2011, 28, 1359–1380. [Google Scholar] [CrossRef] [PubMed]
  18. Sharifi-Rad, J.; Sureda, A.; Tenore, G.C.; Daglia, M.; Sharifi-Rad, M.; Valussi, M.; Tundis, R.; Sharifi-Rad, M.; Loizzo, M.R.; Ademiluyi, A.O.; et al. Biological Activities of Essential Oils: From Plant Chemoecology to Traditional Healing Systems. Molecules 2017, 22, 70. [Google Scholar] [CrossRef] [PubMed]
  19. Šimpraga, M.; Ghimire, R.P.; Van Der Straeten, D.; Blande, J.D.; Kasurinen, A.; Sorvari, J.; Holopainen, T.; Adriaenssens, S.; Holopainen, J.K.; Kivimäenpää, M. Unravelling the functions of biogenic volatiles in boreal and temperate forest ecosystems. Eur. J. For. Res. 2019, 138, 763–787. [Google Scholar] [CrossRef] [Green Version]
  20. Ditengou, F.A.; Müller, A.; Rosenkranz, M.; Felten, J.; Lasok, H.; van Doorn, M.M.; Legué, V.; Palme, K.; Schnitzler, J.-P.; Polle, A. Volatile signalling by sesquiterpenes from ectomycorrhizal fungi reprogrammes root architecture. Nat. Commun. 2015, 6, 6279. [Google Scholar] [CrossRef]
  21. Bourtsoukidis, E.; Behrendt, T.; Yañez-Serrano, A.M.; Hellén, H.; Diamantopoulos, E.; Catão, E.; Ashworth, K.; Pozzer, A.; Quesada, C.A.; Martins, D.L.; et al. Strong sesquiterpene emissions from Amazonian soils. Nat. Commun. 2018, 9, 2226. [Google Scholar] [CrossRef]
  22. Mofikoya, A.O.; Miura, K.; Ghimire, R.P.; Blande, J.D.; Kivimäenpää, M.; Holopainen, T.; Holopainen, J.K. Understorey Rhododendron tomentosum and Leaf Trichome Density Affect Mountain Birch VOC Emissions in the Subarctic. Sci. Rep. 2018, 8, 13261. [Google Scholar] [CrossRef] [Green Version]
  23. Salehi, B.; Upadhyay, S.; Erdogan Orhan, I.; Kumar Jugran, A.; LD Jayaweera, S.; A Dias, D.; Sharopov, F.; Taheri, Y.; Martins, N.; Baghalpour, N.; et al. Therapeutic Potential of α- and β-Pinene: A Miracle Gift of Nature. Biomolecules 2019, 9, 738. [Google Scholar] [CrossRef] [Green Version]
  24. Vieira, A.J.; Beserra, F.P.; Souza, M.C.; Totti, B.M.; Rozza, A.L. Limonene: Aroma of innovation in health and disease. Chem. Biol. Interact. 2018, 283, 97–106. [Google Scholar] [CrossRef] [Green Version]
  25. Antonelli, M.; Firenzuoli, F.; Salvarani, C.; Gensini, G.F.; Donelli, D. Reading and interpreting reviews for health professionals: A practical review. Intern. Emerg. Med. 2020. [Google Scholar] [CrossRef]
  26. Green, B.N.; Johnson, C.D.; Adams, A. Writing narrative literature reviews for peer-reviewed journals: Secrets of the trade. J. Chiropr. Med. 2006, 5, 101–117. [Google Scholar] [CrossRef] [Green Version]
  27. Ferrari, R. Writing narrative style literature reviews. Med. Writ. 2015, 24, 230–235. [Google Scholar] [CrossRef]
  28. Tholl, D.; Sohrabi, R.; Huh, J.-H.; Lee, S. The biochemistry of homoterpenes—Common constituents of floral and herbivore-induced plant volatile bouquets. Phytochemistry 2011, 72, 1635–1646. [Google Scholar] [CrossRef] [PubMed]
  29. Heil, M. Indirect defence via tritrophic interactions. New Phytol. 2008, 178, 41–61. [Google Scholar] [CrossRef] [PubMed]
  30. Brilli, F.; Ciccioli, P.; Frattoni, M.; Prestininzi, M.; Spanedda, A.F.; Loreto, F. Constitutive and herbivore-induced monoterpenes emitted by Populus × euroamericana leaves are key volatiles that orient Chrysomela populi beetles. Plant Cell Environ. 2009, 32, 542–552. [Google Scholar] [CrossRef] [PubMed]
  31. Meneguzzo, F.; Albanese, L.; Bartolini, G.; Zabini, F. Temporal and Spatial Variability of Volatile Organic Compounds in the Forest Atmosphere. Int. J. Environ. Res. Public Health 2019, 16, 4915. [Google Scholar] [CrossRef] [Green Version]
  32. Bach, A.; Yáñez-Serrano, A.M.; Llusià, J.; Filella, I.; Maneja, R.; Penuelas, J. Human Breathable Air in a Mediterranean Forest: Characterization of Monoterpene Concentrations under the Canopy. Int. J. Environ. Res. Public Health 2020, 17, 4391. [Google Scholar] [CrossRef]
  33. Simon, V. Estimating the emission of volatile organic compounds (VOC) from the French forest ecosystem. Atmos. Environ. 2001, 35, 115–126. [Google Scholar] [CrossRef]
  34. Oh, G.-Y.; Park, G.-H.; Kim, I.-S.; Bae, J.-S.; Park, H.-Y.; Seo, Y.-G.; Yang, S.-I.; Lee, J.-K.; Jeong, S.-H.; Lee, W.-J. Comparison of Major Monoterpene Concentrations in the Ambient Air of South Korea Forests. J. Korean Soc. For. Sci. 2010, 99, 698–705. [Google Scholar]
  35. Lee, J.; Cho, K.S.; Jeon, Y.; Kim, J.B.; Lim, Y.-R.; Lee, K.; Lee, I.-S. Characteristics and distribution of terpenes in South Korean forests. Hangug Hwangyeong Saengtae Haghoeji 2017, 41, 19. [Google Scholar] [CrossRef] [Green Version]
  36. Lerdau, M.; Litvak, M.; Palmer, P.; Monson, R. Controls over monoterpene emissions from boreal forest conifers. Tree Physiol. 1997, 17, 563–569. [Google Scholar] [CrossRef] [PubMed]
  37. Llusia, J.; Peñuelas, J.; Guenther, A.; Rapparini, F. Seasonal variations in terpene emission factors of dominant species in four ecosystems in NE Spain. Atmos. Environ. 2013, 70, 149–158. [Google Scholar] [CrossRef] [Green Version]
  38. Kesselmeier, J.; Staudt, M. Biogenic volatile organic compounds (VOC): An overview on emission, physiology and ecology. J. Atmos. Chem. 1999, 33, 23–88. [Google Scholar] [CrossRef]
  39. Isidorov, V.A.; Zenkevich, I.G.; Ioffe, B.V. Volatile organic compounds in the atmosphere of forests. Atmos. Environ. 1985, 19, 1–8. [Google Scholar] [CrossRef]
  40. Peters, R.J.B.; Johannes, A.D.V.; Duivenbode, R.V.; Duyzer, J.H.; Verhagen, H.L.M. The determination of terpenes in forest air. Atmos. Environ. 1994, 28, 2413–2419. [Google Scholar] [CrossRef]
  41. Kim, J.-C.; Kim, K.-H. Factors Affecting Ambient Monoterpene Levels in a Pine Forest. J. Environ. Sci. Int. 2002, 11, 811–818. [Google Scholar] [CrossRef] [Green Version]
  42. Lee, Y.-K.; Woo, J.-S.; Choi, S.-R.; Shin, E.-S. Comparison of Phytoncide (monoterpene) Concentration by Type of Recreational Forest. J. Environ. Health Sci. 2015, 41, 241–248. [Google Scholar] [CrossRef] [Green Version]
  43. Noe, S.M.; Hüve, K.; Niinemets, Ü.; Copolovici, L. Seasonal variation in vertical volatile compounds air concentrations within a remote hemiboreal mixed forest. Atmos. Chem. Phys. 2012, 12, 3909–3926. [Google Scholar] [CrossRef] [Green Version]
  44. Janson, R. Monoterpene concentrations in and above a forest of scots pine. J. Atmos. Chem. 1992, 14, 385–394. [Google Scholar] [CrossRef]
  45. Falk Filipsson, A.; Bard, J.; Karlsson, S. Concise International Chemical Assessment Document 5: Limonene. Available online: https://www.who.int/ipcs/publications/cicad/en/cicad05.pdf?ua=1#:~:text=Limonene%2C%20like%20other%20monoterpenes%2C%20occurs,large%20(Str%C3%B6mvall%2C%201992) (accessed on 27 August 2020).
  46. Courtois, E.A.; Paine, C.E.T.; Blandinieres, P.-A.; Stien, D.; Bessiere, J.-M.; Houel, E.; Baraloto, C.; Chave, J. Diversity of the volatile organic compounds emitted by 55 species of tropical trees: A survey in French Guiana. J. Chem. Ecol. 2009, 35, 1349–1362. [Google Scholar] [CrossRef]
  47. Kim, L.; Galbally, I.E.; Porter, N.; Weeks, I.A.; Lawson, S.J. BVOC emissions from mechanical wounding of leaves and branches of Eucalyptus sideroxylon (red ironbark). J. Atmos. Chem. 2011, 68, 265–279. [Google Scholar] [CrossRef]
  48. Helmig, D.; Klinger, L.F.; Guenther, A.; Vierling, L.; Geron, C.; Zimmerman, P. Biogenic volatile organic compound emissions (BVOCs). I. Identifications from three continental sites in the U.S. Chemosphere 1999, 38, 2163–2187. [Google Scholar] [CrossRef] [Green Version]
  49. Courtois, E.A.; Dexter, K.G.; Paine, C.E.T.; Stien, D.; Engel, J.; Baraloto, C.; Chave, J. Evolutionary patterns of volatile terpene emissions across 202 tropical tree species. Ecol. Evol. 2016, 6, 2854–2864. [Google Scholar] [CrossRef] [Green Version]
  50. Brasseur, G.P.; Prinn, R.G.; Pszenny, A.A.P. Atmospheric Chemistry in a Changing World: An Integration and Synthesis of a Decade of Tropospheric Chemistry Research; Springer: Berlin/Heidelberg, Germany, 2003; ISBN 9783642623967. [Google Scholar]
  51. Holopainen, J.K.; Blande, J.D. Where do herbivore-induced plant volatiles go? Front. Plant Sci. 2013, 4, 185. [Google Scholar] [CrossRef] [Green Version]
  52. Natürliche Aromatische Rohstoffe—Vokabular; Beuth Verlag GmbH: Berlin, Germany, 2015.
  53. Senatore, F. Oli Essenziali; EMSI: Rome, Italy, 2000. [Google Scholar]
  54. Rai, M.; Kon, K. Fighting Multidrug Resistance with Herbal Extracts, Essential Oils and Their Components; Academic Press: Cambridge, MA, USA, 2013; ISBN 9780124017085. [Google Scholar]
  55. Bagetta, G.; Cosentino, M.; Sakurada, T. Aromatherapy: Basic Mechanisms and Evidence Based Clinical Use; CRC Press: Boca Raton, FL, USA, 2015; ISBN 9781482246643. [Google Scholar]
  56. Buckle, J. Clinical Aromatherapy: Essential Oils in Healthcare; Elsevier: Amsterdam, The Netherlands, 2014; ISBN 9780702054402. [Google Scholar]
  57. Husnu Can Baser, K.; Buchbauer, G. Handbook of Essential Oils: Science, Technology, and Applications, 2nd ed.; CRC Press: Boca Raton, FL, USA, 2015; ISBN 9781466590472. [Google Scholar]
  58. Sumitomo, K.; Akutsu, H.; Fukuyama, S.; Minoshima, A.; Kukita, S.; Yamamura, Y.; Sato, Y.; Hayasaka, T.; Osanai, S.; Funakoshi, H.; et al. Conifer-Derived Monoterpenes and Forest Walking. Mass Spectrom. 2015, 4, A0042. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Miller, J.A.; Hakim, I.A.; Chew, W.; Thompson, P.; Thomson, C.A.; Chow, H.-H.S. Adipose tissue accumulation of d-limonene with the consumption of a lemonade preparation rich in d-limonene content. Nutr. Cancer 2010, 62, 783–788. [Google Scholar] [CrossRef] [PubMed]
  60. Schmidt, L.; Göen, T. Human metabolism of α-pinene and metabolite kinetics after oral administration. Arch. Toxicol. 2017, 91, 677–687. [Google Scholar] [CrossRef]
  61. Peterfalvi, A.; Miko, E.; Nagy, T.; Reger, B.; Simon, D.; Miseta, A.; Czéh, B.; Szereday, L. Much More than a Pleasant Scent: A Review on Essential Oils Supporting the Immune System. Molecules 2019, 24, 4530. [Google Scholar] [CrossRef] [Green Version]
  62. Li, Q.; Kobayashi, M.; Wakayama, Y.; Inagaki, H.; Katsumata, M.; Hirata, Y.; Hirata, K.; Shimizu, T.; Kawada, T.; Park, B.J.; et al. Effect of phytoncide from trees on human natural killer cell function. Int. J. Immunopathol. Pharmacol. 2009, 22, 951–959. [Google Scholar] [CrossRef]
  63. Franco, L.S.; Shanahan, D.F.; Fuller, R.A. A Review of the Benefits of Nature Experiences: More than Meets the Eye. Int. J. Environ. Res. Public Health 2017, 14, 864. [Google Scholar] [CrossRef] [Green Version]
  64. Li, Q.; Nakadai, A.; Matsushima, H.; Miyazaki, Y.; Krensky, A.M.; Kawada, T.; Morimoto, K. Phytoncides (Wood Essential Oils) Induce Human Natural Killer Cell Activity. Immunopharmacol. Immunotoxicol. 2006, 28, 319–333. [Google Scholar] [CrossRef] [PubMed]
  65. Kang, S.; Lee, J.S.; Lee, H.C.; Petriello, M.C.; Kim, B.Y.; Do, J.T.; Lim, D.-S.; Lee, H.G.; Han, S.G. Phytoncide Extracted from Pinecone Decreases LPS-Induced Inflammatory Responses in Bovine Mammary Epithelial Cells. J. Microbiol. Biotechnol. 2016, 26, 579–587. [Google Scholar] [CrossRef] [PubMed]
  66. Cho, J.Y.; Chang, H.-J.; Lee, S.-K.; Kim, H.-J.; Hwang, J.-K.; Chun, H.S. Amelioration of dextran sulfate sodium-induced colitis in mice by oral administration of beta-caryophyllene, a sesquiterpene. Life Sci. 2007, 80, 932–939. [Google Scholar] [CrossRef] [PubMed]
  67. Kim, M.-G.; Kim, S.-M.; Min, J.-H.; Kwon, O.-K.; Park, M.-H.; Park, J.-W.; Ahn, H.I.; Hwang, J.-Y.; Oh, S.-R.; Lee, J.-W.; et al. Anti-inflammatory effects of linalool on ovalbumin-induced pulmonary inflammation. Int. Immunopharmacol. 2019, 74, 105706. [Google Scholar] [CrossRef] [PubMed]
  68. Siqueira, H.D.S.; Neto, B.S.; Sousa, D.P.; Gomes, B.S.; da Silva, F.V.; Cunha, F.V.M.; Wanderley, C.W.S.; Pinheiro, G.; Cândido, A.G.F.; Wong, D.V.T.; et al. α-Phellandrene, a cyclic monoterpene, attenuates inflammatory response through neutrophil migration inhibition and mast cell degranulation. Life Sci. 2016, 160, 27–33. [Google Scholar] [CrossRef] [PubMed]
  69. Zhao, C.; Sun, J.; Fang, C.; Tang, F. 1,8-cineol attenuates LPS-induced acute pulmonary inflammation in mice. Inflammation 2014, 37, 566–572. [Google Scholar] [CrossRef]
  70. Rufino, A.T.; Ribeiro, M.; Sousa, C.; Judas, F.; Salgueiro, L.; Cavaleiro, C.; Mendes, A.F. Evaluation of the anti-inflammatory, anti-catabolic and pro-anabolic effects of E-caryophyllene, myrcene and limonene in a cell model of osteoarthritis. Eur. J. Pharmacol. 2015, 750, 141–150. [Google Scholar] [CrossRef]
  71. Wang, L.; Liang, Q.; Lin, A.; Wu, Y.; Min, H.; Song, S.; Wang, Y.; Wang, H.; Yi, L.; Gao, Q. Borneol alleviates brain injury in sepsis mice by blocking neuronal effect of endotoxin. Life Sci. 2019, 232, 116647. [Google Scholar] [CrossRef]
  72. Bento, A.F.; Marcon, R.; Dutra, R.C.; Claudino, R.F.; Cola, M.; Leite, D.F.P.; Calixto, J.B. β-Caryophyllene inhibits dextran sulfate sodium-induced colitis in mice through CB2 receptor activation and PPARγ pathway. Am. J. Pathol. 2011, 178, 1153–1166. [Google Scholar] [CrossRef]
  73. Horváth, B.; Mukhopadhyay, P.; Kechrid, M.; Patel, V.; Tanchian, G.; Wink, D.A.; Gertsch, J.; Pacher, P. β-Caryophyllene ameliorates cisplatin-induced nephrotoxicity in a cannabinoid 2 receptor-dependent manner. Free Radic. Biol. Med. 2012, 52, 1325–1333. [Google Scholar] [CrossRef] [Green Version]
  74. Li, Y.; Lv, O.; Zhou, F.; Li, Q.; Wu, Z.; Zheng, Y. Linalool Inhibits LPS-Induced Inflammation in BV2 Microglia Cells by Activating Nrf2. Neurochem. Res. 2015, 40, 1520–1525. [Google Scholar] [CrossRef]
  75. Hur, J.; Pak, S.C.; Koo, B.-S.; Jeon, S. Borneol alleviates oxidative stress via upregulation of Nrf2 and Bcl-2 in SH-SY5Y cells. Pharm. Biol. 2013, 51, 30–35. [Google Scholar] [CrossRef] [PubMed]
  76. Lou, J.; Cao, G.; Li, R.; Liu, J.; Dong, Z.; Xu, L. β-Caryophyllene Attenuates Focal Cerebral Ischemia-Reperfusion Injury by Nrf2/HO-1 Pathway in Rats. Neurochem. Res. 2016, 41, 1291–1304. [Google Scholar] [CrossRef] [PubMed]
  77. Cutillas, A.-B.; Carrasco, A.; Martinez-Gutierrez, R.; Tomas, V.; Tudela, J. Thymus mastichina L. essential oils from Murcia (Spain): Composition and antioxidant, antienzymatic and antimicrobial bioactivities. PLoS ONE 2018, 13, e0190790. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Scherer, M.M.d.C.; de Christo Scherer, M.M.; Marques, F.M.; Figueira, M.M.; Peisino, M.C.O.; Schmitt, E.F.P.; Kondratyuk, T.P.; Endringer, D.C.; Scherer, R.; Fronza, M. Wound healing activity of terpinolene and α-phellandrene by attenuating inflammation and oxidative stress in vitro. J. Tissue Viability 2019, 28, 94–99. [Google Scholar] [CrossRef] [PubMed]
  79. Hwang, E.; Ngo, H.T.T.; Park, B.; Seo, S.-A.; Yang, J.-E.; Yi, T.-H. Myrcene, an Aromatic Volatile Compound, Ameliorates Human Skin Extrinsic Aging via Regulation of MMPs Production. Am. J. Chin. Med. 2017, 45, 1113–1124. [Google Scholar] [CrossRef]
  80. Porres-Martínez, M.; González-Burgos, E.; Carretero, M.E.; Gómez-Serranillos, M.P. Major selected monoterpenes α-pinene and 1,8-cineole found in Salvia lavandulifolia (Spanish sage) essential oil as regulators of cellular redox balance. Pharm. Biol. 2015, 53, 921–929. [Google Scholar] [CrossRef]
  81. Khan, A.; Vaibhav, K.; Javed, H.; Tabassum, R.; Ahmed, M.E.; Khan, M.M.; Khan, M.B.; Shrivastava, P.; Islam, F.; Siddiqui, M.S.; et al. 1,8-cineole (eucalyptol) mitigates inflammation in amyloid Beta toxicated PC12 cells: Relevance to Alzheimer’s disease. Neurochem. Res. 2014, 39, 344–352. [Google Scholar] [CrossRef]
  82. Hu, Y.; Zeng, Z.; Wang, B.; Guo, S. Trans-caryophyllene inhibits amyloid β (Aβ) oligomer-induced neuroinflammation in BV-2 microglial cells. Int. Immunopharmacol. 2017, 51, 91–98. [Google Scholar] [CrossRef]
  83. Guo, K.; Mou, X.; Huang, J.; Xiong, N.; Li, H. Trans-caryophyllene suppresses hypoxia-induced neuroinflammatory responses by inhibiting NF-κB activation in microglia. J. Mol. Neurosci. 2014, 54, 41–48. [Google Scholar] [CrossRef]
  84. Shin, M.; Liu, Q.F.; Choi, B.; Shin, C.; Lee, B.; Yuan, C.; Song, Y.J.; Yun, H.S.; Lee, I.-S.; Koo, B.-S.; et al. Neuroprotective Effects of Limonene (+) against Aβ42-Induced Neurotoxicity in a Drosophila Model of Alzheimer’s Disease. Biol. Pharm. Bull. 2020, 43, 409–417. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Sabogal-Guáqueta, A.M.; Osorio, E.; Cardona-Gómez, G.P. Linalool reverses neuropathological and behavioral impairments in old triple transgenic Alzheimer’s mice. Neuropharmacology 2016, 102, 111–120. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Hansen, M.M.; Jones, R.; Tocchini, K. Shinrin-Yoku (Forest Bathing) and Nature Therapy: A State-of-the-Art Review. Int. J. Environ. Res. Public Health 2017, 14, 851. [Google Scholar] [CrossRef]
  87. Oh, B.; Lee, K.J.; Zaslawski, C.; Yeung, A.; Rosenthal, D.; Larkey, L.; Back, M. Health and well-being benefits of spending time in forests: Systematic review. Environ. Health Prev. Med. 2017, 22, 71. [Google Scholar] [CrossRef]
  88. do Vale, T.G.; do Vale, T.G.; Couto Furtado, E.; Santos, J.G.; Viana, G.S.B. Central effects of citral, myrcene and limonene, constituents of essential oil chemotypes from Lippia alba (Mill.) N.E. Brown. Phytomedicine 2002, 9, 709–714. [Google Scholar] [CrossRef] [PubMed]
  89. Silva, M.I.G.; de Aquino Neto, M.R.; Teixeira Neto, P.F.; Moura, B.A.; do Amaral, J.F.; de Sousa, D.P.; Vasconcelos, S.M.M.; de Sousa, F.C.F. Central nervous system activity of acute administration of isopulegol in mice. Pharmacol. Biochem. Behav. 2007, 88, 141–147. [Google Scholar] [CrossRef] [PubMed]
  90. Cheng, W.-W.; Lin, C.-T.; Chu, F.-H.; Chang, S.-T.; Wang, S.-Y. Neuropharmacological activities of phytoncide released from Cryptomeria japonica. J. Wood Sci. 2009, 55, 27–31. [Google Scholar] [CrossRef]
  91. Woo, J.; Lee, C.J. Sleep-enhancing Effects of Phytoncide via Behavioral, Electrophysiological, and Molecular Modeling Approaches. Exp. Neurobiol. 2020, 29, 120–129. [Google Scholar] [CrossRef]
  92. Granger, R.E.; Campbell, E.L.; Johnston, G.A.R. (+)- And (−)-borneol: Efficacious positive modulators of GABA action at human recombinant alpha1beta2gamma2L GABA(A) receptors. Biochem. Pharmacol. 2005, 69, 1101–1111. [Google Scholar] [CrossRef]
  93. Kessler, A.; Sahin-Nadeem, H.; Lummis, S.C.R.; Weigel, I.; Pischetsrieder, M.; Buettner, A.; Villmann, C. GABA(A) receptor modulation by terpenoids from Sideritis extracts. Mol. Nutr. Food Res. 2014, 58, 851–862. [Google Scholar] [CrossRef] [Green Version]
  94. Linck, V.M.; da Silva, A.L.; Figueiró, M.; Caramão, E.B.; Moreno, P.R.H.; Elisabetsky, E. Effects of inhaled Linalool in anxiety, social interaction and aggressive behavior in mice. Phytomedicine 2010, 17, 679–683. [Google Scholar] [CrossRef] [PubMed]
  95. Souto-Maior, F.N.; de Carvalho, F.L.; de Morais, L.C.S.L.; Netto, S.M.; de Sousa, D.P.; de Almeida, R.N. Anxiolytic-like effects of inhaled linalool oxide in experimental mouse anxiety models. Pharmacol. Biochem. Behav. 2011, 100, 259–263. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Guzmán-Gutiérrez, S.L.; Bonilla-Jaime, H.; Gómez-Cansino, R.; Reyes-Chilpa, R. Linalool and β-pinene exert their antidepressant-like activity through the monoaminergic pathway. Life Sci. 2015, 128, 24–29. [Google Scholar] [CrossRef] [PubMed]
  97. Guzmán-Gutiérrez, S.L.; Gómez-Cansino, R.; García-Zebadúa, J.C.; Jiménez-Pérez, N.C.; Reyes-Chilpa, R. Antidepressant activity of Litsea glaucescens essential oil: Identification of β-pinene and linalool as active principles. J. Ethnopharmacol. 2012, 143, 673–679. [Google Scholar] [CrossRef]
  98. Song, C.; Ikei, H.; Miyazaki, Y. Physiological Effects of Nature Therapy: A Review of the Research in Japan. Int. J. Environ. Res. Public Health 2016, 13, 781. [Google Scholar] [CrossRef]
  99. Antonelli, M.; Barbieri, G.; Donelli, D. Effects of forest bathing (shinrin-yoku) on levels of cortisol as a stress biomarker: A systematic review and meta-analysis. Int. J. Biometeorol. 2019, 63, 1117–1134. [Google Scholar] [CrossRef]
  100. Kawakami, K.; Kawamoto, M.; Nomura, M.; Otani, H.; Nabika, T.; Gonda, T. Effects of phytoncides on blood pressure under restraint stress in SHRSP. Clin. Exp. Pharmacol. Physiol. 2004, 31 (Suppl. S2), S27–S28. [Google Scholar] [CrossRef]
  101. Akutsu, H.; Kikusui, T.; Takeuchi, Y.; Sano, K.; Hatanaka, A.; Mori, Y. Alleviating effects of plant-derived fragrances on stress-induced hyperthermia in rats. Physiol. Behav. 2002, 75, 355–360. [Google Scholar] [CrossRef]
  102. Kusuhara, M.; Maruyama, K.; Ishii, H.; Masuda, Y.; Sakurai, K.; Tamai, E.; Urakami, K. A Fragrant Environment Containing α-Pinene Suppresses Tumor Growth in Mice by Modulating the Hypothalamus/Sympathetic Nerve/Leptin Axis and Immune System. Integr. Cancer Ther. 2019, 18, 1534735419845139. [Google Scholar] [CrossRef]
  103. Okano, S.; Honda, Y.; Kodama, T.; Kimura, M. The Effects of Frankincense Essential Oil on Stress in Rats. J. Oleo Sci. 2019, 68, 1003–1009. [Google Scholar] [CrossRef] [Green Version]
  104. Tang, X.-P.; Guo, X.-H.; Geng, D.; Weng, L.-J. d-Limonene protects PC12 cells against corticosterone-induced neurotoxicity by activating the AMPK pathway. Environ. Toxicol. Pharmacol. 2019, 70, 103192. [Google Scholar] [CrossRef] [PubMed]
  105. Thomas, A.F.; Bessière, Y. Limonene. Nat. Prod. Rep. 1989, 6, 291–309. [Google Scholar] [CrossRef]
  106. Tisserand, R.; Young, R. Essential Oil Safety: A Guide for Health Care Professionals; Churchill Livingstone/Elsevier: London, UK, 2013; ISBN 9780443062414. [Google Scholar]
  107. Souza, M.C.; Siani, A.C.; Ramos, M.F.S.; Menezes-de-Lima, O.J.; Henriques, M.G.M.O. Evaluation of anti-inflammatory activity of essential oils from two Asteraceae species. Pharmazie 2003, 58, 582–586. [Google Scholar] [PubMed]
  108. Santana, H.S.R.; de Carvalho, F.O.; Silva, E.R.; Santos, N.G.L.; Shanmugam, S.; Santos, D.N.; Wisniewski, J.O.; Junior, J.S.C.; Nunes, P.S.; Araujo, A.A.S.; et al. Anti-Inflammatory Activity of Limonene in the Prevention and Control of Injuries in the Respiratory System: A Systematic Review. Curr. Pharm. Des. 2020, 26, 2182–2191. [Google Scholar] [CrossRef] [PubMed]
  109. Ostan, R.; Béné, M.C.; Spazzafumo, L.; Pinto, A.; Donini, L.M.; Pryen, F.; Charrouf, Z.; Valentini, L.; Lochs, H.; Bourdel-Marchasson, I.; et al. Impact of diet and nutraceutical supplementation on inflammation in elderly people. Results from the RISTOMED study, an open-label randomized control trial. Clin. Nutr. 2016, 35, 812–818. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Blowman, K.; Magalhães, M.; Lemos, M.F.L.; Cabral, C.; Pires, I.M. Anticancer Properties of Essential Oils and Other Natural Products. Evid. Based Complementary Altern. Med. 2018, 2018, 3149362. [Google Scholar] [CrossRef]
  111. Boehm, K.; Büssing, A.; Ostermann, T. Aromatherapy as an adjuvant treatment in cancer care—A descriptive systematic review. Afr. J. Tradit. Complement. Altern. Med. 2012, 9, 503–518. [Google Scholar] [CrossRef]
  112. Wattenberg, L.W.; Coccia, J.B. Inhibition of 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone carcinogenesis in mice by D-limonene and citrus fruit oils. Carcinogenesis 1991, 12, 115–117. [Google Scholar] [CrossRef]
  113. Haag, J.D.; Lindstrom, M.J.; Gould, M.N. Limonene-induced regression of mammary carcinomas. Cancer Res. 1992, 52, 4021–4026. [Google Scholar]
  114. Vigushin, D.M.; Poon, G.K.; Boddy, A.; English, J.; Halbert, G.W.; Pagonis, C.; Jarman, M.; Coombes, R.C. Phase I and pharmacokinetic study of D-limonene in patients with advanced cancer. Cancer Research Campaign Phase I/II Clinical Trials Committee. Cancer Chemother. Pharmacol. 1998, 42, 111–117. [Google Scholar] [CrossRef]
  115. Miller, J.A.; Lang, J.E.; Ley, M.; Nagle, R.; Hsu, C.-H.; Thompson, P.A.; Cordova, C.; Waer, A.; Chow, H.-H.S. Human breast tissue disposition and bioactivity of limonene in women with early-stage breast cancer. Cancer Prev. Res. 2013, 6, 577–584. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Erasto, P.; Viljoen, A.M. Limonene—A Review: Biosynthetic, Ecological and Pharmacological Relevance. Nat. Prod. Commun. 2008, 3, 1934578X0800300728. [Google Scholar] [CrossRef] [Green Version]
  117. Igimi, H.; Tamura, R.; Toraishi, K.; Yamamoto, F.; Kataoka, A.; Ikejiri, Y.; Hisatsugu, T.; Shimura, H. Medical dissolution of gallstones. Clinical experience of d-limonene as a simple, safe, and effective solvent. Dig. Dis. Sci. 1991, 36, 200–208. [Google Scholar] [CrossRef] [PubMed]
  118. Chen, S.-W.; Wu, P.-J.; Chiang, B.-H. In vitro neuropeptide Y mRNA expressing model for screening essences that may affect appetite using Rolf B1.T cells. J. Agric. Food Chem. 2012, 60, 7824–7829. [Google Scholar] [CrossRef] [PubMed]
  119. Yoon, W.-J.; Lee, N.H.; Hyun, C.-G. Limonene suppresses lipopolysaccharide-induced production of nitric oxide, prostaglandin E2, and pro-inflammatory cytokines in RAW 264.7 macrophages. J. Oleo Sci. 2010, 59, 415–421. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Bai, J.; Zheng, Y.; Wang, G.; Liu, P. Protective Effect of D-Limonene against Oxidative Stress-Induced Cell Damage in Human Lens Epithelial Cells via the p38 Pathway. Oxid. Med. Cell. Longev. 2016, 2016, 5962832. [Google Scholar] [CrossRef]
  121. Berliocchi, L.; Chiappini, C.; Adornetto, A.; Gentile, D.; Cerri, S.; Russo, R.; Bagetta, G.; Corasaniti, M.T. Early LC3 lipidation induced by D-limonene does not rely on mTOR inhibition, ERK activation and ROS production and it is associated with reduced clonogenic capacity of SH-SY5Y neuroblastoma cells. Phytomedicine 2018, 40, 98–105. [Google Scholar] [CrossRef]
  122. Del Toro-Arreola, S.; Flores-Torales, E.; Torres-Lozano, C.; Del Toro-Arreola, A.; Tostado-Pelayo, K.; Guadalupe Ramirez-Dueñas, M.; Daneri-Navarro, A. Effect of D-limonene on immune response in BALB/c mice with lymphoma. Int. Immunopharmacol. 2005, 5, 829–838. [Google Scholar] [CrossRef] [PubMed]
  123. Roberto, D.; Micucci, P.; Sebastian, T.; Graciela, F.; Anesini, C. Antioxidant activity of limonene on normal murine lymphocytes: Relation to H2O2 modulation and cell proliferation. Basic Clin. Pharmacol. Toxicol. 2010, 106, 38–44. [Google Scholar] [CrossRef] [PubMed]
  124. Hirota, R.; Nakamura, H.; Bhatti, S.A.; Ngatu, N.R.; Muzembo, B.A.; Dumavibhat, N.; Eitoku, M.; Sawamura, M.; Suganuma, N. Limonene inhalation reduces allergic airway inflammation in Dermatophagoides farinae-treated mice. Inhal. Toxicol. 2012, 24, 373–381. [Google Scholar] [CrossRef] [PubMed]
  125. Chi, G.; Wei, M.; Xie, X.; Soromou, L.W.; Liu, F.; Zhao, S. Suppression of MAPK and NF-κB pathways by limonene contributes to attenuation of lipopolysaccharide-induced inflammatory responses in acute lung injury. Inflammation 2013, 36, 501–511. [Google Scholar] [CrossRef] [PubMed]
  126. Rehman, M.U.; Tahir, M.; Khan, A.Q.; Khan, R.; Oday-O-Hamiza; Lateef, A.; Hassan, S.K.; Rashid, S.; Ali, N.; Zeeshan, M.; et al. D-limonene suppresses doxorubicin-induced oxidative stress and inflammation via repression of COX-2, iNOS, and NFκB in kidneys of Wistar rats. Exp. Biol. Med. 2014, 239, 465–476. [Google Scholar] [CrossRef] [PubMed]
  127. De Almeida, A.A.C.; Silva, R.O.; Nicolau, L.A.D.; de Brito, T.V.; de Sousa, D.P.; Barbosa, A.L.D.R.; de Freitas, R.M.; da Sila Lopes, L.; Medeiros, J.-V.R.; Ferreira, P.M.P. Physio-pharmacological Investigations About the Anti-inflammatory and Antinociceptive Efficacy of (+)-Limonene Epoxide. Inflammation 2017, 40, 511–522. [Google Scholar] [CrossRef] [PubMed]
  128. Yu, L.; Yan, J.; Sun, Z. D-limonene exhibits anti-inflammatory and antioxidant properties in an ulcerative colitis rat model via regulation of iNOS, COX-2, PGE2 and ERK signaling pathways. Mol. Med. Rep. 2017, 15, 2339–2346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Yu, X.; Lin, H.; Wang, Y.; Lv, W.; Zhang, S.; Qian, Y.; Deng, X.; Feng, N.; Yu, H.; Qian, B. D-limonene exhibits antitumor activity by inducing autophagy and apoptosis in lung cancer. OncoTargets Ther. 2018, 11, 1833–1847. [Google Scholar] [CrossRef] [Green Version]
  130. Durço, A.O.; de Souza, D.S.; Heimfarth, L.; Miguel-dos-Santos, R.; Rabelo, T.K.; de Oliveira Barreto, T.; Rhana, P.; Santana, M.N.S.; Braga, W.F.; dos Santos Cruz, J.; et al. d-Limonene Ameliorates Myocardial Infarction Injury by Reducing Reactive Oxygen Species and Cell Apoptosis in a Murine Model. J. Nat. Prod. 2019, 82, 3010–3019. [Google Scholar] [CrossRef]
  131. De Almeida, A.A.C.; de Carvalho, R.B.F.; Silva, O.A.; de Sousa, D.P.; de Freitas, R.M. Potential antioxidant and anxiolytic effects of (+)-limonene epoxide in mice after marble-burying test. Pharmacol. Biochem. Behav. 2014, 118, 69–78. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Yun, J. Limonene inhibits methamphetamine-induced locomotor activity via regulation of 5-HT neuronal function and dopamine release. Phytomedicine 2014, 21, 883–887. [Google Scholar] [CrossRef]
  133. Piccinelli, A.C.; Santos, J.A.; Konkiewitz, E.C.; Oesterreich, S.A.; Formagio, A.S.N.; Croda, J.; Ziff, E.B.; Kassuya, C.A.L. Antihyperalgesic and antidepressive actions of (R)-(+)-limonene, α-phellandrene, and essential oil fromSchinus terebinthifoliusfruits in a neuropathic pain model. Nutr. Neurosci. 2015, 18, 217–224. [Google Scholar] [CrossRef]
  134. Zhang, L.-L.; Yang, Z.-Y.; Fan, G.; Ren, J.-N.; Yin, K.-J.; Pan, S.-Y. Antidepressant-like Effect of Citrus sinensis (L.) Osbeck Essential Oil and Its Main Component Limonene on Mice. J. Agric. Food Chem. 2019, 67, 13817–13828. [Google Scholar] [CrossRef]
  135. Lima, N.G.P.B.; De Sousa, D.P.; Pimenta, F.C.F.; Alves, M.F.; De Souza, F.S.; Macedo, R.O.; Cardoso, R.B.; de Morais, L.C.S.L.; Melo Diniz, M.d.F.F.; de Almeida, R.N. Anxiolytic-like activity and GC-MS analysis of (R)-(+)-limonene fragrance, a natural compound found in foods and plants. Pharmacol. Biochem. Behav. 2013, 103, 450–454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Do Amaral, J.F.; Silva, M.I.G.; Neto, M.R.d.A.; Neto, P.F.T.; Moura, B.A.; de Melo, C.T.V.; de Araújo, F.L.O.; de Sousa, D.P.; de Vasconcelos, P.F.; de Vasconcelos, S.M.M.; et al. Antinociceptive effect of the monoterpene R-(+)-limonene in mice. Biol. Pharm. Bull. 2007, 30, 1217–1220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Piccinelli, A.C.; Morato, P.N.; Dos Santos Barbosa, M.; Croda, J.; Sampson, J.; Kong, X.; Konkiewitz, E.C.; Ziff, E.B.; Amaya-Farfan, J.; Kassuya, C.A.L. Limonene reduces hyperalgesia induced by gp120 and cytokines by modulation of IL-1 β and protein expression in spinal cord of mice. Life Sci. 2017, 174, 28–34. [Google Scholar] [CrossRef] [PubMed]
  138. PubChem Alpha-Pinene. Available online: https://pubchem.ncbi.nlm.nih.gov/compound/6654 (accessed on 18 June 2020).
  139. PubChem Beta-Pinene. Available online: https://pubchem.ncbi.nlm.nih.gov/compound/14896 (accessed on 18 June 2020).
  140. Matthys, H.; de Mey, C.; Carls, C.; Ryś, A.; Geib, A.; Wittig, T. Efficacy and tolerability of myrtol standardized in acute bronchitis. A multi-centre, randomised, double-blind, placebo-controlled parallel group clinical trial vs. cefuroxime and ambroxol. Arzneimittelforschung 2000, 50, 700–711. [Google Scholar] [CrossRef]
  141. Ghavami, M.B.; Ahmadi, S. Effectiveness of eucalyptus and cinnamon essential oils compared to permethrin in treatment of head lice infestation. J. Adv. Med. Biomed. Res. 2017, 25, 86–98. [Google Scholar]
  142. Oladimeji, F.A.; Orafidiya, O.O.; Ogunniyi, T.A.; Adewunmi, T.A. Pediculocidal and scabicidal properties of Lippia multiflora essential oil. J. Ethnopharmacol. 2000, 72, 305–311. [Google Scholar] [CrossRef]
  143. Moss, L.; Rouse, M.; Wesnes, K.A.; Moss, M. Differential effects of the aromas of Salvia species on memory and mood. Hum. Psychopharmacol. 2010, 25, 388–396. [Google Scholar] [CrossRef]
  144. Tildesley, N.T.J.; Kennedy, D.O.; Perry, E.K.; Ballard, C.G.; Savelev, S.; Wesnes, K.A.; Scholey, A.B. Salvia lavandulaefolia (Spanish sage) enhances memory in healthy young volunteers. Pharmacol. Biochem. Behav. 2003, 75, 669–674. [Google Scholar] [CrossRef]
  145. Perry, N.S.L.; Bollen, C.; Perry, E.K.; Ballard, C. Salvia for dementia therapy: Review of pharmacological activity and pilot tolerability clinical trial. Pharmacol. Biochem. Behav. 2003, 75, 651–659. [Google Scholar] [CrossRef]
  146. Matsuo, A.L.; Figueiredo, C.R.; Arruda, D.C.; Pereira, F.V.; Scutti, J.A.B.; Massaoka, M.H.; Travassos, L.R.; Sartorelli, P.; Lago, J.H.G. α-Pinene isolated from Schinus terebinthifolius Raddi (Anacardiaceae) induces apoptosis and confers antimetastatic protection in a melanoma model. Biochem. Biophys. Res. Commun. 2011, 411, 449–454. [Google Scholar] [CrossRef] [Green Version]
  147. Yoshida, N.; Takada, T.; Yamamura, Y.; Adachi, I.; Suzuki, H.; Kawakami, J. Inhibitory effects of terpenoids on multidrug resistance-associated protein 2- and breast cancer resistance protein-mediated transport. Drug Metab. Dispos. 2008, 36, 1206–1211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Chen, W.; Liu, Y.; Li, M.; Mao, J.; Zhang, L.; Huang, R.; Jin, X.; Ye, L. Anti-tumor effect of α-pinene on human hepatoma cell lines through inducing G2/M cell cycle arrest. J. Pharmacol. Sci. 2015, 127, 332–338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Nam, S.-Y.; Chung, C.-K.; Seo, J.-H.; Rah, S.-Y.; Kim, H.-M.; Jeong, H.-J. The therapeutic efficacy of α-pinene in an experimental mouse model of allergic rhinitis. Int. Immunopharmacol. 2014, 23, 273–282. [Google Scholar] [CrossRef] [PubMed]
  150. Soković, M.; Glamočlija, J.; Marin, P.D.; Brkić, D.; van Griensven, L.J.L.D. Antibacterial effects of the essential oils of commonly consumed medicinal herbs using an in vitro model. Molecules 2010, 15, 7532–7546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  151. Elaissi, A.; Rouis, Z.; Mabrouk, S.; Salah, K.B.H.; Aouni, M.; Khouja, M.L.; Farhat, F.; Chemli, R.; Harzallah-Skhiri, F. Correlation between chemical composition and antibacterial activity of essential oils from fifteen Eucalyptus species growing in the Korbous and Jbel Abderrahman arboreta (North East Tunisia). Molecules 2012, 17, 3044–3057. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Kim, D.-S.; Lee, H.-J.; Jeon, Y.-D.; Han, Y.-H.; Kee, J.-Y.; Kim, H.-J.; Shin, H.-J.; Kang, J.; Lee, B.S.; Kim, S.-H.; et al. Alpha-Pinene Exhibits Anti-Inflammatory Activity Through the Suppression of MAPKs and the NF-κB Pathway in Mouse Peritoneal Macrophages. Am. J. Chin. Med. 2015, 43, 731–742. [Google Scholar] [CrossRef]
  153. Rufino, A.T.; Ribeiro, M.; Judas, F.; Salgueiro, L.; Lopes, M.C.; Cavaleiro, C.; Mendes, A.F. Anti-inflammatory and chondroprotective activity of (+)-α-pinene: Structural and enantiomeric selectivity. J. Nat. Prod. 2014, 77, 264–269. [Google Scholar] [CrossRef]
  154. Türkez, H.; Aydın, E. In vitro assessment of cytogenetic and oxidative effects of α-pinene. Toxicol. Ind. Health 2016, 32, 168–176. [Google Scholar] [CrossRef]
  155. Khoshnazar, M.; Bigdeli, M.R.; Parvardeh, S.; Pouriran, R. Attenuating effect of α-pinene on neurobehavioural deficit, oxidative damage and inflammatory response following focal ischaemic stroke in rat. J. Pharm. Pharmacol. 2019, 71, 1725–1733. [Google Scholar] [CrossRef]
  156. Rahbar, I.; Abbasnejad, M.; Haghani, J.; Raoof, M.; Kooshki, R.; Esmaeili-Mahani, S. The effect of central administration of alpha-pinene on capsaicin-induced dental pulp nociception. Int. Endod. J. 2019, 52, 307–317. [Google Scholar] [CrossRef]
  157. Yamaoka, S.; Tomita, T.; Imaizumi, Y.; Watanabe, K.; Hatanaka, A. Effects of plant-derived odors on sleep-wakefulness and circadian rhythmicity in rats. Chem. Senses 2005, 30 (Suppl. S1), i264–i265. [Google Scholar] [CrossRef] [Green Version]
  158. Satou, T.; Kasuya, H.; Maeda, K.; Koike, K. Daily inhalation of α-pinene in mice: Effects on behavior and organ accumulation. Phytother. Res. 2014, 28, 1284–1287. [Google Scholar] [CrossRef] [PubMed]
  159. Yang, H.; Woo, J.; Pae, A.N.; Um, M.Y.; Cho, N.-C.; Park, K.D.; Yoon, M.; Kim, J.; Justin Lee, C.; Cho, S. α-Pinene, a Major Constituent of Pine Tree Oils, Enhances Non-Rapid Eye Movement Sleep in Mice through GABAA-benzodiazepine Receptors. Mol. Pharmacol. 2016, 90, 530–539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Kong, Y.; Wang, T.; Wang, R.; Ma, Y.; Song, S.; Liu, J.; Hu, W.; Li, S. Inhalation of Roman chamomile essential oil attenuates depressive-like behaviors in Wistar Kyoto rats. Sci. China Life Sci. 2017, 60, 647–655. [Google Scholar] [CrossRef] [PubMed]
  161. Ueno, H.; Shimada, A.; Suemitsu, S.; Murakami, S.; Kitamura, N.; Wani, K.; Matsumoto, Y.; Okamoto, M.; Ishihara, T. Attenuation Effects of Alpha-Pinene Inhalation on Mice with Dizocilpine-Induced Psychiatric-Like Behaviour. Evid. Based Complement. Alternat. Med. 2019, 2019, 2745453. [Google Scholar] [CrossRef] [Green Version]
  162. Him, A.; Ozbek, H.; Turel, I.; Oner, A.C. Antinociceptive activity of alpha-pinene and fenchone. Pharmacologyonline 2008, 3, 363–369. [Google Scholar]
  163. Quintão, N.L.; da Silva, G.F.; Antonialli, C.S.; Rocha, L.W.; Cechinel Filho, V.; Cicció, J.F. Chemical composition and evaluation of the anti-hypernociceptive effect of the essential oil extracted from the leaves of Ugni myricoides on inflammatory and neuropathic models of pain in mice. Planta Med. 2010, 76, 1411–1418. [Google Scholar] [CrossRef] [Green Version]
  164. Kennedy, D.; Okello, E.; Chazot, P.; Howes, M.-J.; Ohiomokhare, S.; Jackson, P.; Haskell-Ramsay, C.; Khan, J.; Forster, J.; Wightman, E. Volatile Terpenes and Brain Function: Investigation of the Cognitive and Mood Effects of Mentha × Piperita L. Essential Oil with in Vitro Properties Relevant to Central Nervous System Function. Nutrients 2018, 10, 1029. [Google Scholar] [CrossRef] [Green Version]
  165. Kennedy, D.O.; Wightman, E.L. Herbal extracts and phytochemicals: Plant secondary metabolites and the enhancement of human brain function. Adv. Nutr. 2011, 2, 32–50. [Google Scholar] [CrossRef]
  166. Falk-Filipsson, A.; Löf, A.; Hagberg, M.; Hjelm, E.W.; Wang, Z. d-limonene exposure to humans by inhalation: Uptake, distribution, elimination, and effects on the pulmonary function. J. Toxicol. Environ. Health 1993, 38, 77–88. [Google Scholar] [CrossRef]
  167. Kohlert, C.; van Rensen, I.; März, R.; Schindler, G.; Graefe, E.U.; Veit, M. Bioavailability and pharmacokinetics of natural volatile terpenes in animals and humans. Planta Med. 2000, 66, 495–505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  168. Porres-Martínez, M.; González-Burgos, E.; Carretero, M.E.; Gómez-Serranillos, M.P. In vitro neuroprotective potential of the monoterpenes α-pinene and 1,8-cineole against H2O2-induced oxidative stress in PC12 cells. Z. Naturforsch. C 2016, 71, 191–199. [Google Scholar] [CrossRef] [PubMed]
  169. Lorenzetti, B.B.; Souza, G.E.; Sarti, S.J.; Santos Filho, D.; Ferreira, S.H. Myrcene mimics the peripheral analgesic activity of lemongrass tea. J. Ethnopharmacol. 1991, 34, 43–48. [Google Scholar] [CrossRef]
  170. De-Oliveira, A.C.; Ribeiro-Pinto, L.F.; Paumgartten, J.R. In vitro inhibition of CYP2B1 monooxygenase by beta-myrcene and other monoterpenoid compounds. Toxicol. Lett. 1997, 92, 39–46. [Google Scholar] [CrossRef]
  171. Rao, V.S.; Menezes, A.M.; Viana, G.S. Effect of myrcene on nociception in mice. J. Pharm. Pharmacol. 1990, 42, 877–878. [Google Scholar] [CrossRef] [PubMed]
  172. Suntar, I.; Khan, H.; Patel, S.; Celano, R.; Rastrelli, L. An Overview on Citrus aurantium L.: Its Functions as Food Ingredient and Therapeutic Agent. Oxid. Med. Cell. Longev. 2018, 2018, 7864269. [Google Scholar] [CrossRef] [Green Version]
  173. Kim, S.; Choi, Y.; Choi, S.; Choi, Y.; Park, T. Dietary camphene attenuates hepatic steatosis and insulin resistance in mice. Obesity 2014, 22, 408–417. [Google Scholar] [CrossRef]
  174. Girola, N.; Figueiredo, C.R.; Farias, C.F.; Azevedo, R.A.; Ferreira, A.K.; Teixeira, S.F.; Capello, T.M.; Martins, E.G.A.; Matsuo, A.L.; Travassos, L.R.; et al. Camphene isolated from essential oil of Piper cernuum (Piperaceae) induces intrinsic apoptosis in melanoma cells and displays antitumor activity in vivo. Biochem. Biophys. Res. Commun. 2015, 467, 928–934. [Google Scholar] [CrossRef]
  175. Quintans-Júnior, L.; Moreira, J.C.F.; Pasquali, M.A.B.; Rabie, S.M.S.; Pires, A.S.; Schröder, R.; Rabelo, T.K.; Santos, J.P.A.; Lima, P.S.S.; Cavalcanti, S.C.H.; et al. Antinociceptive Activity and Redox Profile of the Monoterpenes (+)-Camphene, p-Cymene, and Geranyl Acetate in Experimental Models. ISRN Toxicol. 2013, 2013, 459530. [Google Scholar] [CrossRef] [Green Version]
  176. Vallianou, I.; Peroulis, N.; Pantazis, P.; Hadzopoulou-Cladaras, M. Camphene, a plant-derived monoterpene, reduces plasma cholesterol and triglycerides in hyperlipidemic rats independently of HMG-CoA reductase activity. PLoS ONE 2011, 6, e20516. [Google Scholar] [CrossRef] [Green Version]
  177. Van den Berg, M.; Wendel-Vos, W.; van Poppel, M.; Kemper, H.; van Mechelen, W.; Maas, J. Health benefits of green spaces in the living environment: A systematic review of epidemiological studies. Urban For. Urban Green. 2015, 14, 806–816. [Google Scholar] [CrossRef]
  178. Beyer, S.V. Singing to the Plants: A Guide to Mestizo Shamanism in the Upper Amazon; UNM Press: Albuquerque, NM, USA, 2010; ISBN 9780826347305. [Google Scholar]
  179. Hamayon, R. Shamanism and the hunters of the Siberian forest: Soul, life force, spirit. In The Handbook of Contemporary Animism; Acumen Publishing: Slough, UK, 2013; pp. 284–293. [Google Scholar]
  180. Maas, J.; Verheij, R.A.; de Vries, S.; Spreeuwenberg, P.; Schellevis, F.G.; Groenewegen, P.P. Morbidity is related to a green living environment. J. Epidemiol. Community Health 2009, 63, 967–973. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  181. Lee, A.C.K.; Maheswaran, R. The health benefits of urban green spaces: A review of the evidence. J. Public Health 2011, 33, 212–222. [Google Scholar] [CrossRef] [PubMed]
  182. Kotte, D.; Li, Q.; Shin, W.S. International Handbook of Forest Therapy; Cambridge Scholars Publishing: Cambridge, UK, 2019; ISBN 9781527541740. [Google Scholar]
  183. ABOUT US|INFOM. Available online: https://www.infom.org/aboutus/index.html (accessed on 8 July 2020).
  184. Martellucci, S. L’Idea Paesaggio. Caratteri Interattivi del Progetto Architettonico e Urbano; Alinea Editrice: Florence, Italy, 2007; ISBN 9788860551320. [Google Scholar]
  185. Delendi, M.L. Il Progetto di Paesaggio come Dispositivo Terapeutico; Gangemi Editore Spa: Milan, Italy, 2015; ISBN 9788849293487. [Google Scholar]
  186. Locatelli, E. Scent. Esperienze Olfattive; Politecnico di Milano: Milan, Italy, 2014. [Google Scholar]
  187. Gillis, K.; Gatersleben, B. A Review of Psychological Literature on the Health and Wellbeing Benefits of Biophilic Design. Buildings 2015, 5, 948–963. [Google Scholar] [CrossRef] [Green Version]
  188. Marcus, C.C.; Barnes, M. Healing Gardens: Therapeutic Benefits and Design Recommendations; John Wiley & Sons: Hoboken, NJ, USA, 1999; ISBN 9780471192039. [Google Scholar]
  189. Mencagli, M.; Nieri, M. La Terapia Segreta degli Alberi. L’Energia Nascosta delle Piante e dei Boschi per il Nostro Benessere; Sperling & Kupfer: Milan, Italy, 2020; ISBN 9788855440035. [Google Scholar]
  190. Firenzuoli, F. Lo Smart Garden del CERFIT (Careggi A.O.U.). Available online: https://www.regione.toscana.it/documents/10180/15989178/MC_41_careggi.pdf/d7e05406-afdc-4867-b8db-f3f94e9cf1f9 (accessed on 29 July 2020).
Figure 1. BVOCs’ functions in relation to biotic and abiotic stresses. Adapted from Laothawornkitkul et al. (2009) [8].
Figure 1. BVOCs’ functions in relation to biotic and abiotic stresses. Adapted from Laothawornkitkul et al. (2009) [8].
Ijerph 17 06506 g001
Figure 2. Main biosynthetic pathways for BVOCs. In green and yellow colors, the methylerythritol phosphate (MEP) and mevalonate (MVA) pathways to isoprenoids, respectively, in blue color, the lipoxygenase (LOX) pathway to GLVs, and in purple color, the shikimate pathway to aromatic compounds. Adapted from Maffei et al. (2011) [17].
Figure 2. Main biosynthetic pathways for BVOCs. In green and yellow colors, the methylerythritol phosphate (MEP) and mevalonate (MVA) pathways to isoprenoids, respectively, in blue color, the lipoxygenase (LOX) pathway to GLVs, and in purple color, the shikimate pathway to aromatic compounds. Adapted from Maffei et al. (2011) [17].
Ijerph 17 06506 g002
Figure 3. Atmospheric concentration of BVOCs under forest canopy [16,31,32,34,38,39,40,41,42,43,44]. Abbreviations: Tot. terpenes = total concentration of terpenes; LOX = products of the lipoxygenase (LOX) pathway.
Figure 3. Atmospheric concentration of BVOCs under forest canopy [16,31,32,34,38,39,40,41,42,43,44]. Abbreviations: Tot. terpenes = total concentration of terpenes; LOX = products of the lipoxygenase (LOX) pathway.
Ijerph 17 06506 g003
Figure 4. Atmospheric concentrations of seven monoterpenes under the canopy of a Scots pine forest [44].
Figure 4. Atmospheric concentrations of seven monoterpenes under the canopy of a Scots pine forest [44].
Ijerph 17 06506 g004
Table 1. Functions of constitutive and herbivore-induced forest Volatile Organic Compounds (VOCs).
Table 1. Functions of constitutive and herbivore-induced forest Volatile Organic Compounds (VOCs).
Constitutive Forest VOCsHerbivore-Induced Plant Volatiles (HIPVs)
Reduction of abiotic stress. Isoprene and monoterpenes increase general thermal tolerance of photosynthesis, protect photosynthetic apparatus and its activity under high-temperature stress by stabilizing the thylakoid membranes and quenching Reactive Oxygen Species (ROS)Reduction of abiotic stress. Isoprene and monoterpenes increase general thermal tolerance of photosynthesis, protect photosynthetic apparatus and its activity under high-temperature stress by stabilizing the thylakoid membranes and quenching Reactive Oxygen Species (ROS)
Defense against herbivores. Comprises toxic, repellent, anti-nutritive constitutive BVOCs (biogenic Volatile Organic Compounds) or HIPVs, as well as growth and reproductive reducersDefense against herbivores, mainly indirectly but also directly. HIPVs and volatile compounds that attract, nourish, or otherwise favor another organism that reduces herbivore pressure
Inter-plant signaling. HIPVs, especially Green Leaf Volatiles (GLVs), and constitutive BVOCs can travel from a herbivore-damaged part to other plants (both conspecific and heterospecific), activating defense genes and priming a more vigorous response after an attackInter- and intra-plant signaling. HIPVs, especially GLVs, and constitutive BVOCs can travel from a herbivore-damaged part to an undamaged one, or to other plants (both conspecific and heterospecific), activating defense genes and priming a more vigorous response after an attack
Defense against microbial pathogensDefense against microbial pathogens
Allelopathy. Inhibition of competing species’ seed germination and competition
Attraction of pollinators and seed dispersers
Table 2. Forest BVOCs and their chemical characteristics listed on the basis of the magnitude of emissions (in descending order).
Table 2. Forest BVOCs and their chemical characteristics listed on the basis of the magnitude of emissions (in descending order).
MoleculeChemical FamilyIUPACFormulaStructureCAS numberBoiling Point (at 760 mmHg)Molar Mass (g/mol)I/C 1C/D 2E 3P 4
IsopreneIsoprenoids2-methylbuta-1,3-dieneC5H8 Ijerph 17 06506 i00178-79-534.1 °C68.12CD****+++
cis-3-Hexen-1-olGLVs(Z)-hex-3-en-1-olC6H12O Ijerph 17 06506 i002928-96-1156.5 °C100.16ID***+++
cis-3-HexenalGLVs(Z)-hex-3-enalC6H10O Ijerph 17 06506 i0036789-80-6126 °C98.14ID***+++
cis-3-Hexenyl acetateGLVs[(Z)-hex-3-enyl] acetateC8H14O2 Ijerph 17 06506 i0043681-71-8174.2 °C142.2ID***+++
d-LimoneneMonoterpene hydrocarbons(4R)-1-methyl-4-prop-1-en-2-ylcyclohexeneC10H16 Ijerph 17 06506 i00565996-98-7175.4 °C136.23C, ID***+/+++
α-PineneMonoterpene hydrocarbons2,6,6-trimethylbicyclo[3.1.1]hept-2-eneC10H16 Ijerph 17 06506 i00667762-73-6156 °C136.23C, ID***+/+++
(E)-β-OcimeneMonoterpene hydrocarbons(3E)-3,7-dimethylocta-1,3,6-trieneC10H16 Ijerph 17 06506 i0073779-61-1175.2 °C136.23C, ID**+/++
1,8-CineoleMonoterpenoid ethers1,3,3-trimethyl-2-oxabicyclo[2.2.2]octaneC10H18O Ijerph 17 06506 i008470-82-6176.4 °C154.25C, ID**
CamphorMonoterpenoid ketones1,7,7-trimethylbicyclo[2.2.1]heptan-2-oneC10H16O Ijerph 17 06506 i00976-22-2205.7 °C152.23 **
LinaloolMonoterpenoid alcohol3,7-dimethylocta-1,6-dien-3-olC10H18O Ijerph 17 06506 i01078-70-6197.5 °C154.25C, LD**+/++
p-CymeneAromatic monoterpene hydrocarbons1-methyl-4-propan-2-ylbenzeneC10H14 Ijerph 17 06506 i01199-87-6177 °C134.22CD**
SabineneMonoterpene hydrocarbons4-methylidene-1-propan-2-ylbicyclo[3.1.0]hexaneC10H16 Ijerph 17 06506 i0123387-41-5164 °C136.23CD**
β-CaryophylleneSesquiterpene hydrocarbons(1R,4E,9S)-4,11,11-trimethyl-8-methylidenebicyclo[7.2.0]undec-4-eneC15H24 Ijerph 17 06506 i01387-44-5NA204.35C, ID**+/++
β-MyrceneMonoterpene hydrocarbons7-methyl-3-methylideneocta-1,6-dieneC10H16 Ijerph 17 06506 i014123-35-3167 °C136.23CD**
β-PineneMonoterpene hydrocarbons6,6-dimethyl-2-methylidenebicyclo[3.1.1]heptaneC10H16 Ijerph 17 06506 i015127-91-3166.0 °C136.234C, ID**
β 3-CareneMonoterpene hydrocarbons3,7,7-trimethylbicyclo[4.1.0]hept-3-eneC10H16 Ijerph 17 06506 i01674806-04-5171.4 °C136.234CD**
(E)-Linalool-oxideMonoterpenoid oxide2-[(2S,5S)-5-ethenyl-5-methyloxolan-2-yl]propan-2-olC10H18O2 Ijerph 17 06506 i01711063-78-8NA170.25CD*
(Z)-Linalool-oxideMonoterpenoid oxide2-(5-ethenyl-5-methyloxolan-2-yl)propan-2-olC10H18O2 Ijerph 17 06506 i01814049-11-7224.2 °C170.25CD*
BorneolMonoterpenoid alcohol1,7,7-trimethylbicyclo[2.2.1]heptan-2-olC10H18O Ijerph 17 06506 i019464-45-9 212.0 °C154.25C *
Bornyl acetateMonoteropene-derived ester(1,7,7-trimethyl-2-bicyclo[2.2.1]heptanyl) acetateC12H20O2 Ijerph 17 06506 i02020347-65-3223.5 °C196.286C *
CampheneMonoterpene hydrocarbons2,2-dimethyl-3-methylidenebicyclo[2.2.1]heptaneC10H16 Ijerph 17 06506 i02179-92-5157.5 °C136.234CD*
Terpinen-4-olMonoterpenoid alcohol4-methyl-1-propan-2-ylcyclohex-3-en-1-olC10H18O Ijerph 17 06506 i022562-74-3209.0 °C154.25C *
α-CopaeneSesquiterpene hydrocarbons(1R)-1,3-dimethyl-8-propan-2-yltricyclo[4.4.0.02,7]dec-3-eneC15H24 Ijerph 17 06506 i0233856-25-5248.5 °C204.35I *
α-HumuleneSesquiterpene hydrocarbons(1E,4E,8E)-2,6,6,9-tetramethylcycloundeca-1,4,8-trieneC15H24 Ijerph 17 06506 i0246753-98-6166-168 °C204.35CD*
α-PhellandreneMonoterpene hydrocarbons2-methyl-5-propan-2-ylcyclohexa-1,3-dieneC10H16 Ijerph 17 06506 i02599-83-2171.5 °C136.234C *
α-TerpineneMonoterpene hydrocarbons1-methyl-4-propan-2-ylcyclohexa-1,3-dieneC10H16 Ijerph 17 06506 i02699-86-5174.1 °C136.234CD*
α-TerpineolMonoterpenoid alcohol2-(4-methylcyclohex-3-en-1-yl)propan-2-olC10H18O Ijerph 17 06506 i02710482-56-1217.5 °C154.249C *
α-TerpinoleneMonoterpene hydrocarbons1-methyl-4-propan-2-ylidenecyclohexeneC10H16 Ijerph 17 06506 i0281124-27-2186.0 °C138.25CD*
β-PhellandreneMonoterpene hydrocarbons3-methylidene-6-propan-2-ylcyclohexeneC10H16 Ijerph 17 06506 i029555-10-2175 °C136.234C, I *+/+++
β-TerpineneMonoterpene hydrocarbons1-methyl-4-propan-2-ylcyclohexa-1,4-dieneC10H16 Ijerph 17 06506 i03099-85-4183.0 °C136.234C, ID*
(Z)-β-OcimeneMonoterpene hydrocarbons(3Z)-3,7-dimethylocta-1,3,6-trieneC10H16 Ijerph 17 06506 i03113877-91-3175.2 °C136.234C, ID +/++
BergamoteneSesquiterpene hydrocarbons6-methyl-2-methylidene-6-(4-methylpent-3-enyl)bicyclo[3.1.1]heptaneC15H24 Ijerph 17 06506 i0327663-66-3NA208.38I
DMNTHomoterpene hydrocarbons4,8-dimethylnona-1,3,7-trieneC11H18 Ijerph 17 06506 i03319945-61-0195.6 °C150.26I +/++
LongifoleneSesquiterpene hydrocarbons3,3,7-trimethyl-8-methylidenetricyclo[5.4.0.02,9]undecaneC15H24 Ijerph 17 06506 i034475-20-7252.2 °C204.35CD
Methyl jasmonateJasmonate estermethyl 2-[(1R,2R)-3-oxo-2-[(Z)-pent-2-enyl]cyclopentyl]acetateC13H20O3 Ijerph 17 06506 i03539924-52-2302.9 °C224.296I
Methyl salicylate Benzoate ester methyl 2-hydroxybenzoate C8H8O3 Ijerph 17 06506 i036 119-36-8 222.0 °C 152.147 I ++++
TMTTHomoterpene hydrocarbons (3E,7Z)-4,8,12-trimethyltrideca-1,3,7,11-tetraene C16H26 Ijerph 17 06506 i03762235-06-7293.2 °C 218.38 I +/++
α-ThujeneMonoterpene hydrocarbons 2-methyl-5-propan-2-ylbicyclo[3.1.0]hex-2-en C10H16 Ijerph 17 06506 i038 2867-05-2 152 °C 136.23 C
β-FarneseneSesquiterpene hydrocarbons 7,11-dimethyl-3-methylidenedodeca-1,6,10-triene C15H24 Ijerph 17 06506 i039 18794-84-8 279.6 °C 204.35 I ++
1 I/C = Inducible/constitutive: forest VOC types. 2 C/D = Conifers/deciduous: tree types. 3 E = Emissions [3,8,42]: highly abundant emissions: ****; abundant emissions: ***; moderately abundant emissions: **; common emissions: *. 4 P = Persistence [3,8,42]: less than 10 min: +; 10–60 min: ++; 1 h–24 h: +++; more than 24 h: ++++.
Table 3. Average content of D-limonene in some tree-derived essential oils [106].
Table 3. Average content of D-limonene in some tree-derived essential oils [106].
Plant SourceBotanical FamilyPart of the Plant UsedContent (%) *
Boswellia rivae Engl.BurseraceaeOleoresin28.0–45.0
Boswellia sacra FlueckBurseraceaeOleoresin6.0–21.9
Bursera graveolens (Kunth) Triana et PlanchBurseraceaeOleoresin58.6–63.3
Canarium luzonicum (Blume) A. Gray, C. vulgare Leenh.BurseraceaeOleoresin26.9–65.0
Ravensara aromatica SonneratLauraceaeLeaves13.9–22.5
Abies alba Mill.PinaceaeLeaves and branches28.5–54.7
Abies spectabilis (D. Don) SpachPinaceaeLeaves and branches29.6
Pinus mugo TurraPinaceaeLeaves and branches6.1–37.1
Citrus spp.RutaceaeFruit peel27.0–95.0
* Content (%) = the relative quantity, expressed as a range of percentages, of D-limonene included in tree-derived essential oils.
Table 4. Summary of findings of clinical studies and preclinical experiments with animal models about anti-proliferative effects of D-limonene.
Table 4. Summary of findings of clinical studies and preclinical experiments with animal models about anti-proliferative effects of D-limonene.
TypeDosesResultsRef.
Animal model—mice25 mg orally
administered
Limonene reduces mice stomach/lung tumors by 3%[112]
Animal model—rats7.5–10% dietBreast cancer regression in 89% of animals[113]
Phase I—humans0.5–12 g/m2Partial response in breast cancer patients
Absence of progression in subjects with colorectal cancer
[114]
Phase II—humans8 g/m2No responses in patients with solid tumors[114]
Open-label—humans2 g/die22% reduction in cyclin D1 in early-stage breast cancer[115]
Table 5. Preclinical evidence of limonene biological activity.
Table 5. Preclinical evidence of limonene biological activity.
Functional ResponseModelMolecules/Mechanisms Involved in Targeted PathwaysRef.
Anti-inflammatory
Antioxidant
Murine raw 264.7 cell lineTNF-α, IL-1, IL-6[119]
Human chondrocytesNF-kB, NO, iNOS, p38, JNK[70]
Human lens epithelial cellsROS, CASP, MAPK, Bcl-2/Bax[120]
Human neuroblastoma cellsLC3, clonogenic capacity[121]
Fruit flyNO[84]
BALB/c miceNO[122]
BALB/c miceCatalase, peroxidase[123]
BALB/c miceIL-5, IL-13, MCP-1, TGF-β[124]
BALB/c miceNF-kB, p38, JNK, ERK[125]
Wistar ratsNF-kB, COX-2, iNOS[126]
Swiss miceIL-1β[127]
Sprague–Dawley ratsCOX2, ERK, iNOS, MMP-2, MMP-9, PGE, TGF-β[128]
BALB/c miceApoptosis-related genes[129]
Swiss miceOxidative stress[130]
Anxiolytic
Antidepressant
Sedative
ICR miceElevated plus maze test[90]
MiceSleeping time[88]
Swiss miceMBT assay, anxiolytic effect[131]
RatsLocomotion, dopamine level[132]
RatsImmobility in forced swim test[133]
CUMS miceBody weight, sucrose preference, mobility[134]
Mus musculus albino miceElevated Plus Maze (EPM) test[135]
AnalgesicSwiss miceInduced nociception[136]
ICR miceWrithing test[90]
RatsMechanical hyperalgesia[133]
Swiss miceWrithing test[127]
Swiss miceMechanical hyperalgesia, IL-1β, TNF-α[137]
Abbreviations: CASP, caspase; COX-2, cyclooxygenase-2; ERK, extracellular signal-regulated kinase; ICR, imprinted control region; IL, interleukin; iNOS, inducible NO synthase; JNK, c-jun N-terminal kinase; LC3, microtubule-associated protein light chain 3; MAPK, mitogen-activated protein kinase; MBT, marble burying test; MCP, monocyte chemoattractant protein; NF-kB, nuclear factor-B; NO, nitric oxide; p38, protein 38; PGE, prostaglandin E; ROS, reactive oxygen species; TGF, tumor growth factor; TNF, tumor necrosis factor.
Table 6. Tree species whose total essential oil contains more than 20% of α-pinene [106].
Table 6. Tree species whose total essential oil contains more than 20% of α-pinene [106].
Plant SourceBotanical FamilyPart of the Plant UsedContent (%) *
Boswellia frereana BirdwoodBurseraceaeOleoresin41.7–80.0
Boswellia sacra FlueckBurseraceaeOleoresin10.3–51.3
Cupressus sempervirens L.CupressaceaeLeaves20.4–52.7
Juniperus communis L.CupressaceaeFruit24.1–55.4
Juniperus phoenicea L.CupressaceaeLeaves and branches41.8–53.5
Dryobalanops aromatica GaertnDipterocarpaceaeWood54.3
Abies alba Mill.PinaceaeCones18.0–31.7
Larix laricina Du RoiPinaceaeLeaves and branches38.5
Picea abies (Mill.) BrittonPinaceaeLeaves14.2–21.5
Pinus divaricata AitonPinaceaeLeaves and branches23.1–32.1
Pinus mugo TurraPinaceaeLeaves4.1–31.5
Pinus nigra J. F. X ArnoldPinaceaeLeaves11.5–35.1
Pinus resinosa Ait.PinaceaeLeaves and branches47.7–52.8
Pinus strobus LPinaceaeLeaves30.8–36.8
Pinus sylvestris LPinaceaeLeaves20.3–45.8
* Content (%) = the relative quantity, expressed as a range of percentages, of α-pinene included in tree-derived essential oils.
Table 7. Tree species whose total essential oil contains more than 20% of β-pinene [106].
Table 7. Tree species whose total essential oil contains more than 20% of β-pinene [106].
Plant SourceBotanical FamilyPart of the Plant UsedContent (%) *
Abies alba Mill.PinaceaeCones3.0–22.5
Abies balsamea L.PinaceaeLeaves and twigs28.1–56.1
Picea abies (Mill.) BrittonPinaceaeLeaves and twigs4.8–31.9
Picea glauca (Moench) VossPinaceaeLeaves and twigs23.0
Pinus mugo TurraPinaceaeLeaves and twigs1.3–20.7
Pinus ponderosa Douglas ex P. Lawson & C. LawsonPinaceaeLeaves and twigs28.9
Pinus resinosa Ait.PinaceaeLeaves and twigs29.4–29.9
Pinus strobus L.PinaceaeLeaves and twigs31.1–33.3
Pinus sylvestris L.PinaceaeLeaves and twigs1.9–33.3
Tsuga canadensis (L.) CarrierePinaceaeLeaves and twigs20.8
Citrus x aurantifolia Christm.RutaceaeFruit peel21.1
* Content (%) = the relative quantity, expressed as a range of percentages, of β-pinene included in tree-derived essential oils.
Table 8. Preclinical evidence of α-pinene biological activity.
Table 8. Preclinical evidence of α-pinene biological activity.
ModelTargetReference
Anti-inflammatory
Antioxidant
Murine macrophagesNF-kB,
ERK, JNK
[152]
Human U373-MG cellsROS, peroxidase[80]
Human chondrocytesNF-kB,
IL-1β, JNK, iNOS, MMP-1, MMP-13
[153]
Human lymphocytesTotal antioxidant capacity[154]
MouseIg-E, IL-4[149]
Wistar ratsSuperoxide dismutase, catalase, peroxidase, NO, IL-6[155]
C57BL/6 miceCD4, CD8 and NK cells[102]
Wistar ratsCOX2[156]
Anxiolytic
Antidepressant
Sedative
Sprague–Dawley ratsSleep rhythm[157]
MiceBDNF, TH, EPM test[158]
ICR and C57BL/6N miceGABA BZD receptor, sleep behavior[159]
Wistar–Kyoto miceForced swim test, oxidative phosphorylation expression[160]
Wistar ratsSensorimotor severity score[155]
C57BL/6 miceSchizophrenia-like behavior[161]
AnalgesicBALB/c miceTail-flick test[162]
MiceNeuropathic pain[163]
Wistar ratsNociception[156]
Abbreviations: BDNF, brain-derived neurotrophic factor; BZD, benzodiazepine; EPM, elevated plus maze; ERK, extracellular signal-regulated kinase; GABA, γ-aminobutyric acid; Ig-E, immunoglobulin E; JNK, c-jun N-terminal kinase; MMP-1, metalloproteinase 1; MMP-13, metalloproteinase 13; NF-kB, nuclear factor-B; ROS, reactive oxygen species; TH, tyrosine hydroxylase.

Share and Cite

MDPI and ACS Style

Antonelli, M.; Donelli, D.; Barbieri, G.; Valussi, M.; Maggini, V.; Firenzuoli, F. Forest Volatile Organic Compounds and Their Effects on Human Health: A State-of-the-Art Review. Int. J. Environ. Res. Public Health 2020, 17, 6506. https://0-doi-org.brum.beds.ac.uk/10.3390/ijerph17186506

AMA Style

Antonelli M, Donelli D, Barbieri G, Valussi M, Maggini V, Firenzuoli F. Forest Volatile Organic Compounds and Their Effects on Human Health: A State-of-the-Art Review. International Journal of Environmental Research and Public Health. 2020; 17(18):6506. https://0-doi-org.brum.beds.ac.uk/10.3390/ijerph17186506

Chicago/Turabian Style

Antonelli, Michele, Davide Donelli, Grazia Barbieri, Marco Valussi, Valentina Maggini, and Fabio Firenzuoli. 2020. "Forest Volatile Organic Compounds and Their Effects on Human Health: A State-of-the-Art Review" International Journal of Environmental Research and Public Health 17, no. 18: 6506. https://0-doi-org.brum.beds.ac.uk/10.3390/ijerph17186506

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop