bs_bs_banner

Review

Journal of Pharmacy And Pharmacology

Selected terpenoids from medicinal plants modulate endoplasmic reticulum stress in metabolic disorders Natasha Beukes, Ruby-Ann Levendal and Carminita L. Frost Department of Biochemistry and Microbiology, Nelson Mandela Metropolitan University, Port Elizabeth, South Africa

Keywords apoptosis; phytochemicals; signalling pathways; unfolded protein response Correspondence Carminita L. Frost, Department of Biochemistry and Microbiology, Nelson Mandela Metropolitan University, PO Box 77000, 6031 Port Elizabeth, South Africa. E-mail: [email protected] Received November 25, 2013 Accepted March 16, 2014 doi: 10.1111/jphp.12267 Chemical compounds: Artemisinin (PubChem CID: 68827); Cannabidiol (PubChem CID: 26346); Cannabinol (PubChem CID: 2543); Dehydrocostuslactone (PubChem CID: 73174); Marrubiin (PubChem CID: 73401); Thapsigarin (PubChem CID:446378); Δ9-tetrahydrocannabinol (PubChem CID: 2977)

Abstract Objectives The majority of research performed on cellular stress and apoptosis focuses on mitochondrial dysfunction; however, the importance of the endoplasmic reticulum dysfunction and the link to metabolic diseases has gained a substantial interest. This review focuses on the potential of terpenoids to influence endoplasmic reticulum stress and the possible role terpenoids play as the treatment of metabolic diseases. Key findings Metabolic diseases develop as a result of a cascade of cellular pathways. In most cases, cells are able to compensate for the disruption of the cellular homeostasis although the initiation of response pathways; however, chronic stress initiates apoptotic pathways. This reviewed (1) showed the importance of phytoterpenoids to influence endoplasmic reticulum (ER) stress and homeostasis, (2) showed how regulating ER stress affect the cell survival and death, and (3) highlighted some examples of how the progression of metabolic diseases can be influenced by ER. Summary Due to the substantial number of terpenoids that have been identified in literature, this review gave examples of 21 terpenoids that have been documented to have an effect on the different proteins associated with ER stress, how these plant terpenoids influence ER dysfunction and metabolic diseases such as diabetes, cancer, liver, and neurological diseases and parasitic infections.

Current findings of the role of medicinal plants in the traditional treatment of metabolic diseases An extensive percentage of the world’s developing countries depend on the use of traditional medicines as an alternative to Western medicines. The use of plants and plant-derived remedies to treat a variety of different ailments have been used for centuries within different cultures globally. Plantderived traditional remedies are used to treat a wide variety of ailments, ranging from mild dermatological problems to serious metabolic conditions, such as diabetes, cardiovascular disease and cancer.[1–6] Research in the fields of ethnopharmacology is growing exponentially with the discovery of numerous novel treatments derived from natural plant products. Over the last few decades, a substantial amount of research has focused on identifying the different medicinal plants and the biologically active compounds (e.g. terpenoids, flavonoids,

alkaloids, glycosides, saponins) that have the potential to be an alternative therapeutic treatment for a variety of different ailments. This review will focus on terpenoids and their potential use in alleviating endoplasmic reticulum (ER) stress found in different metabolic disorders.

The endoplasmic reticulum and its importance The ER is an essential cellular organelle responsible for the production, folding, post-transcriptional modifications of nascent proteins, maintaining calcium homeostasis, and the degradation of secretory and transmembrane proteins via the ER-associated degradation (ERAD) pathway.[7,8] For example, the ER degradation enhancing

© 2014 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 66, pp. 1505–1525

1505

Terpenoids and ER stress

Natasha Beukes et al.

α-mannosidase-like protein 1 (EDEM-1) family, expression of genes involved in DNA repair, genes involved in the maintenance and expansion of the ER, proteins involved in lipid biosynthesis and maintaining the redox potential of the cell.[9–11] By definition, ER homeostasis is the balance between the ER’s protein loading and its protein folding capacity. Any disturbance in the ER’s homeostasis would lead to the production and accumulation of unfolded and incorrectly folded proteins. It is the accumulation of these proteins within the ER that is responsible for the ER stress and the initiation of the unfolded protein response (UPR).[12]

Terpenoids linked to endoplasmic reticulum stress Terpenoids can be modified to produce thousands of different intermediates and metabolites, to date approximately 40 000 plant and animal terpenoids have been identified; however, the majority have not been extensively studied.[13,14] All terpenoids are derived from one of two isomers, isopentyl diphosphate and dimethylallyl diphosphate via the traditional mevolonate or methylerythritol phosphate pathway. However, there have been documented cases where plants use both pathways in the synthesis of terpenoids.[15–17] Because of the vast number of terpenoids that have been identified, a small fraction has been studied as possible treatments for a variety of ailments. This review has selected a few terpenoids that have shown promise as possible treatment options in metabolic diseases. These include: abscisic acid,[18,19] lycopene,[20,21] carotene,[22] cannabidiol (CBD),[23–25] cannabinol (CBN),[26] tetrahydrocannabinol (THC),[27] geraniol,[28] limonene,[29] genipin,[30] linalool,[28,29] menthol,[31] perillyl alcohol,[32] kujigamberol,[33] marrubiin,[34,35] sarcodonin,[36,37] ganoderiol F,[38] artemisinin,[39–41] dehydrocostus lactone,[42–44] farnesol,[28,45] parthenolide[46] and thapsigargin[47] (Table 1).

Endoplasmic reticulum stress ER stress was first described by Kozutsumi and associates when they observed an increased production of glucoseregulated protein 78 (GRP78; also known as immunoglobulin heavy-chain binding protein (BiP)/heat-shock 70 kDa protein) resulting from the accumulation of unfolded proteins in the ER.[48] To prevent ER stress from developing, cells need to tightly regulate ER homeostasis. ER stress can be induced through the treatment with compounds that inhibit glycosylation (e.g. tunicamycin); disruptions in the cell’s calcium homeostasis; the overexpression of proteins, accumulation of incorrectly folded/unfolded proteins, star1506

vation due to nutrient depletion, oxidative conditions, hypoxia, accumulation of free fatty acids and, in some cases, viral infections.[7,8,49–52] Once ER homeostasis has been disrupted, ER stress triggers the activation of three transmembrane proteins: inositol requiring enzyme 1 (IRE1), PKR-like ER kinase (PERK) and activating transcription factor 6 (ATF6). These transmembrane proteins become activated with the release of GRP78, which preferentially binds to accumulated unbound or incorrectly folded proteins, allowing for the dimerization of IRE1 and PERK complexes into ‘active’ complexes and allows the translocation of ATF6 to the golgi apparatus.[8] The activation of these proteins and protein complexes, subsequently regulates the signalling cascade of the UPR.[12] The PERK pathway is largely involved in the attenuation of protein translation. The IRE1 and ATF6 signalling pathways are primarily responsible for the transcriptional up-regulation of ER chaperones and initiate the ERAD pathway.[8] Thus, the activation of UPR pathways may lead to the attenuation of protein translation, except ER resident chaperones and the proteins associated with ERAD (e.g. EDEM). The expression of these proteins are up-regulated to decrease the demands of ER function and to degrade any incorrectly folded and unfolded proteins that have accumulated in the ER lumen.[7] In cases where the ER stress remains irresolvable, the cells initiate the process of apoptosis. Apoptosis unlike necrosis is a cellprogrammed death response to a variety of stimuli and is characterized by a variety of cellular changes including chromatin and cytoplasm condensation (resulting in the organelles becoming densely packed), cell shrinkage and pyknosis.[53] The UPR pathways are complex, a ‘cross-talk’ between the UPR signalling cascades and mitochondrial dysfunction is well-documented, and the functional relationship between the ER and mitochondria is essential to maintain cellular homeostasis. If persistent ER stress and the activation of the IRE1 signalling cascade occurs, the initiation of apoptosis may occur either via the phosphorylation of β-cell lymphoma 2 (Bcl-2) and Bcl-2 interacting mediator of cell death (Bim), or the release of cytochrome C, which would initiate the caspase signalling pathway (Figure 1). There have also been documented cases indicating that mitochondrial dysfunction increases the level of ER stress as seen by the up-regulation of phosphorylated eukaryote translation initiation factor 2 alpha (eIF2α), GRP78, C/EBP homologous protein (CHOP) and the activation of c-Jun N-terminal kinase (JNK) signalling.[66] The UPR (Figure 1) can be regulated by a number of proteins, the most common include: BiP/GRP78, growth arrest and DNA damage-inducible protein 34 (GADD34), 58 kDa inhibitor of protein kinase (P58IPK),

© 2014 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 66, pp. 1505–1525

Natasha Beukes et al.

Terpenoids and ER stress

Table 1 A summary of the chemical structures of the few terpenoids mentioned in this review that have shown promise in alleviating metabolic diseases Structure and classification of different terpenoids

Name

Plant source

Reference(s)

Abscisic acid

Rosa arvensis, Bibes nigrum, Fagus sylvatica, Malus domestica

[18,19]

Lycopene

Solanum lycopersicum, Carica papaya L. Citrullus lanatus

[20,21]

Carotene

Daucus carota L., Spirulina platensis

[22]

Cannabidiol (CBD)

Cannabis sativa

[23–25]

Cannabinol (CBN)

Cannabis sativa

[26]

Δ9-tetrahydrocannabinol (THC)

Cannabis sativa

[27]

Geraniol

Citrus spp., Lavandula angustifolia Mill, Cymbopogon winterianus

[28]

Limonene

Citrus spp.

[29]

Genipin – also classified as an iridoils

Gardenia fructus

[30]

Carotenoids CH3 CH3 O

CH3

OH O

CH3

OH

CH3 H3C

CH3

CH3

CH3 CH3

CH3

CH3

CH3

H3C CH3

CH3

CH3

H3C

CH3

CH3

CH3

CH3

H3C CH3

Cannabinoids CH3 OH H2C

HO CH3

CH3

CH3 OH

H3C H3C

O

CH3

CH3 OH

H3C H3C

O

CH3

Monoterpenoids CH3 HO CH3 CH3

H2C CH3 H3C

O

HO

O HO

O

CH3

© 2014 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 66, pp. 1505–1525

1507

Terpenoids and ER stress

Table 1

Natasha Beukes et al.

Continued

Structure and classification of different terpenoids CH3

H3C

OH CH2

H3C

Name

Plant source

Reference(s)

Linalool

Lavandula angustifolia, Ocimum sanctum, Bursera aloexylon

[28,29]

Menthol

Lavandula angustifolia, Ocimum sanctum, Bursera aloexylon

[31]

Perillyl alcohol

Prunus spp., Lavandula angustifolia

[32]

Kujigamberol

Kuji amber

[33]

Marrubiin

Leonotis leonurus L. (Lamiaceae), Marrubium vulgare L. (Lamiaceae)

[34,35]

Sarcodonin

Sarcodon scabrosus, Sarcodon Leucopus, Hydnellum suaveolens, Hydnellum geogerirum, Phellodon niger

[36,37]

Ganoderiol F

Ganoderma amboinense L., (Lamiaceae)

[38]

OH CH3 H3C CH3

CH2 CH3

HO

Diterpenoids CH3

H3C

CH3

HO

CH3

O

HO H3C

H3C

CH3

O O

H3C H3C

OO

O O

– OH CH O 3 N+

O O O H3C

OH OH

O

CH3 N OH

O CH3

H3C

Triterpenoids OH H3C CH3

H3C

CH3

H3C H3C

OH

O

1508

© 2014 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 66, pp. 1505–1525

Natasha Beukes et al.

Table 1

Terpenoids and ER stress

Continued

Structure and classification of different terpenoids

Name

Plant source

Reference(s)

Artemisinin

Artemisia annua, Artemisia indica

[39–41]

Dehydrocostus lactone – also classified as a lactone

Saussurea lappa C.B. Clarke (Asteraceae), Laurusnobilis L., Magnolia sieboldii L., Aucklandia lappa

[42–44]

Farnesol also classified as a fatty alcohol

Matricaria recutita L., Zea mays

[28,45]

Parthenolide

Tanacetum parthenium

[46]

Thapsigargin also classified as a guaiane

Thapsia garganica

[47]

Sesquiterpenoids CH3 O

H3C O

O

CH3 O

H2C CH2

O O H2C

CH3

CH3

CH3

HO O

CH3

O

O H3C

H2C CH3 H3C HO

CH3 O O

CH3

H3C O

O CH3O

O

HO HO H3C O

O

CH3

O

H3C

and wolfram syndrome 1 (WFS1/wolframin). GRP78 is an ER chaperone and is considered a master UPR regulator. This protein is responsible for the prevention of premature UPR activation. GADD34 is responsible for the dephosphorylation of eIF2α, through the recruitment of phosphatsase 1, restoring the eIF2α to its unphosphorylated state. P58IPK is considered a negative feedback regulator of PERK signalling. Similarly to GRP78, this protein’s main function is to maintain ER homeostasis. The phosphorylation of eIF2α, results in the attenuation of general protein translation. The expression of P58IPK is mediated by ATF6 in the pancreas and interacts with the kinase domain of PERK, inhibiting the kinase activity. WFS1 is an important and key negative regulator of UPR. If a mutation in this protein’s gene occurs, people can develop Wolfram syndrome. WFS1 is located in the ER and appears to have a protective function against ER stress.[12] Very little is known about the protein’s function; thus, the protein’s functional mechanism is unclear. However, Oslowski and Urano[12] have recently reported that WFS1 may play a role in regulating ATF6α activation.

Relationship between the unfolded protein response and terpenoids Inositol requiring enzyme 1 or endoplasmic reticulum to nucleus signalling 1 pathway, and the associated terpenoids The activation of the type I transmembrane protein, IRE1 and the subsequently signalling pathway is considered to be one of the main UPR pathways. IRE1 is triggered as early response to the accumulation of incorrectly folded proteins in the ER under both pathological and physiological conditions.[67] When the ER is under a tolerable amount of stress, IRE1 undergoes dimerization, and autophosphorylation after GRP78 has been released, resulting in the activation of the IRE1 RNAse domain. However, it has been suggested that the IRE1 signalling pathway may remain partially active in cells at a basal level.[68] The IRE1 signalling pathways have a dual cellular function, as this signalling pathway has both pro-apoptotic branch via the activation of tumour necrosis factor receptor-associated factor 2 (TRAF2), apoptosis signal regulating kinase 1 (ASK1), JNK signalling pathway, and a pro-survival branch via the activation of X-box

© 2014 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 66, pp. 1505–1525

1509

Terpenoids and ER stress

Natasha Beukes et al.

ER Ca2+

Grp 78 Bound

IP 3 receptor IRE1

PCP12 - TRAF2

PERK kinase

IRE1

PERK kinase

ATF 6 (p90)

2+

Ca

Mitochondria

Cyt C

JIK–recruited to IRE1 prior TRAF2 binding

XBP1

BAX BAK Cyt C

PCP12

CP12

EIF 2α P

TRAF2

EIF 2α

GADD 34

Apaptosome CP9

ASK1

P Bcl-2

Reduces I-κB

NF-κB XBP 1s

P

Bim

P38 MAPK

CP3

Golgi Apparatus

Calpain

ATF 6α ATF 4

JNK Bcl-2 Bim

SAPK JUN

P

APOPTOSIS

General CHOP pathway

CHOP, ER chaperones, CHOP, Cytokines, DnaJ, CHOP, Cytokines, ERO1β, ERAD ERAD proteins. TRB3, GADD34, AA proteins, ORP150. transport & Nucleus metabolism.

Figure 1 A schematic representation of the main unfolded protein response pathways and the apoptotic ‘cross-talk’ between the endoplasmic reticulum stress and mitochondrial dysfunction. Apoptosis can be induced through different pathways that are initiated by the activation of the different unfolded protein response transmembrane proteins: insitol requiring enzyme 1 (green pathway), PKR-like endoplasmic reticulum kinase (red pathway) and activating transcription factor 6 (blue pathway). A ‘cross-talk’ between the different pathways may occur; thus, apoptosis can occur due to both endoplasmic reticulum (unfolded protein response pathways) and mitochondrial dysfunction (yellow pathway). However, the activation of caspase 9 and the subsequent caspase-induced apoptosis can occur independently from mitochondrial dysfunction.[8,54–65]

binding protein (XBP1).[69,70] Activation of IRE1 occurs when the cell is experiencing severe hypoxia or if there is an increase in the expression of interleukins (IL-1β, IL-6 and IL-8), vascular endothelial growth factor (VEGF) A, connective tissue growth factor and matrix proteins, such as thrombospondin 1 and metallopeptidase 9.[71–73] Both branches of the IRE1 pathway has been documented to be modulated by several interacting proteins including β-cell lymphoma 2 (Bcl-2), Bcl-2-associated X protein (BAX) and Bcl-2 antagonist/killer protein (Figure 1). Protein tyrosine phosphatase 1 B (PTP1B) has also been documented to affect the IRE1 pathway. PTP1B has been observed to disrupt the splicing of XBP1, subsequently attenuating the up-regulation of XBP1’s target genes (e.g. EDEM) and induce the phosphorylation of JNK.[74] 1510

The UPR signalling pathway the expression and regulation of different genes and proteins that are associated with cell survival and apoptosis. However, with the apoptotic signalling pathway, there is ‘cross-talk between the ER and the mitochondia protein signalling pathway (Figure 1).[8,54–64] IRE1 has been documented to have two catalytic domains. The first has a serine/threonine transautophosphorylation activity and the second, an endoribonuclease activity, which is responsible for initiating the splicing of XBP1.[75,76] The cleavage of XBP1 messenger RNA (mRNA) in an unconventional splicing event, forcing a translational frame producing the active transcription factor XBP1.[8] The active form of XBP1 is responsible for regulation and expression of a variety of cellular chaperones and ERAD enzymes. The active transcription factor (XBP1s) is integral

© 2014 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 66, pp. 1505–1525

Natasha Beukes et al.

Table 2

Terpenoids and ER stress

Examples of terpenoids that have an effect on various parts of the IRE1 signalling pathway

Terpenoid

Protein target

Cannabidiol (CBD) Dehydrocostuslactone (DHE)

XBP1

Cannabidiol (CBD)

ASK1

Dehydrocostuslactone (DHE)

p38

Ganoderiol F

MAPK

Cannabidiol (CBD) Dehydrocostuslactone (DHE)

JNK

Potential mode of action/ pathway

Reference(s)

CBD and DHE activated the endoribonuclease domain of IRE1 and induced the splicing of XBP1 mRNA in the hepatic stellate cells (HSCs) and non-small lung cancer cells A549 and NCI-460. The cannabidiol induced the dephosphorylation of ASK1 triggering the apoptotic signalling cascade in mouse HSC, rat HSC and LX-2 cells (immobilized human HSC line derived from a cirrhotic patient). Induces the accumulation of calcium in the cytosol, possibly via the PPARγ and the calcium/calmodulin dependent kinase II signalling pathway and induces the activation and accumulation of p38 in the non-small lung cancer cells A549 and NCI-460. Ganoderiol F activates the mitogen-activated protein kinase (MAPK) and up-regulates the expression of cyclin-dependent kinase inhibitor p16, resulting in HepG2 cells senescences CBD and DHE induced the accumulation of phophorylated JNK as a result of the activated ASK1 in non-small lung cancer cells A549 and NCI-460.

[25,44]

[25]

[44,78]

[38]

[25,44]

ASK1, apoptosis signal regulating kinase 1; JNK, c-Jun N-terminal kinase; MAPK, mitogen-activated protein kinase; PPARγ, peroxisome proliferationactivated receptor; XBP1, X-box binding protein.

to the anti-apoptotic signalling branch of the IRE1 signalling pathway. The active XBP1 is responsible for regulation the expression of proteins implemented in ensuring correct protein folding, trafficking and secretion; active XBP1 has also been linked to the restoration of ER homeostasis and general cell survival.[69,77] IRE1 has been documented to be responsible for a number of cellular functions, including the reduction of the ER workload, although the cleavage of ER associated mRNA and enhancing the synthesis of pro-insulin; thus, IRE1 has an important role in the production of insulin. However, under intolerable ER stress, IRE1 can activate an apoptotic pathway (Figure 1). This occurs when IRE1 recruits the protein TRAF2 and phosphorylates JNK, which is responsible for the regulation of the Bcl-2 family of proteins that induce apoptosis.[12] TRAF2 is an adaptor protein that binds to JIK : IRE1 complex and promotes the activation of JNK indirectly through the ASK1.[70] Although the IRE1 signalling pathway and the link between mitochondrial dysfunction and apoptosis via the activation of caspases is well-established; a limited number of studies on the effect terpenoids have on the IRE1 signalling cascade have been documented (Table 2).[25,38,44,78] Double-stranded RNA-activated protein kinase-like endoplasmic reticulum kinase pathway and the associated terpenoids The second most characterized ER type I transmembrane protein is PERK (also known as pancreatic eukaryotic ini-

tiation factor 2α kinase; EIF2AK3).[54,67] This protein complex plays an important role in the functioning of β-cells under normal physiological conditions. PERK aids in the regulation of β-cell development and proliferation, and the production of pro-insulin in response to hyperglycaemic conditions. Under conditions that induce ER stress, PERK, similarly to IRE1, dimerizes and autophosphorylates once GRP78 has been released, triggering the phosphorylation of eIF2α, which is a subunit of the guanosine triphosphate-binding complex eIF2,[79] attenuating general mRNA translation under tolerable ER stress conditions. Under normal conditions, protein translation is initiated once eIF2 recruits the methionyl-tRNA to the ribosome. Once PERK is activated and eIF2α is phosphorylated, the activity of eIF2 complex is disrupted, and general protein translation ceases, reducing the protein load in the ER.[80] The phosphorylation of the alpha subunit of eIF2 favours the translation of ATF4 mRNA, which subsequently produces the active transcription factor and nephrin. However, it must be noted that the phosphorylation of eIF2α is not always synonymous with ER stress, and as such, ER dysfunction should be confirmed with additional experiments other than the detection of phosphorylated eIF2α.[80] Under irresolvable ER stress conditions, the continuous phosphorylation of eIF2α, induction of ATF4 and ATF3 would induce cell death through the up-regulation of the pro-apoptotic factor CHOP (Figure 1).[12] Recently, Oslowski and Urano[12] discover a novel transcription factor that appears to antagonize apoptosis and through the transcriptional regulation of protein kinase B (AKT1) mediates β-cell survival.

© 2014 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 66, pp. 1505–1525

1511

Terpenoids and ER stress

Table 3

Natasha Beukes et al.

Examples of some terpenoids that affect the cellular proteins specific to the PERK pathway

Terpenoid

Protein target

Cannabidiol (CBD) Δ9-tetrahydrocannabinol (THC)

eIF2α

Cannabidiol (CBD)

ATF4

Potential mode of action/pathway

Reference(s)

CBD and THC allowed the trascription factor to be phosphorylated via the activation of the PERK pathway in the hepatic stellate cell lines LX-2 and U86MG. CBD induced of the PERK pathway resulted in ATF4 accumulating in the nucleus of mouse HSC LX-2 cells.

[25,94]

[25]

ATF4, activating transcription factor 4; eIF2α, eukaryote translation initiation factor 2 alpha; HSC, hepatic stellate cell; PERK, PKR-like endoplasmic reticulum kinase.

ATF4 regulates the expression of the genes responsible for the restoration of ER homeostasis (e.g. GADD34), the production and metabolism of amino acids, regulating the antioxidant stress response, the transcription of other prosurvival genes (GRP78, GRP94), Tribbles homologue 3 (TRB3), the receptor activator nuclear factor-kappa B (NF-κB) ligand VEGF as well as the expression of CHOP (also known as DNA-damage inducible transcript/DNAdamage inducible gene 153).[7,50,81–83] Increased expression of GADD34 increases the rate eIF2α dephosphorylation in a negative feedback loop involving protein phosphatase 1, allowing for protein translation to be resumed.[84,85] Increased expression of CHOP can be induced through the initiation of all three UPR transmembrane sensors, e.g. IRE1, PERK and ATF6; therefore, it has been traditionally considered an apoptotic inducer. CHOP has been documented to activate the ER oxidase, GADD34, TRB3, and the proapoptotic factors Bim and death receptor 5 while decreasing the expression of the prosurvival protein Bcl-2.[83,86–91] This is not always the case in all cell types and some neurological diseases, e.g. murine models with Pelizaeus–Merzbacher disease.[92] In addition to the activation of the eIF2 signalling cascade, PERK has also been documented to phosphorylate the nuclear respiratory factor 2, which functions by binding to the antioxidant response element, resulting in the transcriptional activation of the gene-encoding detoxification enzymes.[93] Unlike the IRE1 signalling pathway, the PERK pathway is more simplistic because this pathway is not considered the initial UPR pathway, and a limited amount research has been performed on the effects of terpenoids. In some cases, terpenoids may have an effect on more than one protein in the signal pathway, for example CBD (Table 3).[25,94] Activating transcription factor 6 pathway and the associated terpenoids The third UPR signalling pathway primarily involves the activation and cleavage of ATF6. Unlike IRE1 and PERK, ATF6 is classified as a type II transmembrane protein.[67] 1512

Under ER stress conditions, GRP78 dissociates from the luminal domain of ATF6, releasing the protein into the cytoplasm and facilitating the translocation of ATF6 to the Golgi apparatus via the coat protein II vesicles. At the Golgi apparatus, the transcription factor is cleaved into its active form by site-1 and site-2 proteases.[95,96] The active transcription factor, which is the cytoplasmic bZIP domain, is subsequently translocated to the nucleus where it up-regulates the expression of homeostatic factors such as chaperones (e.g. GRP78 and calnexin – allowing for incorrectly folded proteins to be refolded), as well as proteins associated with ERAD and lipid biosynthesis[7,12,95,97–101] (Figure 1). In general the detection of the cleaved ATF6 is considered a good biomarker of ER stress; however, it may be difficult to detect.[80] As far as the authors of this review are aware, the only terpenoid that has been documented to affect ATF6α is CBD, which induces the cleavage of the transcription factor in the nucleus.[25] Aside from the proteins already mentioned, which are specific to the different UPR pathways, there are several proteins that are associated with more than one of the UPR pathways or other cellular pathways that may be associated with ER stress (Table 4).[25,30,33,44,102–109]

Is there a role for terpenoids from medicinal plants in alleviating endoplasmic reticulum stress and metabolic disorders? Literature has indicated that the core programs that contribute to the proper executive phase of apoptosis appear to be expressed constitutively in virtually every cell. The intracellular signals (e.g. UPR, mitochondrial dysfunction, mammalian target of rapamycin, NF-κB) that are involved in the induction of apoptosis are often regulators of other cellular responses. However, there is accumulating evidence that multiple signal pathways can interact during a process called ‘cross-talk’. A cell’s response to a given stimulus may alter significantly not only between cellular compartments but also within a cell populations.[110] Because of the ‘crosstalk’ that occurs between signalling pathways both intrinsic

© 2014 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 66, pp. 1505–1525

Natasha Beukes et al.

Table 4

Terpenoids and ER stress

Terpenoids that have been shown to affect some of the proteins associated with more than one unfolded protein response pathway

Terpenoid

Protein targeta

Potential mode of action

Reference(s)

Dehydrocostuslactonev (DHE) Genipin

Bip/Grp78

[30,44,102–108]

Cannabidiol (CBD) Parthenolide

Calnexin NF-κB

Kujigamberol

GSK3-β

Cannabidiol (CBD) Dehydrocostuslactone (DHE)

CHOP

DHE increased the expression of the chaperone in lung cancer cell lines A549 and NCI-H460. Genipin suppresses the up-regulation of Bip in Neuro2a cells after the cells were treated with the calcium ionophore A23187. Genipin has also been documented to have neuroprotective properties in the rat primary hippocampal neurons when treated with β-amyloid peptide. In Neuro2a cells, genipin has shown to have neuroprotective effects against the cytotoxicity reaction induced by serum deprivation and oxidative stress. CBD up-regulates the expression of calnexin in mouse HSC LX-2 cells. Parthenolide modifies the p65 subunit of the nuclear factor by alkylation on the cysteine residues, inhibiting its activity in Jurkat T cells. Kujigamberol has been noted to inhibit GSK3-β, that is believed to restore growth in the mutant yeast strain YNS17 by affecting the calcium signal transduction. Kujigamberol is also cytotoxic to the promyelocytic leukemia cell line HL60. Both terpenoids up-regulates the expression of the pro-apoptotic protein CHOP via the phosphorylation of PERK and the activation of the PERK signalling cascade in the hepatic stellate cells LX-2 and A549 cell lines and lung cancer cell line, NCI-H460, and the multiple myeloma cells OPM2. MM.1S.

[25] [109]

[33]

[25,44]

Bip, immunoglobulin heavy-chain binding protein; CHOP, C/EBP homologous protein; Grp78, glucose-regulated protein 78; GSK3-β, glycogen synthase kinase 3 beta; HSC, hepatic stellate cell; NF-κB, nuclear factor-kappaB.

and extrinsic, the effect different terpenoids have on the specific signalling pathways may also have an effect-related pathway that may not necessarily be considered in the original experiments. The progression of metabolic diseases and, in some cases, viral infections is complex and involves the activation of different signalling pathways. This review focused on the role of the ER and the UPR pathways; however, the ‘cross-talk’ that occurs between the UPR signalling pathways and other cellular pathways need to be kept in mind. As in the case of most of the different active components found in plants, there are several different types of terpenoids/isoprenols that can be classified as polyisoprenoid alcohols, phytol, carotenoids and abietic acid, all of which can be derivatized to produce additional terpenoids.[14] Terpenoids have been documented to have several different functions such as the regulation of cation channels, induce apoptosis and suppress tumour proliferation,[13,111–114] which raises the question: Can terpenoids be used to prevent or aid in the alleviation of metabolic diseases? If so, what are the biological target, and do these phytochemicals distinguish between healthy and ‘diseased/infected’ cells? For the treatment of metabolic disorders, a number of possible therapeutic targets have been identified including nuclear receptors and several different proteins involved in the URP signalling cascades. The cellular response to ER stress does not simply involve the activation of UPR pathways to determine whether the

cell survives or triggers apoptosis. Under tolerable ER stress conditions where the IRE1 pathway is triggered, the signalling cascade may initiate the ER overload response as an alternative pathway to activate NF-κB or GSK3-β to trigger anti-apoptotic responses.[115,116] Nuclear receptors are of great interest, as they appear to be the main target for the different active compounds present in plants.[117,118] Nuclear receptors can generally be classified into: (1) steroid hormones such as mineral corticoid, glucocorticoid, androgen, estrogen and progesterone receptors; (2) receptors that have the conserved characteristics of nuclear receptors but are not associated with endogenous ligands; and (3) receptors that to associate with endogenous ligands, such as the peroxisome proliferationactivated receptors (PPARs), thyroid hormone receptor and retinoic acid receptors. In normal cell differentiation, very low levels of NF-κB is required for controlled cell proliferation; however, under conditions where uncontrolled cell proliferation occurs (cancerous/tumour cells), the expression and activity of NF-κB is increased. Thus, the potential to use terpenoids (e.g. pathenolide, artemisinin, resveratol) to specifically target proteins crucial to cell proliferation as well as having the ability to target proteins involved in apoptotic pathways, for example p53 pathway[119] demonstrates the potential terpenoids to influence multiple cellular processes. As a result of some terpenoids, especially the lactone-derived terpenoids are in use in clinical trials for the treatment of cancers. The use of the endocannabinoid

© 2014 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 66, pp. 1505–1525

1513

Terpenoids and ER stress

Natasha Beukes et al.

system to mediate the activation of HSCs and subsequently fibrosis has been documented,[120] and the use of cannabinoids in the treatment of cancer or the alleviation of associated conditions (e.g. pain and inflammation) is well-established.[121–123]

Metabolic diseases and infections that have been linked to endoplasmic reticulum stress and unfolded protein response pathways The importance of the ER and its dysfunctional prevalence is increasingly being observed in different metabolic diseases and infections that may trigger a metabolic response. A variety of diseases are associated with ER stress and dysfunction, and as such, this review looks to provide a few examples of diseases, indicating the different links to ER stress, the respective proteins associated with the UPR signalling pathways and some terpenoids that have been documented to have an effect on the UPR pathways and diseases. As previously mentioned, the use of plants and plantderived compounds are well-established as medicinal treatments for a wide variety of ailments and diseases (as an alternative to synthetic medicines). Various terpenoids have been identified for their potential and promising role in treating diseases[25,30,33,44,102–109] (Table 4). ER dysfunction has been documented to be linked to a variety of different diseases including lung diseases, cancers, diabetes and diabetic-related diseases/complications, malaria, neurological diseases, inflammatory diseases, viral infections etc. This review provides a few examples of diseases that have been linked to ER stress and dysfunction, and identifies terpenoids that have been associated with the different treatments[13,20,21,32,34,35,40,41,55,56,114,124–155] (Table 5).

Diabetes mellitus Diabetes is a common metabolic disorder that is characterized with chronically high glucose levels in the blood because of the inability of the body to utilize glucose as a result of an absence or deficiency in insulin production by β-cells. Diabetes is generally classified as either type 1 diabetes mellitus (T1DM) or type 2 diabetes mellitus (T2DM); however, a rare medical condition, Wolcott–Rallison syndrome, which is an autosomal recessive form of juvenile onset diabetes, has also been documented.[156] Experimental, clinical and genetic evidence indicating that ER stress plays a role in β-cell dysfunction and death during the progression of T1DM, T2DM and genetic forms such as Wolcott–Rallison syndrome is well-documented and as such is briefly mentioned in this review. In T1DM, β-cells are the direct target of the autoimmune invasion that results in insulitis and β-cell death, and as such, T1DM is 1514

classified as an autoimmune form of diabetes. In T1DM, ER stress has been documented to occur resulting from exposure to pro-inflammatory cytokines, increased nitric oxide production, depletion of the ER calcium stores and the activation of the transcription factor ATF3, resulting in the up-regulation of CHOP and subsequently inducing the apoptotic pathways.[157,158] In T2DM, metabolic stress is thought to activate the innate immune system, resulting in an increase in cytokines, islet-associated macrophages, insulin resistance, and the reduction of β-cell mass and ultimately apoptosis.[159] A number of factors contribute to cell apoptosis in T2DM, including glucotoxicity, lipotoxicity, ER stress, oxidative stress and amyloid deposition.[160] The apoptosis of β cells in T2DM may be contributed to a variety of different components, including the activation of the IRE : TRAF2 : JNK signalling pathway, the up-regulation of CHOP ATF4, spliced XBP1 and GSK3-β.[161,162] The first documented case, linking ER stress to diabetes was reported in 1972 by Wolcott and Rallison. They documented the presence of a genetic mutation in the gene EIF2AK3, encoding the catalytic domain of the UPR transmembrane sensor PERK, thus resulting in a defective PERK signalling pathway, that leads to β-cell dysfunction and apoptosis.[163,164] Diabetes has also been linked to a variety of different ailments, for example blindness, cardiovascular diseases, kidney failure and obesity.[165] The occurrence of insulin resistance in T2DM is well-established. Insulin resistance has been linked to the prothrombic state and coagulation. The prothrombic state that is characterized by increased fibrinogen levels, increased plasminogen activator inhibitor and abnormalities in platelet function that would result in thrombosis. Insulin resistance affects the prothrombic state and coagulation by cosegregating with abnormalities that may occur in coagulation.[166] Well-documented terpnoids that have been linked to diabetes and diabetes-related ailments include cannabinoids and abscisic acid (Table 4). The antithrombic effect of Cannabis sativa and the cannabinoids THC and CBN were observed in vivo and in vitro using obese rats and human plasma respectively. Indicating the potential use of cannabinoids in patients where traditional anticoagulants (e.g. heparin and warfarin) cannot be used.[167] In 3T3-L1 cells, THC has been documented to decrease the rate of adipogenesis and increase insulininduced glucose uptake, relative to increasing concentrations of the cannabinoid.[27] The cannabis extract has also been shown to reduce the effects caused by a cafeteriainduced obesity, especially with regards to fat depots, insulin level regulation and cytokine production.[168] Abscisic acid has been documented to up-regulate the expression of PPARγ, and increase glucose intolerance and decreases the level of fasting glucose.

© 2014 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 66, pp. 1505–1525

Metabolic disorder(s)

Malaria Leishmaniasia Cancer

Cancer Malaria

Cancer

Leishmaniasia

Cancer

Cancer

Cancer Dengue fever

Cholesterol Cancer

Artemisinin

Thapsigargin

Sarcodonin

Linalool

Geraniol

Farnesol

© 2014 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 66, pp. 1505–1525

d-Limonene

Menthol Perillyl alcohol

HMG-CoA reductase Mannose-6-phosphate/ insulin-like growth factor II receptor

HMG-CoA

PPARγ PPARα

PPARγ

NOS Mitochondria Kinetoplast chromatin

BAX, Bcl-2 Caspases

Inhibits the HMG-CoA reductase activity and reduce cholesterol in rodent models. Has a chemopreventive/therapeutic properties against, liver, mammary, cancer in Fisher 344 rats and human MIA PaCa-2 cells. Perillyl alcohol inhibited tumour growth in Fisher 344 rats by increasing the expression of mRNA encoding mannose-6-phosphate/insulin-like growth factor II receptor and transforming growth factor B types I, II and III receptors.

Antitumour activity in a variety of rodent organs (mammary, liver, lungs, stomach and skin). Often serves as a precursor to the synthesis of different terpenoids via the mevalonte pathway, for example perillyl alchol. Inhibits the HMG-CoA reductase activity and reduce cholesterol in rodents Has a larvicidal activity against the Aedes aegypti mosquito.

Demonstrated to activate nuclear receptors, thus transcriptionally affecting cancer cell growth in human MIA PaCa2 cells. In adipose tissue (3T3 cells), PPARγ expression is up-regulated and activation has been seen to improve insulin resistance. In HepG2 cells, treatment with farnesol up-regulates the expression of PPARα. In liver cells, activation of PPARα would reduce the circulating levels of lipids. May serve as an intermediate in the synthesis of different terpenoids in the mevalonate pathway. Induce apoptosis in cervical cancer and lymphoma.

In adipose tissue, PPARγ activation has been seen to improve insulin resistance. Suppresses the hepatic 3-hydroxy-3 methylglutaryl coenzyme A (HMG-CoA) reductase activity and reduces serum cholesterolin rats and humans. Has a concentration dependent impact on tumour cell proliferation in the murine cells: P388, B16 melanoma, Morris hepatoma 7777 cells and human MIA PaCa2 pancreatic ductal adenocarcinoma cells. By suppressing the mevalonate pathway in tumour cells and not normal cells. May serve as an intermediate in the synthesis of different terpenoids in the mevalonate pathway. Has an in vivo antitumour activity against murine hepatoma, melanoma and leukaemia cells.

Modulates the expression of the pro-apoptotic factors (BAX, Bcl-2) that would subsequently result in mitochondrial dysfunction and apoptosis via the induction of the caspase signalling cascade in Hela cells. The main mechanism of killing leishmanial parasites is by controlling the expression of NO. Linalool has been shown to increase the production of NO in L. amaxonensis-infected macrophages and not in the mammalian Vero cells (kidney epithelial cells from African green monkeys). The chronic increase in NO would induce the activation of the UPR process. Linalool also affects mitochondrial dysfunction, resulting in mitochondrial swelling. Linalool has also been documented to change the chromatin organization.

This lactone has been well-documented to diffuse through the cell membranes and block the calcium pump via lipophilic interactions, inhibiting the Ca2+ transporting ATPases in Plasmodium falciparum. The same mechanism is proposed in cancer cells. Blocking the SERCA pump, prevents the Ca2+ flux between the ER and the cytosol, resulting in the accumulation of Ca2+ and subsequent ER stress and apoptosis.

Hypothesized to kill the Plasmodium spp. gametocytes by inhibiting the activity of the sarcoendoplasmic reticulum Ca2+ ATPase pump. Leishmania spp. have been documented to reduce the expression of inducible nitric oxide synthase (iNOS) and subsequently nitric oxide in macrophages from BALB/c mice. By treating Leishmania-infected macrophages with artemisinin, the expression of iNOS and NO increased, and the levels of INF-γ decreased to a level comparable with uninfected macrophages. When bound to Fe(II), artemisinin is cleaved, producing free radicals toxic to tumour cells and may contribute to the anti-leishmanial activity. Artemisinin inhibits the activity of NF-κB that is elevated in tumour cells, rendering the cells sensitive to chemotherapy.

SERCA pump iNOS

SERCA pump

Biological impact/function/mechanism(s)

Biological target(s)

Examples of the terpenoids that have shown promise to alleviate metabolic disorders

Terpenoid name

Table 5

[32,138,145,146]

[140–142,144]

[13,140–143]

[13,133,135–139]

[13,114,131–135]

[130]

[129]

[55,56,124,125,127,128]

[40,41,124–126]

References(s)

Natasha Beukes et al. Terpenoids and ER stress

1515

1516 [34,35,154,155]

ATF, activating transcription factor; Bcl-2, beta-cell lymphoma 2; BAX, beta-cell lymphoma 2 antagonist/killer; HMG-CoA, hepatic 3-hydroxy-3methylglutaryl coenzyme A; IGF, insulin-like growth factor; IL-1β/6, interleukin 1 beta/6; LANCL2, lanthionine synthase C-like 2; MCP-1, monocyte chemoattractant protein 1; Nrf2, nuclear respiratory factor 2; NF-κB, nuclear factor-kappa B; iNOS, inducible nitric oxide synthase, PPAR, peroxisome proliferation-activated receptor; SERCA, sarcoendoplasmic reticulum calcium ATPase pump.

iNOS Insulin Cholesterol Cytokines Fibrin

Using a murine model using Swiss mice, where the mice were exposed to an iNOS inhibitor, the gastroprotective properties of marrubiin was strongly related to the production of nitric oxide, the increased synthesis of mucus and the decrease in gastric acid secretion. Using INS-1 cells, marrubiin increased insulin expression in a dose-dependent manner, up-regulated the expression of glucose transport 2 genes, and increased the respiratory rate and mitochondrial membrane potential under hyperglycaemic conditions. Using male Wistar rats, marrubiin, increased insulin secretion under hyperglycaemic conditions, which was linked to mitochondrial metabolism. As ATP production increased, stimulated by an increase in the mitochondrial membrane potential, increasing oxygen consumption via the respiratory chain. In the obese Wistar rat models, marrubiin treatment was seen to increase HDL-cholesterol levels while normalizing the total cholesterol levels by decreasing the LDL-cholesterol, the atherogenic index. Marrubiin normalized IL-1β and IL-6 levels, and decreased TNF-α levels in obese rats. The anti-inflammatory activity of marrubiin is proposed to be linked to the suppression of the NF-κB signalling pathway. Marrubiin has also been shown normalize fibrin levels in the obese rat model. This terpenoid inhibits platelet aggregation and subsequently suppressing coagulation. Marrubiin inhibits collagen and thrombin-induced Ca2+ mobilization.

Gastric ulcers Diabetes

Marrubiin

[20,21,150–152]

[153]

Leishmania

18β-glycyrrhetinic acid

References(s) [147–149]

Treating murine peritoneal macrophages infected with L. donovani promastigotes, increased iNOS expression would induce UPR, resulting in the release in cytokines. The expression of IL10 and IL4 were down-regulated and the expression of IL-12, INF-γ and TNF-α were up-regulated in the macrophages. 18β-glycyrrhetinic acid induced the migration of NF-κB into the nucleus of the parasite-infected macrophages, decreasing the levels of IκB in the cytoplasm. The decrease in IκB in the cytoplasm increased the phosphorylation of IκBα, activating the kinase activity. However, 18β-glycyrrhetinic acid did not directly affect the activity of IκB kinase.

LPS IGF-1 IL-1β IL-6 MCP-1

Inflammation associated with obesity and atherosclerosis Cardiovascular disease

Carotenoids (e.g. α-carotene, β-carotene and lycopene)

iNOS Cytokines NF-κB signalling

Decreases fasting glucose, improves glucose intolerance and increases the expression of PPARγ and its responsive genes. Abscisic acid increases the PPARγ reporter activity in bone marrow-derived macrophages, which is dependent on the expression if LANCL2 as demonstrated by LANCL2 knock-down studies. Abscisic acid shown to suppress the LPS-mediated expression of prostaglandin E2, MCP-1 production and inflammation through the activation of PPARγ and suppressing NF-κB in T cells. Decreases LPS-mediated inflammation and regulates the innate immune responses through the bifurcating pathway using LANCL2. Suggested to be an endogenous pro-inflammatory cytokine in human granulocytes that has the ability to stimulate the secretion of insulin. α-Carotene acts as the precursor for the synthesis of vitamin A β-carotene inhibits the expression of inflammatory genes in LPS-stimulated macrophages. Lycopene is a strong antioxidant that has been documented to have several mechanisms of activity, including anti-angiogenesis and induction of apoptosis through the inhibition of insulin-like growth factor 1-stimulated cell proliferation, inhibition of LDL oxidation and lipid peroxidation. Lycopene also reduces the expression of inflammatory markers IL-1β, IL-6 and MCP-1.

PPARγ LPS LANCL2 NF-κB

Abscisic acid

Diabetes Inflammation related to obesity

Metabolic disorder(s)

Terpenoid name

Biological impact/function/mechanism(s)

Continued Biological target(s)

Table 5

Terpenoids and ER stress Natasha Beukes et al.

© 2014 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 66, pp. 1505–1525

Natasha Beukes et al.

Obesity is a common occurrence in diabetic individuals. Obesity linked to diabetes has been extensively documented to induce ER stress in a number of organs, including the liver, heart, pancreas, kidneys, adipose tissue and the central nervous system.[169–178] For example, it has been observed that obesity can activate ER stress in the liver by suppressing the insulin receptor signalling through the activation of JNK and the phosphorylation of the insulin receptor substrate[171,179] In a study by Zhang and associates, the effect of genipin on mitochondrial respiration and pancreatic islet function was tested.[180] They observed that genipin not only increase mitochondrial membrane potential by closing KATP plasma membrane channels and increased ATP levels but also showed an acute reversal of high glucose and obesity-associated β-cell dysfunction. These results were observed when genipin was used to inhibit uncoupling protein 2 (UCP2) expression in a dose-dependent manner;[180] thus, genipin has the ability to affect an ER-associated protein, Grp78, and a mitochondrial protein, UCP2.

Inflammatory bowel diseases The two main inflammatory bowel diseases that affect the colon or the intestines are Crohn’s disease and ulcerative colitis, both have been linked to ER stress, especially the IRE1 signalling pathway and its involvement in inflammation.[181–183] However, the activation of the PERK and ATF6 pathways has also been noted.[184] Thus, targeting the UPR pathways associated with the expression of proteins (e.g. pro-inflammatory cytokines) that exaggerate inflammation could possibly serve as a valuable treatment plan to alleviate the discomfort and slowdown the progression of the diseases. Several terpenoids have been documented to alleviate inflammation, examples include abscisic acid and carotenoids (Table 4).

Cancers The field of cancer research is vast due to the wide variety of cancers and carcinomas afflicting the global population. The prevalence of cancer has increased substantially over the last few decades, and as such a substantial amount of research has been and continues to be performed searching for natural compounds that show potential in the treatment of cancers in general. Auf and associates[68] showed that there is a correlation between IRE1 signalling pathway and the reduction of functional tumour blood vessels in glioma, cell invasion and vessel co-option. They showed that by blocking the IRE1 pathway, they modified the glioma expansion by reducing angiogenesis and promoting cell invasion, disrupting the normal tissue architecture and induced reactive gliosis.

Terpenoids and ER stress

They also found that interfering with IRE1 activity resulted in the down-regulation of VEGF-A, IL-1β, IL-6 and IL-8, thus indicating that the IRE1 signalling pathway may be essential for angiogenesis and tumour progression and antiinflammatory action. The use of C. sativa as part of cancer treatments is wellestablished and has been used to relieve a variety of ailments, ranging from pain (Sativex®, GW Pharmaceuticals, Salisbury, UK), inflammation and nausea (Cesamet®, Valeant Pharmaceuticals, Toronto, Canada) for decades, thus is possibly the best example of the use of terpenoids in the treatment of cancers.[26] Cannabis sativa contains a variety of phytochemicals; however, the terpenoids that have shown promise in cancer treatments are commonly known as cannabinoids (THC, CBD, CBN). In relation to terpenoids, cannabinoids are highly regarded for their general antitumour properties against a wide variety of cancers. Antitumour properties have been documented in gliomers, melanomas, lymphomas and carcinomas.[185] In general, cannabinoids have been documented to affect the endocannabinoid system by binding to the cannabinoid receptors 1 and 2 (CB1 and CB2). THC impairs cell proliferation via the CB2 receptor, inhibition of the protumourigenic protein kinase B, impairing angiogenesis and subsequently induce the apoptotic signalling cascade.[186] Treatment with THC has resulted in increased cell proliferation; however, in these cases, it was noted that the increased cell proliferation was due to the use of low dosages and compromised immune systems of the animal models.[187–190] For THC treatment to be effective in inhibiting cell proliferation, the dosage and general immunity need to be taken into account. CBD induces ER stress in a variety of cancers, including gliomas, liver carcinoma and pancreatic cancer.[94,122,191] CBD, unlike THC, has a low affinity for the endocannabinoid receptors CB1 and CB2. It has been proposed to bind to the vanilloid receptors,[23] induce ER stress and inhibit the AKT/mammalian target of rapamycin 1 signalling cascade, initiating the ‘cross-talk’ between the ER stress pathways and mitochondrial dysfunction, and ultimately resulting in autophagy and apoptosis.[192] Over the last few decades, additional research has been performed in search of terpenoids other than cannabinoids that show potential in the treatment of cancers. Additional terpenoid examples and derived therapeutic drugs used in the potential treatment of cancer are available in other review articles. For example, artemisinin (commonly used to treat malaria), thapsigargin and partenolide have been used in the cancer clinical trials because of their ability to induce ER stress by blocking the sarco/ER calcium ATPase pump, and subsequently affecting a variety of downstream metabolic signalling pathways integral to cell survival and proliferation.[126,193]

© 2014 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 66, pp. 1505–1525

1517

Terpenoids and ER stress

Natasha Beukes et al.

Neurodegenerative diseases

Cerebral malaria

As previously mentioned, the ER is essential for the correct folding of proteins and as such ER dysfunction would be expected to play an important role in the development and progression of diseases that may arise from the resultant effects of accumulated incorrectly folded proteins or even protein overloading. The idea that ER dysfunction and the subsequent accumulation of incorrectly folded neuronal proteins may play an important role in the progression of most neurological diseases is gaining popularity; however, it is important to note that not all neurological diseases arise from ER dysfunction. Factors such as the duration and severity of ER stress, the amount of cellular energy expended to alleviate ER dysfunction, and ER overload may also play a role in the different neurological diseases. In some cases, neurological diseases such as Huntington’s disease may induce ER stress as a consequence of the disease, instead of ER stress being associated with the progression of the neurodegenerative disease. Neurological diseases that have been linked to ER dysfunction include: epilepsy, cerebral malaria, Alzheimer’s, Parkinson’s, multiple sclerosis, sleep apnoea and prion diseases. In the past, the intrinsic and extrinsic apoptotic pathways studied in neuronal cells after seizures focused on the mitochondrial and death receptor analysis, respectively. However, in recent years, the importance and role of ER stress and dysfunction has emerged as an alternative intrinsic pathway.[55,194,195] A number of research articles and reviews are available, linking neurological diseases to ER dysfunction and stress, and as such, this review will briefly provide two examples, epilepsy and cerebral malaria, that have not been studied to the same extent as the more common neurological diseases.[54,80,196–200]

The contraction and progression of cerebral malaria develops as a result of complications after the infection of Plasmodium falciparum. Even though a substantial amount of people die from the disease annually, the pathogenesis in humans remains largely unstudied because of experimental analysis only occurring post-mortem.[205] In recent years, a murine model has been used to study the pathogenesis of cerebral malaria after infected with P. berghei. The murine model is used to study cerebral malaria because it displays symptoms typical of humans cerebral malaria. Observations from these studies have indicated that any complications arising from infections, which may involve the activation of platelets, increase expression of cytokines, disruption of the blood-brain barrier and the blockage of microvessels by infected red blood cells. At the stage, where the infection results in cell death, several signalling pathways have been implicated, including mitochondrial dysfunction, activation of calciumdependent and independent kinases (e.g. JNK), caspases and recently ER stress.[205–207] Anand and Babu[205] illustrated the role of ER stress in neuronal cell death in mice infected with P. berghei. They observed the activation of all the signalling pathways involved in UPR and that neuron apoptosis occurred via the activation of the IRE1TRAF2-ASK-JNK signalling pathway; the increased levels and accumulation of CHOP, phosphorylated eIF2α, ATF4, GADD34, BAX, caspase 3 cleavage and the downregulation of the prosurvival proteins (Bcl-2, GRP78, calreticulin, calnexin). This indicates the involvement of both the IRE1 and PERK signalling pathways in the progression of the disease.

Viral infections Epilepsy In general, the role of the ER in neurodegenerative diseases has increasingly been recognized; however, very little is known about the role of ER stress in epilepsy. A recent study by Liu and associates have indicated the expression of ER stress related chaperones (GRP78 and GRP94) were up-regulated after seizures in the temporal neocortex of epileptic patients, confirming what had previously been seen in literature.[201] They also documented the presence of ER stress through the activation of the IRE1α : TRAF2 : JNK signalling pathway, thus indicating that in cases where brain injury has occurred as a result of epileptic seizures, ER stress and specifically the IRE1α signalling pathway may play an important role in the apoptosis of neuronal cells.[201] Other evidence linking ER stress to epilepsy include disturbances in the calcium homeostasis, increased expression of pro-inflammatory cytokines (IL-1β, IL-6 and TNFα), and the up-regulation of XBP1.[202–204] 1518

Like most disease states, viral infections have been documented to affect ER homeostasis. Once the cells are infected, the viral replication proteins increase pressure on the cellular functions and induce the ER’s UPR pathways. However, because irresolvable ER stress generally results in the induction of apoptosis, which would not be beneficial to the survival of the virus. To overcome this obstacle, several viruses have been documented with the ability to regulate the UPR pathways, ensuring that favourable conditions are maintained. Hepatitis C virus affects the ATF6 and IRE1 pathways by cleaving ATF6 into its active conformation, subsequently up-regulating the expression of chaperones, and prevents the activation of XBP1.[208] The dengue virus induces the expression of XBP1, EDEM1, ER DNA J homologue 4, p58IPK and GRP78.[209,210] West Nile virus strains affect all the UPR pathways by up-regulating pro-apoptotic factors (CHOP, caspase 3) and GADD34,[211] XBP1 and INF signalling pathways.[212]

© 2014 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 66, pp. 1505–1525

Natasha Beukes et al.

Terpenoids and ER stress

Conclusions

JUN, SAPK and ATF4 have not been well-established. The exuberant number of terpenoinds that have been identified, increase the possibility that more plant-derived compounds can be used to identify novel treatments for more diseases and viral infections, thus producing a sustainable alternative to the treatment of diseases.

In general, the majority of research that is performed on signal cascades involve the prominent protein that initiate the signalling pathways; the effect terpenoids have on the expression or function of the UPR proteins TRAF2, JIK,

References 1. Paduch R et al. Terpenes: substances useful in human healthcare. Arch Immunol Ther Exp (Warsz) 2007; 5: 315–327. 2. Ukwe CV, Ubaka CM. Hypoglycemic activity of leaves of Acanthus montanus T.Anderson (Acanthaceae) in rats. Int J Diabetes Dev Ctries 2011; 1: 32–36. 3. Ashok Kumar BS et al. Antidiabetic, antihyperlipidemic and antioxidant activities of methanolic extract of Amaranthus viridis Linn in alloxan induced diabetic rats. Exp Toxicol Pathol 2012; 1–2: 75–79. 4. Thakur CP. Emblica officinalis reduces serum, aortic and hepatic cholesterol in rabbits. Experientia 1985; 3: 423–424. 5. Giancarlo S et al. Hypoglycaemic activity of two spices extracts: Rhus coriaria L and Bunium persicum Boiss. Nat Prod Res 2006; 9: 882–886. 6. Scartezzini P, Speroni E. Review on some plants of Indian traditional medicine with antioxidant activity. J Ethnopharmacol 2000; 1–2: 23–43. 7. Asada R et al. The signalling from endoplasmic reticulumresident bZIP transcription factors involved in diverse cellular physiology. J Biochem 2011; 5: 507–518. 8. Cunard R, Sharma K. The endoplasmic reticulum stress response and diabetic kidney disease. Am J Physiol Renal Physiol 2011; 5: 1054–1061. 9. Lee AH et al. XBP-1 regulates a subset of endoplasmic reticulum resident chaperone genes in the unfolded protein response. Mol Cell Biol 2003; 21: 7448–7459.

10. Sriburi R et al. XBP1: a link between the unfolded protein response, lipid biosynthesis, and biogenesis of the endoplasmic reticulum. J Cell Biol 2004; 1: 35–41. 11. Acosta-Alvear D et al. XBP1 controls diverse cell type- and conditionspecific transcriptional regulatory networks. Mol Cell 2007; 1: 53– 66. 12. Oslowski CM, Urano F. The binary switch that controls the life and death decisions of ER stressed β cells. Curr Opin Cell Biol 2011; 2: 207–215. 13. Burke YD et al. Inhibition of pancreatic cancer growth by the dietary isoprenoids farnesol and geraniol. Lipids 1997; 2: 151–156. 14. Goto T et al. Various Terpenoids derived from herbal and dietary plants function as PPAR modulators and regulate carbohydrate and lipid metabolism. PPAR Res 2010; doi: 10.1155/2010/483958 15. Laule O et al. Crosstalk between cytosolic and plastidial pathways of isoprenoid biosynthesis in Arabidopsis thaliana. Proc Natl Acad Sci U S A 2003; 11: 6866–6871. 16. Peñuelas J, Munné-Bosch S. Isoprenoids: an evolutionary pool for photoprotection. Trends Plant Sci 2005; 4: 166–169. 17. Withers ST, Keasling JD. Biosynthesis and engineering of isoprenoid small molecules. Appl Microbiol Biotechnol 2007; 5: 980–990. 18. Milborrow BV. The metabolism of abscisic acid. J Exp Bot 1970; 1: 17–29. 19. Bangerth F. Changes in the ratio of cis-trans to trans-trans abscisic acid during ripening of apple fruits. Planta 1982; 3: 199–203.

© 2014 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 66, pp. 1505–1525

20. Agarwal S, Rao AV. Tomato lycopene and its role in human health and chronic diseases. Can Med Assoc J 2000; 6: 739–744. 21. Gouranton E et al. Lycopene inhibits proinflammatory cytokine and chemokine expression in adipose tissue. J Nutr Biochem 2011; 7: 642–648. 22. Asaduzzaman M et al. Growing carrots hydroponically using perlite substrates. Sci Hortic (Amsterdam) 2013; 159: 113–121. 23. Bisogno T et al. Molecular targets for cannabidiol and its synthetic analogues: effect on vanilloid VR1 receptors and on the cellular uptake and enzymatic hydrolysis of anandamide. Br J Pharmacol 2001; 4: 845–852. 24. Mecha M et al. Cannabidiol protects oligodendrocyte progenitor cells from inflammation-induced apoptosis by attenuating endoplasmic reticulum stress. Cell Death Dis 2012; 6: e331–e338. 25. Lim MP et al. Cannabidiol causes activated hepatic stellate cell death through a mechanism of endoplasmic reticulum stress-induced apoptosis. Cell Death Dis 2011; 6: e170. 26. Caffarel MM et al. Cannabinoids: a new hope for breast cancer therapy? Cancer Treat Rev 2012; 7: 911–918. 27. Gallant M et al. Biological effects of THC and a lipophilic cannabis extract on normal and insulin resistant 3T3-L1 adipocytes. Phytomedicine 2009; 10: 942–949. 28. Ku CM, Lin JY. Anti-inflammatory effects of 27 selected terpenoid compounds tested through modulating Th1/Th2 cytokine secretion profiles using murine primary splenocytes. Food Chem 2013; 2: 1104–1113. 29. Russo EB. Taming THC: potential cannabis synergy and 1519

Terpenoids and ER stress

30.

31.

32.

33.

34.

35.

36.

37.

38.

39.

40.

41.

1520

phytocannabinoid-terpenoid entourage effects. Br J Pharmacol 2011; 7: 1344–1364. Yamazaki M et al. Genipin suppresses A23187-induced cytotoxicity in Neuro2a cells. Biol Pharm Bull 2009; 6: 1043–1046. Croteau RB et al. Menthol biosynthesis and molecular genetics. Naturwissenschaften 2005; 12: 562– 577. Stark MJ et al. Chemotherapy of pancreatic cancer with the monoterpene perillyl alcohol. Cancer Lett 1995; 1: 15–21. Kimura KI et al. Kujigamberol, a new dinorlabdane diterpenoid isolated from 85 million years old Kuji amber using a biotechnological assay. Fitoterapia 2012; 5: 907–912. de Oliveira AP et al. Gastroprotective activity of methanol extract and marrubiin obtained from leaves of Marrubium vulgare L. (Lamiaceae). J Pharm Pharmacol 2011; 9: 1230– 1237. Mnonopi N et al. The cardioprotective effects of marrubiin, a diterpenoid found in Leonotis leonurus extracts. J Ethnopharmacol 2011; 1: 67–75. Kamo T et al. Anti-inflammatory cyathane diterpenoids from Sarcodon scabrosus. Biosci Biotechnol Biochem 2004; 6: 1362–1365. Masubuti H et al. Establishment of benzodioxazine core structure for sarcodonin class of natural products by X-ray analysis. Org Lett 2013; 9: 2076–2079. Chang UM et al. Ganoderiol F, a ganoderma triterpene, induces senescence in hepatoma HepG2 cells. Life Sci 2006; 12: 1129–1139. Anfosso L et al. Microarray expression profiles of angiogenesis-related genes predict tumor cell response to artemisinins. Pharmacogenomics J 2006; 4: 269–278. Krishna S et al. Artemisinins: mechanisms of action and potential for resistance. Drug Resist Updat 2004; 4–5: 233–244. Sen R et al. Efficacy of artemisinin in experimental visceral leishmania-

Natasha Beukes et al.

42.

43.

44.

45.

46.

47.

48.

49.

50.

51.

52.

sis. Int J Antimicrob Agents 2010; 1: 43–49. Barrero AF et al. New sources and antifungal activity of sesquiterpene lactones. Fitoterapia 2000; 1: 60–64. Butturini E et al. Two naturally occurring terpenes, dehydrocostuslactone and costunolide, decrease intracellular GSH content and inhibit STAT3 activation. PLoS ONE 2011; 5: 15–21. Hung JY et al. Oxidative and endoplasmic reticulum stress signaling are involved in dehydrocostuslactone-mediated apoptosis in human non-small cell lung cancer cells. Lung Cancer 2010; 3: 355–365. Jamalian A et al. Chemical composition and antifungal activity of Matricaria recutita flower essential oil against medically important dermatophytes and soil-borne pathogens. J de Mycol Med 2012; 4: 308–315. Ghantous A et al. Parthenolide: from plant shoots to cancer roots. Drug Discov Today 2013; 18: 894–905. Feher JJ et al. Ryanodine-sensitive, thapsigargin-insensitive calcium uptake in rat ventricle homogenates. Mol Cell Biochem 1998; 1–2: 9–17. Kozutsumi Y et al. The presence of malfolded proteins in the endoplasmic reticulum signals the induction of glucose-regulated proteins. Nature 1988; 6163: 462–464. Dorner AJ et al. Increased synthesis of secreted proteins induces expression of glucose-regulated proteins in butyrate-treated Chinese hamster ovary cells. Journal of Biological Chemistry 1989; 34: 20602–20607. Harding HP et al. An integrated stress response regulates amino acid metabolism and resistance to oxidative stress. Mol Cell 2003; 3: 619–633. He B. Viruses, endoplasmic reticulum stress, and interferon responses. Cell Death Differ 2006; 3: 393–403. Koumenis C et al. Regulation of protein synthesis by hypoxia via activation of the endoplasmic reticulum

53.

54.

55.

56.

57.

58.

59.

60.

61.

62.

63.

64.

65.

kinase PERK and phosphorylation of the translation initiation factor eIF2α. Mol Cell Biol 2002; 21: 7405– 7416. Häcker G. The morphology of apoptosis. Cell Tissue Res 2000; 1: 5–17. Doyle KM et al. Unfolded proteins and endoplasmic reticulum stress in neurodegenerative disorders. J Cell Mol Med 2011; 10: 2025–2039. Xu C et al. Endoplasmic reticulum stress: cell life and death decisions. J Clin Investig 2005; 10: 2656–2664. Lai E et al. Endoplasmic reticulum stress: signaling the unfolded protein response. Physiology 2007; 3: 193– 201. Yoshida H. ER stress and diseases. Fed Eur Biochem Soc J 2007; 3: 630– 658. Wu J, Kaufman RJ. From acute ER stress to physiological roles of the unfolded protein response. Cell Death Differ 2006; 3: 374–384. Hotamisligil GS. Endoplasmic reticulum stress and the inflammatory basis of metabolic disease. Cell 2010; 6: 900–917. Ortsäter H, Sjöholm A. A busy cellendoplasmic reticulum stress in the pancreatic β-cell. Mol Cell Endocrinol 2007; 1–2: 1–5. Oyadomari S, Mori M. Roles of CHOP/GADD153 in endoplasmic reticulum stress. Cell Death Differ 2004; 4: 381–389. Oyadomari S et al. Endoplasmic reticulum stress-mediated apoptosis in pancreatic β-cells. Apoptosis 2002; 4: 335–345. Rao RV et al. Coupling endoplasmic reticulum stress to the cell death program. Cell Death Differ 2004; 4: 372–380. Szegezdi E et al. Mediators of endoplasmic reticulum stress-induced apoptosis. Eur Mol Biol Organ Rep 2006; 9: 880–885. Kitao Y et al. Expression of the endoplasmic reticulum molecular chaperone (ORP150) rescues hippocampal neurons from glutamate toxicity. J Clin Invest 2001; 10: 1439– 1450.

© 2014 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 66, pp. 1505–1525

Natasha Beukes et al.

66. Lee JW et al. ER stress is implicated in mitochondrial dysfunctioninduced apoptosis of pancreatic beta cells. Mol Cells 2010; 6: 545–549. 67. Schröder M, Kaufman RJ. The mammalian unfolded protein response. Annu Rev Biochem 2005b; 74: 739– 789. 68. Auf G et al. Inositol-requiring enzyme 1α is a key regulator of angiogenesis and invasion in malignant glioma. Proc Natl Acad Sci U S A 2010; 35: 15553–15558. 69. Lin JH et al. IRE1 signaling affects cell fate during the unfolded protein response. Science 2007; 5852: 944– 949. 70. Urano F et al. Coupling of stress in the ER to activation of JNK protein kinases by transmembrane protein kinase IRE1. Science 2000; 5453: 664– 666. 71. Bellail AC et al. Microregional extracellular matrix heterogeneity in brain modulates glioma cell invasion. Int J Biochem Cell Biol 2004; 6: 1046– 1069. 72. Bennewith KL et al. The role of tumor cell-derived connective tissue growth factor (CTGF/CCN2) in pancreatic tumor growth. Cancer Res 2009; 3: 775–784. 73. Pugh CW, Ratcliffe PJ. Regulation of angiogenesis by hypoxia: role of the HIF system. Nat Med 2003; 6: 677– 684. 74. Hetz C, Glimcher LH. Fine-tuning of the unfolded protein response: assembling the IRE1α Interactome. Mol Cell 2009; 5: 551–561. 75. Ron D, Walter P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol 2007; 7: 519–529. 76. Schröder M, Kaufman RJ. ER stress and the unfolded protein response. Mutat Res 2005a; 1–2: 29–63. 77. Yoshida H et al. XBP1 mRNA is induced by ATF6 and spliced by IRE1 in response to ER stress to produce a highly active transcription factor. Cell 2001; 7: 881–891. 78. Gardner OS et al. Peroxisome proliferator-activated receptor γindependent activation of p38

Terpenoids and ER stress

79.

80.

81.

82.

83.

84.

85.

86.

87.

MAPK by thiazolidinediones involves calcium/calmodulin-dependent protein kinase II and protein kinase R: correlation with endoplasmic reticulum stress. J Biol Chem 2005; 11: 10109–10118. Harding HP et al. Protein translation and folding are coupled by an endoplasmic-reticulum-resident kinase. Nature 1999; 6716: 271–274. Roussel BD et al. Endoplasmic reticulum dysfunction in neurological disease. Lancet Neurol 2013; 1: 105–118. Lu PD et al. Translation reinitiation at alternative open reading frames regulates gene expression in an integrated stress response. J Cell Biol 2004; 1: 27–33. Ameri K, Harris AL. Activating transcription factor 4. Int J Biochem Cell Biol 2008; 1: 14–21. Ohoka N et al. TRB3, a novel ER stress-inducible gene, is induced via ATF4-CHOP pathway and is involved in cell death. Eur Mol Biol Organ J 2005; 6: 1243–1255. Brush MH et al. Growth arrest and DNA damage-inducible protein GADD34 targets protein phosphatase 1α to the endoplasmic reticulum and promotes dephosphorylation of the α subunit of eukaryotic translation initiation factor 2. Mol Cell Biol 2003; 4: 1292– 1303. Ma Y, Hendershot LM. Delineation of a negative feedback regulatory loop that controls protein translation during endoplasmic reticulum stress. J Biol Chem 2003; 37: 34864– 34873. Morse E et al. TRB3 is stimulated in diabetic kidneys, regulated by the ER stress marker CHOP, and is a suppressor of podocyte MCP-1. Am J Physiol Renal Physiol 2010; 5: F965– F972. Fawcett TW et al. Complexes containing activating transcription factor (ATF)/cAMP-responsiveelement-binding protein (CREB) interact with the CCAAT/enhancerbinding protein (C/EBP)-ATF composite site to regulate Gadd153

© 2014 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 66, pp. 1505–1525

88.

89.

90.

91.

92.

93.

94.

95.

96.

97.

98.

expression during the stress response. Biochem J 1999; 1: 135–141. Harding HP et al. Regulated translation initiation controls stressinduced gene expression in mammalian cells. Mol Cell 2000; 5: 1099–1108. Marciniak SJ et al. CHOP induces death by promoting protein synthesis and oxidation in the stressed endoplasmic reticulum. Genes Dev 2004; 24: 3066–3077. Ma Y et al. Two distinct stress signaling pathways converge upon the CHOP promoter during the mammalian unfolded protein response. J Mol Biol 2002; 5: 1351–1365. Selim E et al. Fibrates upregulate TRB3 in lymphocytes independent of PPARα by augmenting CCAAT/enhancer-binding proteinβ (C/EBPβ) expression. Mol Immunol 2007; 6: 1218–1229. Southwood CM et al. The unfolded protein response modulates disease severity in Pelizaeus-Merzbacher disease. Neuron 2002; 4: 585–596. Cullinan SB, Diehl JA. Coordination of ER and oxidative stress signaling: the PERK/Nrf2 signaling pathway. Int J Biochem Cell Biol 2006; 3: 317– 332. Salazar M et al. Cannabinoid action induces autophagy-mediated cell death through stimulation of ER stress in human glioma cells. J Clin Invest 2009; 5: 1359–1372. Ye J et al. ER stress induces cleavage of membrane-bound ATF6 by the same proteases that process SREBPs. Mol Cell 2000; 6: 1355–1364. Shen J et al. ER stress regulation of ATF6 localization by dissociation of BiP/GRP78 binding and unmasking of golgi localization signals. Dev Cell 2002; 1: 99–111. Du Z et al. Inhibition of IFN-α signaling by a PKC- and protein tyrosine phosphatase SHP-2dependent pathway. Proc Natl Acad Sci USA 2005; 29: 10267– 10272. Werner ED et al. Proteasomedependent endoplasmic reticulumassociated protein degradation: an 1521

Terpenoids and ER stress

99.

100.

101.

102.

103.

104.

105.

106.

107.

108.

109.

110.

1522

unconventional route to a familiar fate. Proc Natl Acad Sci U S A 1996; 24: 13797–13801. Yoshida H et al. A time-dependent phase shift in the mammalian unfolded protein response. Dev Cell 2003; 2: 265–271. Haze K et al. Mammalian transcription factor ATF6 is synthesized as a transmembrane protein and activated by proteolysis in response to endoplasmic reticulum stress. Mol Biol Cell 1999; 11: 3787–3799. Li M et al. ATF6 as a transcription activator of the endoplasmic reticulum stress element: Thapsigargin stress-induced changes and synergistic interactions with NF-Y and YY1. Mol Cell Biol 2000; 14: 5096–5106. Suzuki H et al. Characteristic properties of genipin as an activator in neuronal nitric oxide synthase. J Health Sci 2007; 6: 730–733. Yamazaki M, Chiba K. Neurotrophic effects of genipin on Neuro2a cells. J Health Sci 2005; 6: 687–692. Yamazaki M et al. Differences in neuritogenic response to nitric oxide in PC12 and PC12h cells. Neurosci Lett 2006; 2–3: 222–225. Yamazaki M et al. Neuro2a cell death induced by 6-hydroxydopamine is attenuated by genipin. J Health Sci 2008; 6: 638–644. Yamazaki M. Chibaa K. Expression of functional nitric oxide synthase for neuritogenesis in PC12h cells. J Health Sci 2006; 6: 769–773. Yamazaki M et al. Prevention of the neurotoxicity of the amyloid β protein by genipin. Biol Pharm Bull 2001; 12: 1454–1455. Ohkubo T et al. Detection of genipin/geniposide-target molecules by a geniposide overlay method using anti-geniposide antibody. J Health Sc 2004; 2: 193–196. García-Piñeres AJ et al. Cysteine 38 in p65/NF-κB Plays a crucial role in DNA binding inhibition by sesquiterpene lactones. J Biol Chem 2001; 43: 39713–39720. Brüne B et al. Nitric oxide and its role in apoptosis. Eur J Pharmacol 1998; 3: 261–272.

Natasha Beukes et al.

111. He L et al. Isoprenoids suppress the growth of murine B16 melanomas in vitro and in vivo. J Nutr 1997; 5: 668–674. 112. Mo H, Elson CE. Apoptosis and cell-cycle arrest in human and murine tumor cells are initiated by isopreonoids. J Nutr 1999; 4: 804– 813. 113. Roullet JB et al. Farnesol inhibits L-type Ca 2+ channels in vascular smooth muscle cells. J Biol Chem 1997; 51: 32240–32246. 114. Yu SG et al. Geraniol, an inhibitor of mevalonate biosynthesis, suppresses the growth of hepatomas and melanomas transplanted to rats and mice. J Nutr 1995; 11: 2763–2767. 115. Pahl HL, Baeuerle PA. The ER overload response: activation of NF-κB. Trends Biochem Sci 1997; 2: 63–67. 116. Qu L et al. Endoplasmic reticulum stress induces p53 cytoplasmic localization and prevents p53-dependent apoptosis by a pathway involving glycogen synthase kinase-3β. Genes Dev 2004; 3: 261–277. 117. Chawta A et al. Nuclear receptors and lipid physiology: opening the x-files. Science 2001; 5548: 1866– 1870. 118. Lazar MA. East meets West: an herbal tea finds a receptor. J Clin Invest 2004; 1: 23–25. 119. Dey A et al. Double-edged swords as cancer therapeutics: simultaneously targeting p53 and NF-κB pathways. Nat Rev Drug Discov 2008; 12: 1031– 1040. 120. Teixeira-Clerc F et al. CB1 cannabinoid receptor antagonism: a new strategy for the treatment of liver fibrosis. Nat Med 2006; 6: 671– 676. 121. Carracedo A et al. The stressregulated protein p8 mediates cannabinoid-induced apoptosis of tumor cells. Cancer Cell 2006a; 4: 301–312. 122. Carracedo A et al. Cannabinoids induce apoptosis of pancreatic tumor cells via endoplasmic reticulum stress-related genes. Cancer Res 2006b; 13: 6748–6755.

123. Mechoulam R et al. Cannabidiol – recent advances. Chem Biodivers 2007; 8: 1678–1692. 124. Eckstein-Ludwig U et al. Artemisinins target the SERCA of Plasmodium falciparum. Nature 2003; 6951: 957–961. 125. Denmeade SR, Isaacs JT. The SERCA pump as a therapeutic target: making a ‘smart bomb’ for prostate cancer. Cancer Biol Ther 2005; 1: 14–22. 126. Ghantous A et al. What made sesquiterpene lactones reach cancer clinical trials? Drug Discov Today 2010; 15–16: 668–678. 127. Cunha DA et al. Initiation and execution of lipotoxic ER stress in pancreatic β-cells. J Cell Sci 2008; 14: 2308–2318. 128. Gurzov EN et al. Signaling by IL-1β + IFN-γ and ER stress converge on DP5/Hrk activation: a novel mechanism for pancreatic β-cell apoptosis. Cell Death Differ 2009; 11: 1539–1550. 129. Dong M et al. Anti-proliferative and apoptosis-inducible activity of Sarcodonin G from Sarcodon scabrosus in HeLa cells. Int J Oncol 2009; 1: 201–207. 130. Rosa MDSS et al. Antileishmanial activity of a linalool-rich essential oil from Croton cajucara. Antimicrob Agents Chemother 2003; 6: 1895– 1901. 131. Elson CE et al. Impact of lemongrass oil, an essential oil, on serum cholesterol. Lipids 1989; 8: 677–679. 132. Fitch ME et al. Microbiological screening of mevalonate-suppressive minor plant constituents. J Agric Food Chem 1989; 3: 687–691. 133. Lehmann JM et al. An antidiabetic thiazolidinedione is a high affinity ligand for peroxisome proliferatoractivated receptor γ (PPARγ). J Biol Chem 1995; 22: 12953–12956. 134. Shoff SM et al. Concentrationdependent increase in murine P388 and B16 population doubling time by the acyclic monoterpene geraniol. Cancer Res 1991; 1: 37–42. 135. Takahashi N et al. Dual action of isoprenols from herbal medicines on

© 2014 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 66, pp. 1505–1525

Natasha Beukes et al.

136.

137.

138.

139.

140.

141.

142.

143.

144.

145.

both PPARγ and PPARα in 3T3-L1 adipocytes and HepG2 hepatocytes. Fed Eur Biochem Soc Lett 2002; 2–3: 315–322. Adany I et al. Differences in sensitivity to farnesol toxicity between neoplasticallyand non-neoplastically-derived cells in culture. Cancer Lett 1994; 2: 175–179. Forman BM et al. Identification of a nuclear receptor that is activated by farnesol metabolites. Cell 1995; 5: 687–693. Haug JS et al. Directed cell killing (apoptosis) in human lymphoblastoid cells incubated in the presence of farnesol: effect of phosphatidylcholine. Biochim Biophys Acta 1994; 1: 133–140. Voziyan PA et al. Mechanism of farnesol cytotoxicity: further evidence for the role of PKC-dependent signal transduction in farnesolinduced apoptotic cell death. Biochem Biophys Res Commun 1995; 2: 479–486. Clegg RJ et al. Inhibition of hepatic cholesterol synthesis and S-3hydroxy-3-methylglutaryl-CoA reductase by mono and bicyclic monoterpenes administered in vivo. Biochem Pharmacol 1980; 15: 2125– 2127. Clegg RJ et al. The mechanism of cyclic monoterpene inhibition of hepatic 3-hydroxy-3-methylglutaryl coenzyme A reductase in vivo in the rat. J Biol Chem 1982; 5: 2294– 2299. Qureshi AA et al. Inhibition of hepatic mevalonate biosynthesis by the monoterpene, d-limonene. J Agric Food Chem 1988; 6: 1220– 1224. Santos SRL et al. Structure-activity relationships of larvicidal monoterpenes and derivatives against Aedes aegypti Linn. Chemosphere 2011; 1: 150–153. Samarasekera R et al. Insecticidal activity of menthol derivatives against mosquitoes. Pest Manag Sci 2008; 3: 290–295. Haag JD, Gould MN. Mammary carcinoma regression induced by

Terpenoids and ER stress

146.

147.

148.

149.

150.

151.

152.

153.

154.

155.

156.

perillyl alcohol, a hydroxylated analog of limonene. Cancer Chemother Pharmacol 1994; 6: 477–483. Mills JJ et al. Induction of apoptosis in liver tumors by the monoterpene perillyl alcohol. Cancer Res 1995; 5: 979–983. Bassaganya-Riera J et al. Abscisic acid regulates inflammation via ligand-binding domain-independent activation of peroxisome proliferator-activated receptor γ. J Biol Chem 2011; 4: 2504–2516. Bruzzone S et al. Abscisic acid is an endogenous stimulator of insulin release from human pancreatic islets with cyclic ADP ribose as second messenger. J Biol Chem 2008; 47: 32188–32197. Guri AJ et al. Dietary abscisic acid ameliorates glucose tolerance and obesity-related inflammation in db/db mice fed high-fat diets. Clin Nutr 2007; 1: 107–116. Burrows TL et al. Validation of overweight children’s fruit and vegetable intake using plasma carotenoids. Obesity 2009; 1: 162–168. Kun Y et al. Lycopene: its properties and relationship to human health. Food Rev Int 2006; 4: 309–333. Vivekananthan DP et al. Use of antioxidant vitamins for the prevention of cardiovascular disease: metaanalysis of randomised trials. Lancet 2003; 9374: 2017–2023. Ukil A et al. 18β-glycyrrhetinic acid triggers curative Th1 response and nitric oxide up-regulation in experimental visceral leishmaniasis associated with the activation of NF-κB. J Immunol 2005; 2: 1161–1169. Mnonopi N et al. Marrubiin, a constituent of Leonotis leonurus, alleviates diabetic symptoms. Phytomedicine 2012; 6: 488–493. Kee NLA et al. Antithrombotic/ anticoagulant and anticancer activities of selected medicinal plants from South Africa. Afr J Biotechnol 2008; 3: 217–223. Wolcott CD, Rallison ML. Infancyonset diabetes mellitus and multiple epiphyseal dysplasia. J Pediatr 1972; 2: 292–297.

© 2014 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 66, pp. 1505–1525

157. Oyadomari S et al. Nitric oxideinduced apoptosis in pancreatic β cells is mediated by the endoplasmic reticulum stress pathway. Proc Natl Acad Sci U S A 2001; 19: 10845– 10850. 158. Hartman MG et al. Role for activating transcription factor 3 in stressinduced β-cell apoptosis. Mol Cell Biol 2004; 13: 5721–5732. 159. Butler AE et al. β-cell deficit and increased β-cell apoptosis in humans with type 2 diabetes. Diabetes 2003; 1: 102–110. 160. Ramadan JW et al. The central role of calcium in the effects of cytokines on beta-cell function: Implications for type 1 and type 2 diabetes. Cell Calcium 2011; 6: 481–490. 161. Lipson KL et al. Regulation of insulin biosynthesis in pancreatic beta cells by an endoplasmic reticulumresident protein kinase IRE1. Cell Metab 2006; 3: 245–254. 162. Kharroubi I et al. Free fatty acids and cytokines induce pancreatic β-cell apoptosis by different mechanisms: role of nuclear factor-κB and endoplasmic reticulum stress. Endocrinology 2004; 11: 5087–5096. 163. Delépine M et al. EIF2AK3, encoding translation initiation factor 2-α kinase 3, is mutated in patients with Wolcott-Rallison syndrome. Nat Genet 2000; 4: 406–409. 164. Harding HP et al. Diabetes mellitus and exocrine pancreatic dysfunction in Perk-/- mice reveals a role for translational control in secretory cell survival. Mol Cell 2001; 6: 1153– 1163. 165. Fonseca SG et al. Endoplasmic reticulum stress in β-cells and development of diabetes. Curr Opin Pharmacol 2009; 6: 763–770. 166. Vinik AI et al. Platelet dysfunction in type 2 diabetes. Diabetes Care 2001; 8: 1476–1485. 167. Coetzee C et al. Anticoagulant effects of a Cannabis extract in an obese rat model. Phytomedicine 2007; 5: 333– 337. 168. Levendal RA et al. Cannabis exposure associated with weight reduction and β-cell protection in an obese rat 1523

Terpenoids and ER stress

169.

170.

171.

172.

173.

174.

175.

176.

177.

178.

179.

180.

1524

model. Phytomedicine 2012; 7: 575– 582. Borradaile NM et al. A critical role for eukaryotic elongation factor 1A-1 in lipotoxic cell death. Mol Biol Cell 2006; 2: 770–778. Malhi H, Kaufman RJ. Endoplasmic reticulum stress in liver disease. J Hepatol 2011; 4: 795–809. Özcan U et al. Endoplasmic reticulum stress links obesity, insulin action, and type 2 diabetes. Science 2004; 5695: 457–461. Ozcan L et al. Endoplasmic reticulum stress plays a central role in development of leptin resistance. Cell Metab 2009; 1: 35–51. Dong F, Ren J. Adiponectin improves cardiomyocyte contractile fnction in db/db diabetic obese mice. Obesity 2009; 2: 262–268. Li J et al. Deficiency of Rac1 blocks NADPH oxidase activation, inhibits endoplasmic reticulum stress, and reduces myocardial remodeling in a mouse model of type 1 diabetes. Diabetes 2010; 8: 2033–2042. Martínez de Morentin PB et al. Hypothalamic lipotoxicity and the metabolic syndrome. Biochim Biophys Acta 2010; 3: 350–361. Boden G et al. Increase in endoplasmic reticulum stress-related proteins and genes in adipose tissue of obese, insulin-resistant individuals. Diabetes 2008; 9: 2438– 2444. Sharma NK et al. Endoplasmic reticulum stress markers are associated with obesity in nondiabetic subjects. JCEM 2008; 11: 4532–4541. Tsutsumi A et al. Caloric restriction decreases ER stress in liver and adipose tissue in ob/ob mice. Biochem Biophys Res Commun 2011; 1: 339–344. Özcan U et al. Chemical chaperones reduce ER stress and restore glucose homeostasis in a mouse model of type 2 diabetes. Science 2006; 5790: 1137–1140. Zhang CY et al. Genipin inhibits UCP2-mediated proton leak and

Natasha Beukes et al.

181.

182.

183.

184.

185.

186.

187.

188.

189.

190.

191.

acutely reverses obesity- and high glucose-induced β cell dysfunction in isolated pancreatic islets. Cell Metab 2006; 6: 417–427. Barmada MM et al. A genome scan in 260 inflammatory bowel diseaseaffected relative pairs. Inflamm Bowel Dis 2004; 1: 15–22. Hampe J et al. A genomewide analysis provides evidence for novel linkages in inflammatory bowel disease in a large European cohort. Am J Hum Genet 1999; 3: 808–816. Vermeire S et al. Genome wide scan in a flemish inflammatory bowel disease population: support for the IBD4 locus, population heterogeneity, and epistasis. Gut 2004; 7: 980– 986. Bogaert S et al. Involvement of endoplasmic reticulum stress in inflammatory bowel disease: a different implication for colonic and ileal disease? Public Libr Sci ONE 2011; 6: e25589. Velasco G et al. Towards the use of cannabinoids as antitumour agents. Nat Rev Cancer 2012; 6: 436–444. Caffarel MM et al. Cannabinoids reduce ErbB2-driven breast cancer progression through Akt inhibition. Mol Cancer 2010; 9: 196. Blázquez C et al. Cannabinoid receptors as novel targets for the treatment of melanoma. Fed Am Soc Exp Biol J 2006; 14: E2199–E2208. Gardner B et al. Methanandamide increases COX-2 expression and tumor growth in murine lung cancer. FASEB J 2003; 14: 2157–2159. McKallip RJ et al. Δ-9tetrahydrocannabinol enhances breast cancer growth and metastasis by suppression of the antitumor immune response. J Immunol 2005; 6: 3281–3289. Zhu LX et al. Δ-9tetrahydrocannabinol inhibits antitumor immunity by a CB2 receptor-mediated, cytokinedependent pathway. J Immunol 2000; 1: 373–380. Vara D et al. Anti-tumoral action of cannabinoids on hepatocellular carcinoma: role of AMPK-dependent

192.

193.

194.

195.

196.

197.

198.

199.

200.

201.

202.

activation of autophagy. Cell Death Differ 2011; 7: 1099–1111. Shrivastava A et al. Cannabidiol induces programmed cell death in breast cancer cells by coordinating the cross-talk between apoptosis and autophagy. Mol Cancer Ther 2011; 7: 1161–1172. Shanmugam MK et al. Targeted inhibition of tumor proliferation, survival, and metastasis by pentacyclic triterpenoids: potential role in prevention and therapy of cancer. Cancer Lett 2012; 2: 158–170. Paschen W, Frandsen A. Endoplasmic reticulum dysfunction – a common denominator for cell injury in acute and degenerative diseases of the brain? J Neurochem 2001; 4: 719–725. Yamamoto A et al. Endoplasmic reticulum stress and apoptosis signaling in human temporal lobe epilepsy. J Neuropathol Exp Neurol 2006; 3: 217–225. Lindholm D et al. ER stress and neurodegenerative diseases. Cell Death Differ 2006; 3: 385–392. Weng WC et al. Role of glucoseregulated protein 78 in embryonic development and neurological disorders. J Formos Med Assoc 2011; 7: 428–437. Sokka AL et al. Endoplasmic reticulum stress inhibition protects against excitotoxic neuronal injury in the rat brain. J Neurosci 2007; 4: 901– 908. Mercado G et al. An ERcentric view of Parkinson’s disease. Trends Mol Med 2013; 3: 165–175. Hetz C et al. XBP-1 deficiency in the nervous system protects against amyotrophic lateral sclerosis by increasing autophagy. Genes Dev 2009; 19: 2294–2306. Liu GL et al. Inositol-requiring protein 1α signaling pathway is activated in the temporal cortex of patients with mesial temporal lobe epilepsy. Neurol Sci 2013; 35: 357–364. Liu G et al. Involvement of IRE1α signaling in the hippocampus in patients with mesial temporal lobe

© 2014 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 66, pp. 1505–1525

Natasha Beukes et al.

203.

204.

205.

206.

epilepsy. Brain Res Bull 2011; 1: 94–102. Henshall DC, Simon RP. Epilepsy and apoptosis pathways. J Cereb Blood Flow Metab 2005; 12: 1557– 1572. Yu N et al. A meta-analysis of pro-inflammatory cytokines in the plasma of epileptic patients with recent seizure. Neurosci Lett 2012; 1: 110–115. Anand SS, Babu PP. Endoplasmic reticulum stress and neurodegeneration in experimental cerebral malaria. Neurosignals 2013; 21: 99– 111. de Souza JB, Riley EM. Cerebral malaria: the contribution of studies

Terpenoids and ER stress

in animal models to our understanding of immunopathogenesis. Microbes Infect 2002; 3: 291–300. 207. van der Heyde HC et al. A unified hypothesis for the genesis of cerebral malaria: sequestration, inflammation and hemostasis leading to microcirculatory dysfunction. Trends Parasitol 2006; 11: 503–508. 208. Tardif KD et al. Hepatitis C virus suppresses the IRE1-XBP1 pathway of the unfolded protein response. J Biol Chem 2004; 17: 17158–17164. 209. Wati S et al. Dengue virus infection induces upregulation of GRP78, which acts to chaperone viral antigen production. J Virol 2009; 24: 12871– 12880.

© 2014 Royal Pharmaceutical Society, Journal of Pharmacy and Pharmacology, 66, pp. 1505–1525

210. Yu CY et al. Flavivirus infection activates the XBP1 pathway of the unfolded protein response to cope with endoplasmic reticulum stress. J Virol 2006; 23: 11868–11880. 211. Medigeshi GR et al. West Nile virus infection activates the unfolded protein response, leading to CHOP induction and apoptosis. J Virol 2007; 20: 10849–10860. 212. Ambrose RL, Mackenzie JM. West Nile virus differentially modulates the unfolded protein response to facilitate replication and immune evasion. J Virol 2011; 6: 2723–2732.

1525

Selected terpenoids from medicinal plants modulate endoplasmic reticulum stress in metabolic disorders.

The majority of research performed on cellular stress and apoptosis focuses on mitochondrial dysfunction; however, the importance of the endoplasmic r...
2MB Sizes 0 Downloads 3 Views