You are on page 1of 14

Biomedicine & Pharmacotherapy 84 (2016) 569–582

Available online at

ScienceDirect
www.sciencedirect.com

Relevance of carnosic acid to the treatment of several health disorders:


Molecular targets and mechanisms
Sana Bahria,b,* , Saloua Jameleddinea , Vadim Shlyonskyb
a
Laboratory of Physiology, Faculty of Medicine of Tunis, University of Tunis El Manar, La Rabta 1007, Tunis, Tunisia
b
Laboratory of Physiopathology and Pharmacology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium

A R T I C L E I N F O A B S T R A C T

Article history:
Received 26 June 2016 Carnosic acid is a phenolic diterperne compound found in abundance in sage and rosemary, which are
Received in revised form 29 August 2016 both widely used in traditional medicine. Research over the past decade indicates that carnosic acid has
Accepted 18 September 2016 multiple bioactive properties including antioxidant, anti-inflammatory and anticancer activities among
others. This review summarizes the current in vitro and in vivo data about the efficacy of carnosic acid in
Keywords: the prevention or treatment of various experimental health disorders. The analysis of the literature
Carnosic acid allows an insight into the participation of numerous signaling pathways modulated by carnosic acid, into
Antioxidant its synergistic potential and, thus, into the divergence in cellular mechanisms of action of this molecule.
Anti-inflammatory
ã 2016 Elsevier Masson SAS. All rights reserved.
Antitumor

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 570
2. Methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 570
3. Bioavailability and toxicity of carnosic acid . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 570
4. Anti-cancer effect of carnosic acid . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 570
4.1. Carnosic acid and colorectal cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 570
4.2. Carnosic acid and liver cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 571
4.3. Carnosic acid and renal cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 571
4.4. Carnosic acid and brain cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 572
4.5. Carnosic acid and leukemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 572
4.6. Others anticancer properties of carnosic acid . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 573
5. Carnosic acid and experimental disorders . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 574
5.1. Carnosic acid and liver injuries . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 574
5.2. Carnosic acid and cardiac injuries . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 574
5.3. Carnosic acid and atherosclerosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 574
5.4. Carnosic acid and brain injuries . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 574
5.4.1. Carnosic acid and Alzheimer disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 574
5.4.2. Carnosic acid and Parkinson disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 575
5.4.3. Carnosic acid and retinal degeneration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 575
5.4.4. Carnosic acid protects from other neuronal agressions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 575
5.5. Canosic acid and obesity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 576
5.6. Carnosic acid and gastric lesions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 577
5.7. Carnosic acid and skin lesions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 577
5.8. Other possible therapeutic applications of carnosic acid . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 577
6. Carnosic acid synergizes with other natural compounds . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 577
7. Rosmarinic acid and carnosic acid: possible connection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 578
8. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 578

* Corresponding author at: Laboratory of Physiology, Faculty of Medicine of Tunis, University of Tunis El Manar, La Rabta 1007, Tunis, Tunisia.
E-mail address: bahrisana88@gmail.com (S. Bahri).

http://dx.doi.org/10.1016/j.biopha.2016.09.067
0753-3322/ã 2016 Elsevier Masson SAS. All rights reserved.
570 S. Bahri et al. / Biomedicine & Pharmacotherapy 84 (2016) 569–582

Conflicts of interest . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 579


Ethical responsibilities protection of people and animals . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 579
Confidentiality of data . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 579
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 579

1. Introduction that this molecule exerts a dual protective role in rosemary plant
against environmental constraints by capturing free radicals in
Currently, research on bioactive substances of plant or animal chloroplasts and by preserving the stability of cell membranes [4].
origin earned a growing interest in numerous industrial sectors, Detection of the anticancer activity of CA was among the first
including cosmetics, pharmaceutics and food processing. Notably, research work carried out. CA exerted an antiproliferative action in
plants are rich source of biomolecules generally well tolerated by leukemic cells without induction of apoptotic or necrotic cell death
the human body. Many of these biomolecules served as natural [5]. This study demonstrated that CA could cooperate synergisti-
remedies since ancient times. The enormous technical progress, cally with other natural anticancer compounds, such as vitamin D
discoveries and innovations, which promoted industrialization, and retinoic acid metabolites, to potentiate anticancer effects.
helped pharmaceutical companies to identify natural substances Later on, consequent studies of CA in the field of oncology took a
obtained from plants and to determine their therapeutic potential. considerable place both in vivo and in vitro. However, studies in
The term “medicinal plant” characterizes species that contain other health areas demonstrate that CA is clearly promiscuous
active compounds, which belong mainly to the category of molecule that is useful in the treatment of many other human
secondary metabolites (polyphenols, essential oils) known for diseases.
their medicinal properties. However, caution should be taken as to In this review, we will focus on the biological effects of
what part of the plant to use, since they can differ in characteristics vegetable extracts rich in CA or pure CA that were demonstrated by
and contain more than one active compound in different different research teams. We will assess efficacy of CA in various
proportions. Therefore, plant usage requires a real knowledge to physiological assays and clinical trials published till now.
avoid any risk of poisoning.
Carnosic acid (CA) was discovered first by Linde in Salvia 2. Methods
officinalis L. [1], then by Wenkert et al. [2] in Rosmarinus officinalis L
leaves. Despite the fact that sage and rosemary are hailed since In order to explore available information about CA beneficial
ancient times for their therapeutic properties, the exploration of effects for the treatment/prevention of various diseases, we have
the mechanisms of CA action began only in the early 2000s. Over collected data published between August 2001 and April 2016 in
the last decade, several research teams studied the pharmacologi- the Pubmed database using the following appropriate combina-
cal properties of CA, demonstrating that this molecule may have tions of MeSH (Medical Subject Heading) terms: carnosic acid,
clinical applications for various human diseases. Indeed, studies carnosic acid antioxidant, carnosic acid cancer, carnosic acid brain,
using rosemary extract showed that its properties are closely carnosic acid obesity, carnosic acid breast, carnosic acid liver,
related to its phenolic constituents, especially the most abundant rosemary carnosic acid.
compounds, carnosic acid and rosmarinic acid. On the other hand,
consumption of the whole plant leads to the ingestion of products 3. Bioavailability and toxicity of carnosic acid
other than the active ingredient sought. This requires the
extraction and purification of the active compound in order to At present, there are only two studies on bioavailability of
reveal its effects, to know the dose administered and to avoid the carnosic acid. First, it was reported that 6 h after oral administra-
risk of overdose that can lead to toxicity. tion in rats (64.3  5.8 mg/kg), the bioavailability of CA in its free
Carnosic acid (C20H28O4, Fig. 1), a phenolic diterpene that form was 40.1%, and its excretion in the feces after 24 h was
belongs to the terpene class of secondary metabolites [3], is 15.6  8.2% [6]. CA is absorbed into the bloodstream after oral
localized in rosemary leaves, more precisely in chloroplasts of administration and its traces were found in the rat intestine, liver
trichome cells. The stability of this molecule remains discussed and muscle tissue of abdomen and legs [6]. In another study, Romo
because it can give rise even in planta to several dehydrogenation Vaquero et al. [7] also investigated the bioavailability of CA and
derivatives such as carnosol, rosmanol and isorosmanol. In plants other diterpenoids found in rosemary extract and detected all the
subjected to intense solar radiation and high drought, CA may be metabolites 25 min after oral administration in rats. CA was
transformed into methyl derivatives. This clearly demonstrates pronouncedly present in the intestine, liver, and plasma with some
quantities of CA and its metabolites even in the brain, translating
their potential health benefits in these tissues.
Wang et al. [8] evaluated the acute and 30-day oral toxicity of
CA on Wistar rats and defined acute oral lethal dose (LD50) in the
range of 7100 mg/kg of body weight. In rats treated chronically
with a high-dose of CA, they observed slight reduction in the body
weight gain compared to control group and revealed only weak
pathological changes in the heart, liver, and kidney. These
observations reflect low toxicity profile of the molecule.

4. Anti-cancer effect of carnosic acid

4.1. Carnosic acid and colorectal cancer

The relevance of the current studies to the oncology is based on


the inhibition of the in vitro viability of different cell lines
Fig 1. Carnosic acid.
S. Bahri et al. / Biomedicine & Pharmacotherapy 84 (2016) 569–582 571

originated from colon cancer. Barni et al. [9] initially studied CA death in the same cell type? Common observation in these above
effect on 3 types of cultured colorectal cancer cells: Caco-2, HT29 studies is that CA induced HepG2 cell death by Akt inhibition. This
and LoVo. They revealed a dose-dependent reduction in the leads us to conclude that Akt signaling suppression is implicated
viability of these cells treated with CA with IC50 values within the both in apoptosis and autophagy in these cells. Most likely, slight
range of 24–96 mM. Cellular death followed the induction of culture condition differences including different serum might
apoptosis and inhibition of cell migration and adhesion. This effect trigger HepG2 cells to undergo autophagy instead of apoptosis in
was accompanied by a downregulation of the expression of COX-2 the presence of CA. Arico et al. [15] demonstrated that PI3 kinase/
and by reduction of the activity of secreted proteases such as Akt pathway activation, which is well known to inhibit apoptosis,
urokinase plasminogen activator and metalloproteinases [9]. Kim could also inhibit autophagy in human colon cancer HT-29 cells. In
et al. [10] showed a decrease in the viability of human colon cancer addition, mTOR/Akt signaling pathway, death-associated protein
HCT116 cells treated with CA (20 to 100 mM). It triggered HCT116 kinase (DAPK) and p53 activation were described to participate in
cells apoptosis via ROS generation, p53 and Bax induction, caspase both processes [16].
3–9 activation, PARP cleavage and STAT3 signaling pathway The progress in molecular research on cell death demonstrates
inhibition. The same Korean team demonstrated that CA could that autophagy and apoptosis have a strong molecular conver-
slow down the rapidity of colon cancer formation associated with gence. The discovery of the autophagy related genes (ATG) in
adiposity. In fact, Kim et al. [11] discovered that treatment with CA Saccharomyces cerevisiae, whichare mostly conserved from yeast to
at 1 and 10 mM for 48 h attenuated the proliferation of HT-29 colon human, has allowed a clarification of the mechanism controlling
adenocarcinoma cells co-cultured with 3T3-L1 adipocytes by this process. ATG genes have been identified to manage an orderly
inducing cell cycle arrest and apoptosis. The in vitro molecular process leading to the induction of autophagic vesicles, their
mechanism involved the attenuation of leptin receptor signaling, nucleation, completion, lysosomal fusion and finally their
including downstreaminhibition of Akt and ERK phosphorylation. recycling [17]. Several studies indicate that two types of ATG
Then, Kim and colleagues tested the effect of carnosic acid in a can interact with Bcl-2 member family, FADD and induce
mice model of colitis induced by treatment with dextran sodium activation of caspases. In fact, ATG-5 can participate in the
sulfate as well as in a model of colon cancer induced by formation of autophagosomes involving FADD death domain
azoxymethane. Compared to untreated animals, mice treated with binding [18]. Furthermore, ATG-12 can provoke Bcl-2 inhibition
CA have shown a reduction in the number of colon tumors and a and caspase activation [19–20,21]. Finally, Pattingre et al. [22]
decrease in circulating concentrations of leptin, adiponectin, demonstrated the close crosstalk between beclin-1 and Bcl-2 in
insulin, and insulin-like growth factor 1. In addition, the expression prevention of autophagy initiation. These examples provide the
of leptin and insulin receptors, phosphorylated Akt and ERK, B-cell evidence that some autophagy regulators have the ability to affect
lymphoma extra large (Bcl-xL), and cyclinD1 protein in the colon both apoptosis and autophagy and prove the clear connection
were attenuated by CA treatment [11]. between the machineries of these two processes. In some cases,
With regard to rosemary leaves extract, Borrás-Linares et al. cell death in the presence of CA might be triggered by
[12] reported its antiproliferative action on a group of human colon simultaneous activation of both phenomenon involving Akt
cancer cell lines. The characterization and quantification of various inhibition. This reinforces effectiveness of CA as an anticancer
phenolic compounds in this extract by HPLC-ESI-QTOF-MS allowed agent (Fig. 2).
authors to point out the contribution of CA among other On the other hand, pretreatment with CA can protect and
compounds identified. Thus, they estimated IC50 value for carnosic counteract the toxic effect of some carcinogenic agents. Costa et al.
acid as 8.1 mM for SW 480 cells and 20.4 mM for HT29 cells. [23] demonstrated a dose-dependent protective effect of carnosic
In summary, all these findings cited above suggest that CA acid pretreatment on cell toxicity induced by aflatoxin B1 in HepG2
might have clinical applications for the treatment of the colorectal cells. When cells were pre-treated with relatively low doses of CA
cancer, considered as the third most frequent malignant tumor (20 or 30 mM) for 24 h prior to aflatoxin, it decreased reactive
around the world [9]. oxygen species level and cell death. This study highlighted the
preventive effect of this molecule, which can play a dual role as a
4.2. Carnosic acid and liver cancer preventive and curative herbal drug against some type of cancer.

The exploration of CA effect for the treatment of liver cancer is 4.3. Carnosic acid and renal cancer
not complete. Yet, current data clearly confirm the anticancer
activity of CA on tumor cell lines originated from rat or human liver The studies of CA effect on human renal cancer started only in
tumors. For example, Gao et al. [13] reported that CA caused an the last two years by the Korean researchers Min and Jung [24].
autophagic cell death in human hepatoma cells (HepG2) but not Actual data are still limited to the detection of the in vitro effects in
apoptosis, the latter being the major mechanism exploited by this human renal carcinoma Caki cells, but remain encouraging and
herbal molecule in other tumor cell lines. Data of this study need to be continued.
showed time and dose-dependent (within the range of 20– CA induces apoptosis in human renal carcinoma Caki cells
100 mM) formation of autophagic vacuoles with an increase in the incubated with 20, 40 and 60 mM for 24 h via caspase activation
ratio of cytosolic microtubule-associated protein 1A/1B-light chain associated with poly (ADP-ribose) polymerase (PARP) cleavage,
3 (LC3-I) and its conjugated form LC3-II with phosphatidyletha- intracellular ROS production and endoplasmic reticulum (ER)
nolamine, used to detect autophagy-related processes. Further- stress [24]. The induction of ER stress marker proteins expression,
more, CA exerts this autophagic induction through the inhibition of the activation of transcription factor 4 (ATF4) and CCAAT/
Akt/mTOR pathway [13]. enhancer-binding protein-homologous protein (CHOP), known
In contrast, Xiang et al. [14] demonstrated that CA reduced cell as a major ER stress-mediated pro-apoptotic transcription factor,
viability of HepG2 cells via induction of apoptosis in a dose- were also detected following CA treatment in a dose- and time-
dependent manner (5–100 mM),causing a rapid caspase-3 activa- dependent manner.
tion and a proteolytic cleavage of poly (ADP-ribose) polymerase Jung et al. [25] demonstrated that CA at 10 or 20 mM induced
(PARP) accompanied by a mitochondrial dysfunction and deacti- considerably Tumor necrosis factor-related apoptosis-inducing
vation of Akt. Therefore, we should ask a question: how can one ligand (TRAIL)-mediated apoptosis in human renal carcinoma Caki
explain the fact that CA can activate two different ways of cell cells after 24 h of incubation through caspase-3- 8 and -9
572 S. Bahri et al. / Biomedicine & Pharmacotherapy 84 (2016) 569–582

Fig. 2. Possible connection between autophagy and apoptosis causing HepG2 cell death induced by carnosic acid.
Carnosic acid (CA)promotes the formation of autophagic vacuoles via ATG activation. The latter (probably ATG-12) can induce caspase activation and Bcl2 inhibition both
implicated in apoptosis. CA can also dissipate mitochondrial membrane potential provoking cytochrome c (cyt c) release, nuclear cleavage of PARP and BAX/Bcl 2 activation to
ensure apoptosis. These two processes implicate down regulation of AKT and may involve other molecular pathways separately to induce HepG2 cell death.

activation.TRAIL is known by its binding to death receptors (DR4 associated with caspase-3/-9 and PARP activation and accompa-
and DR5),whose activation leads to apoptosis in various cancer nied by ROS generation and a down-regulation of anti-apoptotic
cells. The investigation of the molecular targets and apoptosis- protein Bcl-2. The molecular investigation showed an increase of
related protein expression showed that CA at 20 mM sensitizes p38 and JNK activation and a decrease in ERK activation [28]. We
TRAIL-mediated apoptosis through the down-regulation of anti- conclude that further experimental studies are needed to
apoptotic proteins such as cellular FLICE-inhibitory protein (c-FLIP) determine the anticarcinogenic effect of CA on several types of
and Bcl-2. This effect includes also an up-regulation of ER stress- brain cancer.
mediated death receptor5 (DR5), and up-regulation of the pro-
apoptotic Bcl-2 family proteins Bim and PUMA expression at the 4.5. Carnosic acid and leukemia
transcriptional levels.
All these results demonstrate clearly that CA could be a potent Leukemia is a malignant progressive tumor characterized by
candidate for the treatment of renal cancer. An association with the formation of immature or abnormal leukocytes by the bone
other cell ligands like TRAIL can improve its action. Indeed, marrow and other blood-forming organs leading to the deletion of
combined action of CA and TRAIL can inhibit Caki cells viability normal blood cells generation. The protective effect of CA against
that is associated with chromatin damage in the nuclei and in leukemia was first described by Steiner et al. [5] in HL-60 and U937
cytoplasmic histone-associated DNA fragments [24]. CA can human myeloid leukemia cells. This effect was explained by the
reduce the mitochondrial membrane potential and induce inhibition of cell proliferation by cell cycle arrest in the G1 phase
cytochrome-crelease by the activation of the pro-apoptotic with an IC 50 in the range of 6–7 mM without inducing apoptotic or
protein (Bax) [24]. necrotic cell death. Cell cycle arrest was evidenced from the
increase in the levels of the universal cyclin-dependent kinase
4.4. Carnosic acid and brain cancer inhibitors p21WAFI and p27Kipl [5]. This anticarcinogenic activity
could be enhanced synergistically by 1,25-dihydroxyvitamin D3
Studies of CA effect on human brain cancer remain scarce. The and by all-trans retinoic acid(ATRA) at low concentration (1 nM).
first study on brain tumor cells has been performed by Kosaka et al. CA at 10 mM cooperated with these two compounds to potentiate
[26], but the aim of this study was not to determine the anticancer their differentiating effects in human myeloid leukemia cells
effect of CA, but rather to identify its role as a stimulator of nerve [5,29]. To detect the mechanisms involved in this differentiation
growth factor (NGF) synthesis in human glioblastoma multiforme enhancement, especially in sublime HL60G, Danilenko et al. [30]
T98G cells. De Olivera et al. [27] took advantage of SH-SY5Y demonstrated that this monocyte differentiation stimulation was
neuroblastoma cell model to determine the protective effect of CA associated with an increase in monocytic serine esterase and
(1 mM for 12 h) against methylglyoxal cytotoxicity, found in CD11b expression. The latter is a member of integrin family
abundance in samples obtained from patients suffering Alzheimer expressed on the surface of many leukocytes that regulates their
disease. The same research team studied the effect of CA in adhesion and migration and contributes to the inflammatory
paraquat model of Parkinson's disease using these cells, demon- process. In addition, this combinatory effect of CA with 1a,25(OH)
strating that this molecule can play a potential neuroprotective 2D3 and ATRA decreased cell proliferation and cell cycle transition
role and can be used in some cases of Parkinson's disease [69]. The from G1 to S phase but increased Nuclear vitamin D receptor (VDR)
antitumor action of CA in brain cancer was reported in vitro by Tsai and Retinoid X receptor a (RXR-a) expression. Separately, CA at
et al. [28]. It has been demonstratedthat CA can decrease the 10 mM and 1a,25(OH)2D3 at 1 nM induced a decrease in the ROS
viability of human neuroblastoma IMR-32 cells in a dose- intracellular level in HL60G cells. Added together, these molecules
dependent manner (in the range of 5–40 mM). It induced apoptosis ameliorated the antioxidant status, increased intracellular
S. Bahri et al. / Biomedicine & Pharmacotherapy 84 (2016) 569–582 573

glutathione level andpotentiated monocytic differentiation via reversing activity and to identify these properties in various cancer
mitogen-activated protein kinase pathway. cell lines.
The effect of another analogue of vitamin D (1,25D(3)) was
investigated in vivo [31]. These authors demonstrated the 4.6. Others anticancer properties of carnosic acid
antileukemic efficacy of CA when combined with the hormonal
form of vitamin D 1,25D(3) analogue, 1,25-dihydroxy-21(3- CA has other therapeutic virtues by killing or reducing the
hydroxy-3-methyl-butyl)-19-nor-cholecalciferol, in a mouse mod- growth of various cancer cell lines and by the treatment of
el of acute myeloid leukemia. This team has performed a angiogenesis-related malignancies. Indeed, angiogenesisis is a
subsequent study and focused on the combined effect of CA with physiological process of neovascularisation from the pre-existing
other plant polyphenols such as curcumin. They demonstrated that blood vessels occurring particularly during embryonic develop-
these two polyphenols produced a synergistic antiproliferative ment. This process appears to be also important for the growth of
effect caused by an important apoptotic cell death in HL-60 and malignant tumors and metastasis. Then an anti-angiogenesis
KG-1a human acute myeloid leukemia cells, accompanied by treatment can prevent the formation of new blood vessels and
caspase 3–8 and-9 activation and this involved both extrinsic and block the progression and the growth of tumors. CA at 10 mM and
intrinsic apoptotic pathways. Curcumin at 5 mM and CA at 10 mM 100 mM has been described to have an anti-angiogenic activity by
can induce the activation of the proapoptotic protein Bid without inhibiting differentiation, proliferation, migration and proteolytic
increasing ROS level or altering Bcl-2 family protein expression capability of endothelial cells and by exerting an antiproliferative
[32]. effect in endothelial and tumor cells through induction of
On the other hand, this apoptotic response to curcumin and CA apoptosis [37]. The use of chicken embryo chorioallantoic
in acute myeloid leukemia cells is associated with an elevation in membrane assay as a model for studying neovascularisation
the cytosolic level of Ca2+ and with the inhibition of its extrusion confirmed the inhibition of both in vitro and in vivo angiogenesis
through the plasma membrane [33]. This Ca2+ release from process described above. CA was described also to suppress
intracellular stores was found to be responsible for caspase cascade angiogenesis in models using human umbilical vein endothelial
activation. Interestingly, this combined treatment did not modify cells and to inhibit microvessel outgrowth on ex vivo angiogenesis
Ca2+ homeostasis and viability in non-neoplastic hematopoietic test using rat aortic rings in a dose-dependent manner (10–
cells, indicating cancer-selective property. 100 mM) [38]. This activity may contribute to the prevention of
CA also synergized with arsenic trioxide (As2O3), used as a drug angiogenesis-related disorders including cancer and neurodegen-
for cancer treatment, on HL-60 human myeloid leukemia cells that erative diseases.
has been described by Wang et al. [34]. Synergic decrease in cell Melanoma is the most serious skin cancer type that develops
viability, the induction of G1 arrest and apoptosis associated with from melanocytes that provide a pigment for skin coloration
the modulation of PTEN/Akt signaling pathway was observed in named melanin.CAwas shown to prevent murine B16-F10 mela-
the presence of both CA at 15 mM and As2O3 at 4 mM. Considering noma cells adhesion and migration at concentrations ranging from
all these results, we deduced that CA has a potent anti-tumor effect 2.5 to 10 mM via the suppression of the mesenchymal marker
against different cell lines of myeloid leukemia. This activity can be expression and via induction of epithelial markers leading to the
boosted by other anticancer compounds, especially by plant- inhibition of the epithelial-mesenchymal transition (EMT) with an
derived polyphenols. inhibition of AKT phosphorylation among other signaling path-
An important feature of cancer cells is the excessive production ways involved [39].
of P-glycoprotein that has the ability to extrude anticarcinogenic CA could be proposed and taken into consideration for the
drugs from the cell providing it a multi-drug resistance (MDR) prevention and/or for the treatment of prostate cancer, but actual
phenotype. Identification of a natural molecule, capable to reverse results remain insufficient and need to be more elaborated. In fact,
this phenotype in human leukemic cell line K562/A02 was the CA at concentrations of 20–100 mM has anti-proliferative effects
main objective of Yu et al. [35] study. In this case, CA was found on androgen-independent human prostate cancer PC-3 cells and in
capable to enhance the cytotoxicity of adriamycin, a chemotherapy androgen refractory prostate cancer DU145 cells caused by
drug commonly used in cancer treatment, by increasing its apoptosis that implicates cytochrome-c release and an increase
intracellular concentration. This reflected in a decrease of the IC50 in Bax/Bcl-2 ratio [40]. In PC-3 cells, these effects involve both
from 16.3  1.6 mg/l when cells are treated with adriamycin alone intrinsic and extrinsic apoptotic pathways as well as suppression of
to 1.4  0.2 mg/l when cells are treated with adriamycin and CA. CA the expression of inhibitor of apoptosis (IAP) family of proteins via
was found to be able to decrease P-glycoprotein level and to the activation of serine/threonine protein phosphatase 2A (PP2A)
suppress MDRL gene expression at the mRNA level. These results and the modulation of Akt/IKK/NF-kB pathway. In DU145 cells, CA
highlight the reversing effect of CA against multidrug resistance induced apoptosis via the intrinsic pathway associated with PARP
mediated by P-glycoprotein in human leukemic cells. cleavage. This study remains the only one that investigated the
Recent study by Wang et al. [36] of CA effect on NOD/SCID acute efficacy of CA to treat human prostate carcinoma. Other studies are
myeloid leukemia mouse model established by K562/A02 cells required to better understand its mechanism.
inoculation has been performed to verify if CA combined with CA was described to exert higher antiproliferative activity
adriamycin can inhibit cancer growth in vivo. Data demonstrated compared to the effect of six different extracts of Rosmarinus
that mice treated with CA (1%, v/v) and adriamycin (3 intraperito- officinalis and rosmarinic acid in two human breast adenocarcino-
neal injection of 1 mg/kg every two days) contained a lower ma cell lines, MCF-7 and MDA-MB-231 [41]. In addition, CA
number of leukemia cells than untreated mice and a higher level of suppressed proliferation, induced G1 cell cycle arrest and
apoptotic cells than in adriamycin-alone treated mice, which displayed significant growth inhibitory activity when associated
demonstrates that CA has an important inhibitory activity on the with curcumin in ER-negative human breast cancer cells [42]. In
malignant cells growth in vivo. fact, it was reported that treatment of these cells with CA for 6 h at
Taken together, current data reveal the antitumor property of low concentration (15 mM) activated the genes involved in
CA in leukemic cell lines, which can be summarized in two major glutathione biosynthesis and transport. At a higher dose
actions: 1) its ability to cooperate with other anticancer agents and (20 mM), CA activated the expression of antioxidant and apoptosis
2) its reversing effect on MDR phenotype. Others studies are genes and suppressed the expression of inhibitors of transcription
required to determine if this cooperation can ameliorate CA MDR and cell cycle genes.
574 S. Bahri et al. / Biomedicine & Pharmacotherapy 84 (2016) 569–582

Taken together, these results are a good start on the way to the levels of several markers of myocardial infarction and lesion,
broad use of CA in the prevention or eradication of several forms of including troponin, myocardial creatine kinase (CK-MB), lactate
cancer. Generally, this molecule appears to exert its antitumor dehydrogenase (LDH), aspartate-aminotransferase (ASAT) and
action by inhibiting cell viability via induction of apoptosis. alanin-aminotransferase (ALAT). CA pretreatment at 50 or
However, most of the studies did not go beyond the in vitro 100 mg/kg can attenuate all these alterations; it also prevented
experimentation. This constitutes the biggest limitation. Further cardiomyocyte apoptosis and decreased myocardial lipid peroxi-
research and preclinical studies are required to fully elucidate and dation and protein oxidation induced by isoproterenol. This occurs
to understand the potential of CA as a preventive or therapeutic via the enhancement of nuclear translocation of Nrf2 and via an
agent against various human carcinomas. up-regulation of the phase II/antioxidant enzyme activities [50].
Recently, Kocak et al. [51] confirmed all these biochemical results
5. Carnosic acid and experimental disorders and reported that in addition to the increase of Nrf2 expression, CA
at 50 mg/kg reduced TNF-a levels, NF-kB, p38 MAPK, and
A number of studies performed over the last decade up to date phosphorylated-JNK 1/2 expressions, and enhanced phosphory-
confirm that CA has beneficial effects in the majority of models of lated extracellular signal-regulated kinase (pERK 1/2). In conclu-
experimental diseases. In this section, we will recapitulate and sion, the reduction of myocardial impairment and apoptosis is
discuss the different data published in this regard. mediated by the antioxidant and anti-inflammatory action of CA.

5.1. Carnosic acid and liver injuries 5.3. Carnosic acid and atherosclerosis

Examination of CA beneficial effect in liver lesions induced by Atherosclerosis is very often accompanied by lipid deposition
various toxic agents advanced in the last three years. They (cholesterol or LDL) on the inner wall of the arteries. Over the years,
highlight specifically its antioxidant and anti-inflammatory deposits progressively impregnate with fibrinogen, platelets, blood
activities in several experimental studies. cells, calcium and solidify, thus blocking the arteries. According to
CA has been studied on lipopolysaccharide [43], ethanol [44] Yu et al. [52], migration and matrix metalloproteinase (MMP)
and acetaminophen [45] induced models of hepatotoxicity in rats. activation in vascular smooth muscle cells could participate in
CA normalized the majority of pathological signs including atherosclerosis evolution and formation. In this regard, they
histological damage, lipid metabolism and oxidative/nitrosative demonstrated that CA at 20 mM inhibited TNF-a-induced migra-
stress by restoring the antioxidant enzymes levels both in serum tion, ROS production, MPP-9 expression, and down regulated
and liver and by reducing the inflammatory cells infiltration in liver nuclear translocation of NF-kB subunits p50 and p65 in human
tissues. Moreover, several in vivo and in vitro reports demonstrated aortic smooth muscle cells. The inhibition of nuclear translocation
that the molecular mechanism implicated in the protective effect of NF-kB subunits p65 and p50 was also reported along the
of CA against liver injuries involves sirtuin-1 (SIRT-1), an NAD- demonstration of CA effect on the expression of cell adhesion
dependent deacetylase that plays an important role in metabolic molecules and on the attachment of monocytes to the endothelium
diseases, such as nonalcoholic fatty liver disease (NAFLD) [46]. For [53]. These authors showed that pretreatment with CA at 20 mM
example, Shan et al. [46] suggested that miR-34a, that targets reduced the U937 cells adhesion to IL-1beta-treated human
SIRT1 expression, might be decreased in response to CA umbilical vein endothelial cells (HUVECs), suppressed ROS
administration in NAFLD rat model at a dose of 60 mg/kg. This production and the expression of cell adhesion molecules
in turn stimulated the SIRT1/p66shc antiapoptotic signaling (ICAM-1, VCAM-1, and E-selectin) via the inhibition of NF-kB.
pathway in rat liver and L02 cells. Tian et al. [44] reported that In conclusion, we have limited experimental data to assess the
10 mM CA pretreatment increased SIRT1 expression following effectiveness of CA in human atherosclerosis. However, the
ethanol exposure in L02 cells, but the p66Shc expression and published studies are encouraging and need to be associated with
mitochondrial translocation were negatively correlated with SIRT1 other clinical trials to better understand this effect on several
expression. Gao et al. [47] demonstrated that CA administered at a pathophysiological signs and clinical manifestations that charac-
dose of 100 mg/kg protected from chronic alcoholic liver injury by terize this disease.
activating the SIRT1/ChREBP and SIRT1/p66shc pathways.
Other in vitro experiments emphasizing the cytotoxicity of 5.4. Carnosic acid and brain injuries
hydrogen peroxide (H2O2) and the protective effect of CA at low
concentration (1 mM), on primary hepatocytes and HepG2 cells 5.4.1. Carnosic acid and Alzheimer disease
have also reported the implication of SIRT1 [48,49]. Indeed, Alzheimer is a degenerative disease that causes progressive
pharmacological inhibition and the use of siRNA mediated SIRT1 decline of cognitive capacity and memory that emerges especially
silencing confirmed the activation of SIRT1 by CA in these two cell due to excessive production and accumulation of beta-amyloid
types. CA was also reported to reverse the increase in ROS level, proteins in some areas of the brain. This forms amyloid plaques
LDH release and the decrease in cell viability produced by H2O2. that are associated with neuronal death and the “distortion” of
There is now enough experimental evidence that CA can attenuate certain structural proteins, called Tau proteins, causing a neurofi-
acute liver lesion and hepatotoxicity especially by alleviating brillary degeneration.a- and g-secretases can counteract beta-
oxidative stress through the SIRT1 pathway. A parallel activation of amyloid formation resulted from the sequential cleavage of its
other signaling pathways may occur depending on the cell type precursor, amyloid precursor protein (APP) by the b- and
and on the molecule inducing liver damage. g-secretases [54]. The pathological mechanism of this disease
implicates also oxidative stress and inflammation.
5.2. Carnosic acid and cardiac injuries Azad et al. [55] reported for the first time the beneficial effect of
CAin experimental rat model of Alzheimer disease. They demon-
Until now, only two published experimental studies focused on strated that carnosic acid at a dose of 3 mg/kg protected
the effect of CA in the context of cardiovascular disorders. They hippocampal neurons from cellular death, especially in the cornu
made use of isoproterenol, a beta-agonist drug, to induce ammonis 1 (CA1) region of the hippocampus, and this in turn
myocardial injury in mice and rats. This drug can induce prevented beta-amyloid-induced neurodegeneration. Rasoolijazi
histopathological lesions and inflammation and to increase serum et al. [56] confirmed these results and explored the effect of CA,
S. Bahri et al. / Biomedicine & Pharmacotherapy 84 (2016) 569–582 575

administered at the same dose of 3 mg/kg, on memory and vivo using cyanide-stimulated non-Swiss albino mouse brain areas
behavioral alteration caused by beta-amyloid. They demonstrated which show several deteriorations similarly to what has been
that this herbal molecule could ameliorate the spatial and learning detected in human brain. Finally, de Olivera et al. [69] investigated
memory deficits. To better understand this mechanism, Meng et al. the role of CA on SH-SY5Y cells treated by paraquat to create an
[57] evaluated the effect of CA at 30 mM on beta-amyloid experimental model of Parkinson disease.
production in SH-SY5Y human neuroblastoma cells and found a The activation of the nuclear factor erythroid-2 related factor 2
reduction of its secretion via tumor necrosis factor-a-converting (Nrf2) plays an important role in the neuroprotective effect of CA in
enzyme activation (a-secretase TACE) without effect on the b-site both in vitro and in vivo models of Parkinsonism induced by several
APP-cleaving enzyme-1 (b-secretase, BACE1). Later, the same neurotoxin [65,68,69]. This effect may be accompanied by the
research team demonstrated that beta-amyloid treatment induced inhibition of JNK and p38 [64–65,70] or by the modulation of PI3K/
SH-SY5Y cells apoptosis by the cleavage of PARP and by the Akt pathways [67,69]. CA exerted also an antioxidant action by
activation of caspase 3–8 and 9, while CA pretreatment at 10 mM suppressing ROS [65] and RNS [66] production, by the increase of
induced a partial inhibition of apoptosis and reduced beta-amyloid total glutathione level (GSH) [65,67] and of glutathione S-
oligomer cellular content [58]. Further studies are required to transferase activity (GST)-P [66], and by the prevention of lipid
elucidate the signaling pathway involved in this protective effect of peroxidation [64]. CA has been described to reverse caspase
CA. However, it is interesting to note that, in SH-SY5Y cells, CA activation [64–65,70] and peroxisome proliferation-activated
pretreatment (1 mM for 12 h) can counteract methylglyoxal receptor gamma (PARPg) cleavage [65,70] induced by several
neurotoxicity by activation of the PI3K/Akt/Nrf2 signaling pathway neurotoxins.
and by up-regulation of Nrf2-dependent antioxidant enzymes [59]. All these results suggest that CA treatment might be a
In this experiment, methylglyoxal was used as a potent inducer of promising management of Parkinson's disease.
glycation. Methylglyoxal seems to be a key molecule in case of
neurodegenerative diseases such as Alzheimer, because samples 5.4.3. Carnosic acid and retinal degeneration
obtained from subjects suffering Alzheimer contain high amount CA may have clinical applications for different diseases
of methylglyoxal. However, it remains important to clarify whether affecting the outer retina. Rezaie et al. [71] demonstrated that in
CA neuroprotection against beta-amyloid neurotoxicity directly retina-derived cell lines ARPE-19 and 661W treated with hydrogen
inhibits glycation or whether it requires the participation of other peroxide, CA at 10 mM triggered the production of antioxidant-
signaling molecules. phase 2 enzymes and was found to reduce the formation of
hyperoxidized peroxiredoxin (Prx2). CA was tested then in rat
5.4.2. Carnosic acid and Parkinson disease model of light-induced retinal degeneration. Compared to
The development and characterization of relevant in vitro/in untreated animals, rats that were given CA (25 mg/kg/day, for
vivo models of Parkinson disease using several neurotoxins seems 6 days) had a significant improvement in outer nuclear layer
to be a practical tool to study the capacity of certain drugs and food thickness and a better electroretinography activity. This study
constituent to counteract or prevent many deleterious effects confirmed that CA might have clinical application to protect the
accompanying this disease. These neurotoxins include dieldrin, an retina and other areas of the brain from degenerative disorders,
organochlorine insecticide involved in sporadic Parkinson's including age-related macular degeneration and retinitis pigmen-
disease development [60]; 6-hydroxydopamine (6-OHDA), a tosa. The latter is a hereditary retinal disease with progressive
neurotoxic organic compound used to kill selectively dopaminer- degeneration of photoreceptors (produced following rod-specific
gic and noradrenergicneurons that constitutes the most commonly gene mutations) leading to partial or complete vision loss.
used drug for the elaboration of animal model of Parkinson's Recently, Kang et al. [72] reported that CA preserved photo-
disease [61]; cyanide, a toxic chemical compound inducing receptors morphology and function in the Pde6b (rd10) mouse
delayed-onset neurological syndrome with the symptoms of model of retinal pigmentosa, through inhibition of oxidative stress,
Parkinsonism [62]; and paraquat, a toxic herbicide used for the endoplasmic reticulum stress and inflammation, and by the
establishment of Parkinson experimental model [63]. All these regulation of multiple cellular pathways in mice including an
neurotoxins were used by various research teams worldwide to upregulation of Nrf2 expression and a downregulation of p-JNK
provide new insights on different mechanisms involved in CA and p-p38 expression among others. These data provide the
induced neuroprotection. evidence that CA could exert neuroprotective effects by defending
To evaluate the effect of various neurotoxins and create an in brain cells from oxidative stress, known by its implication in the
vitro/in vivo model of Parkinson disease, several neuronal cells progression of several retinal diseases.
types/animal models were used. For example, Park et al. [64]
examined the effect of CA pretreatment (at 5 or 10 mM) on dieldrin 5.4.4. Carnosic acid protects from other neuronal agressions
induced-neurotoxicity in cultured dopaminergic cells (SN4741) CA is a catechol ring containing compounds that can be
and showed a dose-dependent protective effect. Chen et al. [65] converted by oxidation to an electrophilic quinone, characterized
demonstrated that CA attenuated SH-SY5Y cell death induced by 6- by its ability to react with specific cysteine residues on targeted
OHDA in a dose-dependent manner within the range of 0.1–1 mM. proteins via thiol (S-) alkylation. The Kelch ECH associating protein
Two years later, Lin et al. [66] explored the contribution of 1 (Keap1) Nrf2 pathway is the major regulator of cytoprotective
glutathione S-transferase (GST)-P in this neuroprotective effect in responses to endogenous and exogenous stresses caused by
the same cell type and used 6-OHDA-treated rats to demonstrate reactive oxygen species and electrophiles [73].
an improvement of the locomotor activity after 1 mM CA The involvement of (Keap1-Nrf2) pathway in the neuro-
pretreatment. Later, they revealed that the reduction in 6- protective effect of CA against oxidative stress and excitotoxicity
OHDA-induced SH-SY5Y cell death by CA at the same dose was was first described by Satoh et al. [74] in cerebrocortical neurons.
associated with the induction of parkin, a Parkinson disease linked They demonstrated that this activation was initiated by S-
gene that in turn up-regulated the ubiquitin-proteasome system alkylation of the Keap1 protein critical cysteine thiol by the
[67]. “electrophilic” quinone-type of CA. Then, Satoh et al. [75]
Zhang et al. [68] demonstrated a potent neuroprotective effect suggested that the protective effect of CA at 10 mM against
of CA at 10 mM against cyanide brain poisoning, both in vitro using oxidative glutamate toxicity on HT22 cells required both free
cultured human and rodent pluripotent neuronal stem cells and in carboxylic acid and catechol hydroxyl moieties to exert a
576 S. Bahri et al. / Biomedicine & Pharmacotherapy 84 (2016) 569–582

regulatory action on cellular redox impairment. This occurs via neuropathic pain model via spinal sirtuin1 activation and down-
activation of the transcriptional antioxidant-responsive element of regulation of p66shc expression [82]. In conclusion, Nrf2 pathway
phase-2 genes, including gamma-glutamyl cysteine ligase, hem- can be a common molecular mechanism for neurotrophic
eoxygenase-1 and NADPH-dependent quinone oxidoreductase, properties of CA, while tissue-specific neuroprotective action of
accompanied by an upregulation of glutathione expression and CA may involve also other effectors.
metabolism [76].
Nrf2 regulates the expression of antioxidant and cytoprotective 5.5. Canosic acid and obesity
proteins, and that requires its binding to the antioxidant-response
element (ARE) within DNA [77]. Several studies cited above Obesity is a serious health disorder caused by accumulation of
demonstrated that Nrf2 can be activated by CA to exert its excessive amount of body fat that may increase the risk of other
cytoprotective effect. Indeed, a prior administration of CA at 1 mg/ diseases such as diabetes, arthritis, heart troubles and cancer.
kg suppressed the inhibition of mitochondrial respiration induced Overweight develops following high-fat diet, lack of physical
by 4-hydroxynonenal (4-HNE)-treatment in rat cerebral cortical activities or may be genetically related. Treatments that limit
cells cultured ex vivo via the reduction of the level of 4-HNE bound adipogenesis progression and differentiation can be a good
to mitochondrial proteins and via Nrf2-ARE activation [77]. This strategy to cure obesity-related diseases. Thus, some studies have
signaling pathway is also implicated in CA neuroprotection from elucidated the antiadipogenic effects of CA (at a dose ranging from
mitochondrial oxidative damage after experimental traumatic 0.1 to 10 mM). These studies have shown an activation of the
brain injury in mice [78]. CA administered after injury at a dose of antioxidant-response element (ARE), the induction of phase2
3 mg/kg maintained mitochondrial respiratory function and enzymes and an up-regulation of intracellular level of total GSH
reduced cytoskeletal breakdown, lipid peroxidation and protein [83].
nitration via Nrf2-ARE activation. CA treatment on the first day of differentiation of preadipocytes
It was also reported that carnosic acid at 15 mM favors neurite 3T3-L1 into adipocytes can inhibit adipogenesis, associated with a
outgrowth and neuronal differentiation of PC12h cells via Nrf2- suppression of mitotic clonal expansion and with expression
p62/ZIP pathway, which enhances the cell surface TrkA neuro- inhibition of critical transcriptional factors of adipogenesis, C/
trophin receptor signaling, known to be implicated in neuronal EBPa and PPARg [84]. This antiadipogenic mechanism involves
survival and differentiation by several neurotrophin such as NGF also an attenuation of lipid accumulation in 3T3-L1 adipocytes [85]
[79]. The latter could be also activated by CA via Nrf2 pathway in and HepG2 cells [86] with a down-regulation of triglycerides level
human T98G glioblastoma cells and normal astrocytes. NGF-gene accumulated in adipocytes especially at CA concentration of 10 mM
induction can be produced by CA in a dose/time dependent manner [85]. CA was described also to repress palmitate-induced cellular
(up to 50 mM, incubated for 2 h or 24 h) in parallel with an increase lipid accumulation in vivo via activation of EGFR and MAPK
in Nrf2 expression as well as that of its target genes [80]. Vaka et al. phosphorylation [86]. However, it should be noted that the use of a
[81] confirmed these results in vivo and demonstrated that nontoxic dose of CA was essential. In fact, Dickmann et al. [87]
intranasal delivery of CA at a dose of 4 mg/kg together with showed that CA exhibits a dose-dependent hepatotoxicity in
chitosan can enhance the endogenous levels of neurotrophins primary human hepatocytes with an IC50 value of 94.8  36.7 mM.
including NGF. This suggests that CA is potential herbal molecule It could still increase activity of some members of cytochrome
that can treat body alterations associated with neurotrophins P450 superfamily enzymes in human liver microsomes when
shortage. administered at 10 mM. These results provide an indication of a
In summary, Nrf2 pathway appears to be an important safe dose of CA to avoid its toxicity and to obtain a beneficial
molecular effector following CA treatment in many experimental pharmacological effect for the induction of weight loss.
studies (Fig. 3). Nonetheless, other signaling pathways may be also Various experimental studies have used rosemary extract rich
involved. For example, CA exhibits anti-nociceptive property in in CA to demonstrate its preventive/curative effect in obesity and

Fig. 3. Alleged involvement of Nuclear factor erythroid-2 related factor -2 (Nrf 2) in the protective effect of carnosic acid against brain disorders.
Carnosic acid (CA) inhibits methylglyoxal-induced glycation increase in Alzheimer brain via activation of Nrf2/PI3K/AKT signaling pathway. CA preserved photoreceptors
morphology and function in an experimental model of retinal pigmentosa and attenuated experimental model of parkinsonism induced by 6-OHDA, Paraquat or Cyanide by
an up-regulation of Nrf2 and a down-regulation of JNK/p38.
S. Bahri et al. / Biomedicine & Pharmacotherapy 84 (2016) 569–582 577

to confirm its reducing effects on body weight gain without 5.7. Carnosic acid and skin lesions
modification of food intake [88–92,93]. This mechanism was
further elucidated by the evaluation of several biochemical Ultraviolet (UV) radiation is able to penetrate deeper into the
markers, such as glycemia, plasma cholesterol level, serum/liver skin to provoke premature dermal ageing characterized by a
triglycerides, insulin level, plasma ALT and AST and free fatty acid gradual loss of elasticity and by the development of dry and rough
levels, which were reduced by the administration of rosemary skin. UV is known to induce oxidative DNA damage by ROS
extract compared with control diet. This resulted from the production and to activate various types of matrix metalloprotei-
inhibition of gastric lipase activity [90] and was accompanied by nase (MMPs) in the skin. This implicates epidermal growth factor
an improvement in glucose tolerance [92]. receptor (EGFR) and extracellular signal-regulated kinase (ERK)-
The antioxidant power of rosemary extract containing 80% of mediated AP-1 signaling [98,99].
CA was evidenced by a reduction of malondialdehyde levels in CA at low concentrations ranging from 0.3 to3 mM has been first
plasma and liver and by an increase in the GSH/GSSG ratio in the reported to suppress the increase of metalloproteinase 1 (MMP-1)
liver [93]. The anti-inflammatory effect was revealed by a mRNA level in human skin fibroblasts exposed to UVA radiation
reduction in circulating IL-1b, leptin, adiponectin and tumor [100]. Park et al. [101] confirmed this dose-dependent inhibitory
necrosis factor alpha levels when rats are treated with rosemary effect of carnosic acid against UVA/UVB induced MMP-1, MMP-3
extract containing 40% of CA [91]. In addition, the high amount of and MMP-9 expression in a dose–dependent manner (3–10 mM) in
fecal lipid excretion was behind the suppression of lipid human dermal fibroblast cell line (Hs68), primary normal human
absorption and the diminution of lipid accumulation in mouse dermal fibroblasts (HDFs) and normal human epidermal kerati-
hepatocytes [93]. The decrease in body weight gain was nocytes (HEKs). CA prevented ERK and AP-1 activation and reduced
attributed also to the modulation of microbiota composition ROS level induced by UV [101].
and a reduction of b-glucosidase content in rat caecum with an CA at 20 mM can also exert an anti-inflammatory action in skin
increase in its fecal evacuation [94]. The molecular mechanisms by attenuating several interleukins and chemoattractant protein
accompanying these hepatic regulations by rosemary extract production, and by inhibiting tumor necrosis factor TNF-a,
implicate a reduction of phase I and phase II gene expression [91], prostaglandin E2 (PGE2) and nitric oxide (NO) release following
an activation of the expression of receptor of advanced glycation an inflammatory reaction [102]. This may be mediated through the
end products (RAGE) [93] and activations of hepatic lipogenic- blockade of the nuclear translocation of nuclear factor (NF)-kB and
related genes with an increase in lipolysis-related genes through the inhibition of Src-family/Syk tyrosine kinases.
expression [92].
The majority of experimental studies designed to evaluate the 5.8. Other possible therapeutic applications of carnosic acid
antiobesity effect of rosemary extract demonstrated that this
molecule can be a promising agent to attenuate metabolic Various publications have shown others properties of CA. Lee
disorders by regulating fatty acid metabolism, hepatic fat et al., [103] reported the antiplatelet activity due in part to the
accumulation, glucose tolerance and various other manifestations. blockage of collagen, arachidonic acid, and thrombin-mediated
cytosolic calcium mobilization with IC50 values of 39  0.3 mM,
5.6. Carnosic acid and gastric lesions 34  1.8 mM and 48  2.9 mM. CA at 60 mM and 150 mM can exert
an antiviral activity against human respiratory syncytial virus
Doolaege et al. [6] demonstrated that CA given in a single dose (hRSV) infection by suppressing its replication, inhibiting both A
orally (64.3  5.8 mg/kg) is absorbed in rats into blood circulation. and B type hRSV and blocking their genes expression [104]. CA at
After this oral administration as well as after single intravenous 100 mM can also attenuate cisplatin-induced experimental neph-
dose (20.5  4.2 mg/kg) traces of CA were located in gut, abdomen, rotoxicity due to its strong antioxidant and antiapoptotic proper-
liver, legs and muscle tissues. The distribution of CA in rats after ties, through the inhibition of lipid peroxidation, caspase-3 and
intragastric administration (90 mg/kg) is limited to stomach and ROS production and through the enhancement of some antioxidant
intestine [95] and the molecule is eliminated through the fecal enzymes levels [105]. Other beneficial property have been
route. To assess its gastroprotective activity, some studies identified to be the counteraction of obesity-associated hypergly-
compared the effect of different CA derivates in vivo using HCl/ cemia and insulin resistance, by the improvement of glucose
EtOH-induced gastric lesions model in mice or rat and in vitro on homeostasis and by the amelioration of peripheral glucose
different gastric cell lines. clearance in skeletal muscle tissue (L6 myotubes) through a
In fact, the gastroprotective action of a single oral dose of CA PME-1/PP2A/PKB signaling pathway when CA is administered at
alkyl derivatives (40 mg/kg) has been demonstrated to correlate 20 mM [106]. Similar results were observed in vitro, where CA at
with the length of the alkyl chain in the molecule and the best 10 mM attenuated insulin resistance in 3T3-L1 adipocytes via Akt-
effect was attributed to oleate and palmitate derivates [96]. dependent FoxO1 signaling pathway [107]. Likewise, CA can exert
According to Theoduloz et al. [97], some derivatives given at 10 mM an anti-inflammatory action against tumor necrosis factor-a (TNF-
show a better preventive action with low cytotoxicity against a)-inducing mRNA expression of inflammatory genes in 3T3-L1
gastric lesions compared to the reference compound lansopra- adipocytes via NF-kB and AP-1 pathways.
zole at the same dose. The elucidation of processes involved in this These studies suggest that CA may regulate several cellular
gastroprotective effect was performed using human cell culture alterations and target different molecular/metabolic mediators in a
models, such as human gastric adenocarcinoma cell line (AGS) and tissue-specific manner, especially depending on the type of the
lung fibroblasts (MRC-5) treated with sodium taurocholate (NaT) lesion occurred.
that produces cell lesions [97]. Gastroprotective properties of CA
included an increase in the cellular reduced glutathione (GSH) and 6. Carnosic acid synergizes with other natural compounds
prostaglandin E2 (PGE2) levels, protection against cell lesion
induced by NaT and inhibition of lipid peroxidation. However, the We have mentioned above that the antileukemic action of CA
signaling pathways behind these effects are still imperfectly was boosted by a combinatory treatment with natural compounds
understood. Further studies using these gastroprotective mole- such as 1a,25(OH)2D3 vitamin D derivate, retinoic acid and
cules derived from CA are required to determine the molecular curcumin. CA also enhanced arsenic trioxide-induced HL60 human
mechanisms involved. myeloid leukemia cells apoptosis. This synergizing property of CA
578 S. Bahri et al. / Biomedicine & Pharmacotherapy 84 (2016) 569–582

has been demonstrated from the beginning of the experiments compromise cell viability. These results highlight the strong ability
designed to explore the anti-carcinogenic effects of this molecule. of RA to bind and increase the effects of CA.
Fuhrman et al. [108] proved also that tomato lycopene (5 mM)
acts synergistically with CA (25 mM), as an effective antioxidant 8. Conclusion
against low-density lipoprotein (LDL) oxidation, known to cause
atherosclerosis. Lycopene belongs to the family of carotenoids. It is Numerous evidences of the beneficial effect of CA for the
a natural pigment that gives red color to plants such as tomato. treatment of various health disorders have been published.
Insoluble in water, lycopene is transported by the blood lipids and Related mechanisms include the inhibition of oxidative stress, the
is accumulated in certain organs, particularly in the liver and attenuation of inflammation and cancer. In the latter case, CA can
prostate. This study showed that lycopene exhibited synergistic attenuate tumor progression via apoptosis induced by ROS
antioxidative effect with other components such as vitamin E, generation and caspase activation. This implicates in some cases
glabridin, rosmarinic acid and garlic against LDL oxidation, induced both intrinsic and extrinsic apoptotic pathways, ER stress
either by copper ions or by the radical generator AAPH. This induction and PPAR cleavage [33]. The molecular pathways
indicates that the molecular recombination phenomenon can involved in this preventive/therapeutic effect of CA against some
occur with this natural compound and is not specific to CA. type of cancer are summarized in Table 1. It is important to
Another original recent study aimed to develop CA derivates in point out that there is no particular or universal signaling
silico able to inhibit acetylcholinesterase activity, in order to pathway taken by CA for the killing of different types of cancer
reverse the lack in acetylcholine content that causes memory cells. Indeed, CA on various types of carcinomas may induce
damage [109]. In this context, CA derivates cooperate with apoptosis, cell cycle arrest, autophagy, and inhibition of cellular
acetylcholinesterase to induce a negative feedback, i.e. down proliferation Table 1. In each case, different signaling pathways
regulation in its expression. This study offers an alternative are involved. In some cases, even opposite modulation of a
strategy to treat Alzheimer's disease. signaling pathway is reported; as can be seen, Akt is activated in
In summary, synergistic action of CA with other molecules may leukemia cell, while it is rather down-regulated in other tissues in
constitute the basis of a combinatory treatment approach against the presence of CA.
the emergence of cancer cells, but also against other dysfunctions Great variability of modulation of signaling pathways is
that may occur in the human organism. It is important to figure out observed in the treatment of other disorders besides cancer. As
whether the synergy occurs at the level of cellular signalization or shown in Table 2, effects of CA can be arbitrary divided into three
the chemical combination takes place in the molecule that creates groups. First group of effects concerns cell viability. Major
new chemical compounds responsible of the enhancement of pathways modulated by CA are sirtuin-1, PI3K/Akt, Keap-1/Nrf2,
several specific properties of CA. MAPK-p38/JNK and ERK1/2. One can witness cell specific action of
CA for MAPK-p38/JNK pathway. While in neuronal cells this
7. Rosmarinic acid and carnosic acid: possible connection pathway is activated to protect against apoptosis [70], MAPK-p38/
JNK is inhibited by CA to produce antiapoptotic effect in myocytes
CA has been described to produce a synergistic beneficial effect [51]. However, it is difficult to compare these two studies because
in human diseases by cooperating with other natural compounds, of uncertainty about the in situ concentration of CA in the study of
such as 25-dihydroxyvitamin D3, all-trans retinoic acid and Kocak et al. [51]. The dose used in this study (50 mg/kg) is rather
curcumin. Cooperation between CA and other phenol compounds high and, given the high bioavailability of CA, this could be
in planta such as rosmarinic acid is very possible to boost its translated into higher micromolar concentration of CA in the
biological activities. This reinforces the power of rosemary extract myocardium. Thus, we could suggest concentration dependent
to treat several human diseases. Indeed, rosmarinic acid (RA, dual effect of CA on MAPK-p38/JNK pathway. Better example of
C18H16O8, Fig. 4) is one of the most important component of dose-dependent signaling pathway modulation is the inhibition of
rosemary and it been isolated from many species of the Lamiaceae PI3K/Akt in cancer cells by high-micromolar CA concentrations and
family [110]. RA and its derivatives are easily produced in cell PI3K/Akt activation in neuronal cells by low-micromolar concen-
cultures in high yield without a special handling, because this trations (Tables 1 and 2). Similarly, in comparison with the Table 1,
substance is one of the secondary metabolites that are synthesized one can note that, at low micromolar concentrations, carnosic acid
continuously in plants [111]. induces rather anti-apoptotic effect in hepatocytes in contrast to
Many biological activities of RA have been reported. These are higher doses. Clearly, further studies are needed to check whether
mainly the anti-inflammatory [112], anti-angiogenic [113], anti- this dual effect of CA on cell viability can be observed in other cell
oxidant [114], anti-apoptotic [115], and anti-fibrotic [116] activi- types.
ties. These properties could be potentiated in cooperation with CA. Second group of CA effects concerns inflammation. These
In this context, our research team demonstrated synergic potential effects include the reduction of TNFa via TACE activation,
between CA and RA in human lung fibroblasts, rat lung fibroblasts, inhibition of NFkB/p65/p50, suppression of PPARg cleavage,
rat alveolar type II cells and L929 cells cultured in vitro (submitted). increase in PGE2 level and inhibition of NO release (Table 2).
We reported an enhancement in the proapoptotic activity of CA in Finally, antioxidative properties of CA are thrown into relief by the
these cells in the presence of RA, while RA alone did not increase of GST-P and the reduction of the redox protein p66shc
expression (Table 2). Again, CA at higher doses can induce ROS
generation in some type of cells, while cell pretreatment with
lower doses protects them from the oxidant stress (Tables 1 and 2).
Taken as a whole, this review highlights the need for further
studies to better understand the mechanisms of action of CA in
some forms of human diseases including cancer. It is important to
show first its bioavailability in human body before any human
clinical studies could be performed. Nonetheless, carnosic acid
shows its strong relevance to the treatment of many health
disorders and finally it could find its way to the list of accepted
Fig. 4. Rosmarinic acid. drugs in phytotherapy.
S. Bahri et al. / Biomedicine & Pharmacotherapy 84 (2016) 569–582 579

Table 1
Summary of different molecular pathways involved in the anticancer effect of carnosic acid.

Type of CA dose Effect on target cancer cells Molecular pathway involved Ref.
cancer
Colorectal 20– Induction of HCT 116 cells apoptosis ROS generation, inhibition of STAT3 [10]
cancer 100 mM
1– Inhibition of HT-29 cells proliferation Inhibition of AKT and ERK [11]
10 mM
Renal 10– Human renal carcinoma CaKi cells apoptosis ROS generation, TRAIL mediated apoptosis [25]
cancer 20 mM
Brain 10– Human neuroblastoma IMR-32 cells apoptosis Inhibition of ERK and activation of p38 and JNK, [28]
cancer 40 mM ROS generation
Leukemia 2.5– G1 cell cycle arrest/apoptosis on HL-60 and U937 human myeloid leukemia cells in the Activation of cyclin dependant kinase inhibitors [5]
7.5 mM presence of 100 mM of curcuma p21 WAFI and p27 Kipl
15 mM G1 cell cycle arrest/apoptosis on HL-60 human myeloid leukemia cells, in the presence Activation of PTEN/AKT [34]
of 4 mM of As2O
Skin cancer 5– Inhibition of murine B16-F10 melanoma cells adhesion and migration Inhibition of AKT [39]
10 mM
Prostate 20– PC-3 cells apoptosis Modulation of AKT/IKK/NF-kB [40]
cancer 100 mM
Liver cancer 20– HepG2 cells autophagy Inhibition of Akt/mTOR pathway [13]
100 mM
5– HepG2 cells apoptosis Inhibition of Akt/mitochondrial dysfunction [14]
100 mM

Table 2
Signaling pathways modulated by carnosic acid.

Target CA Cell type Physiological effect Ref.


dose
Cell survival Sirtuin-1 activation 10 mM L02 cells CA induced a decrease in the release of cytochrome C, AIF and in [46]
mitochondrial apoptosis induced by ethanol
1 mM Primary hepatocytes and CA reversed the increase in ROS level, LDH release and the decrease in cell [48,49]
HepG2 cells viability produced by H2O2
Keap-1/Nrf2 activation 10 mM COS7 and PC12 h cells CA protects neurons from oxidative stress and glutamate-induced [74]
excitotoxicity
PI3 K/AKT activation 1 mM SH-SY5Y cells CA prevents the effects of paraquat on cell viability and redox parameters [69]
MAPK-p38/JNK activation 1 mM SH-SY5Y cells CA reversed the reduction of Bcl-2/Bax ratio, induction of caspase 3 and [70]
PARP cleavage by 6-OHDA
MAPK-p38/JNK inhibition, 50 mg/ Myocardial cells CA reduced myocardial damage and apoptosis [51]
ERK1/2 activation kg

Inflammation Reduction of TNFa level 50 mg/ Myocardial cells CA reduced myocardial impairment [51]
kg
TACE activation 30 mM SH-SY5Y cells CA reduced beta-amyloid secretion [57]
NFkB/p65/p50 inhibition 20 mM aortic smooth muscle CA inhibited vascular smooth muscle migration [52]
cells
20 mM U937 cells CA reduced U937 cells adhesion to HUVECs [53]
PPARg cleavage suppression 10 mM SH-SY5Y cells CA reduced beta-amyloid oligomer cellular content [58]
Increase in PGE2 level 10 mM Gastric adeno-carcinoma CA protected against cell lesion induced by sodium taurocholate [97]
cell
Inhibition of NO release 20 mM Keratinocyte HaCaT cells CA protected against skin inflammatory responses [102]

Oxidative Increase of GST-P level 1 mM SH-SY5Y cells CA prevented 6-OHDA-induced apoptosis and oxidative stress [66]
stress
Reduction of p66shc 1 mM HepG2 human hepatoma CA decreased p66shc protein expression induced by ethanol [47]
expression cells

Conflicts of interest References

The authors declare no conflict of interest. [1] H. Linde, Ein neues diterpen aus salvia officinalis L. und eine notiz zur
konstitution von pikrosalvin, Helv. Chim. Acta 47 (1964) 1234–1239.
[2] E. Wenkert, A. Fuchs, J.D. Mc Chesney, Chemical artifacts from the family
Ethical responsibilities protection of people and animals Labiatae, J. Org. Chem. 30 (1965) 2931–2934.
[3] R.A. Hill, J.D. Connoly, Triterpenoids, Nat. Prod. Rep. 30 (2013) 1028–1065.
[4] S. Munne-Bosch, L. Alegre, Changes in carotenoids, tocopherols and
The authors declare that this study has not been conducted on diterpenes during drought and recovery, and the biological significance of
humans or animals. chlorophyll loss in Rosmarinus officinalis plants, Planta 210 (2000) 925–931.
[5] M. Steiner, I. Priel, J. Giat, J. Levy, Y. Sharoni, M. Danilenko, Carnosic acid
inhibits proliferation and augments differentiation of human leukemic cells
Confidentiality of data induced by 1,25-dihydroxyvitamin D3 and retinoic acid, Nutr. Cancer 41
(2001) 135–144.
[6] E.H. Doolaege, K. Raes, F. De Vos, R. Verhé, S. De Smet, Absorption,
The authors declare that this review does not contain patient distribution and elimination of carnosic acid, a natural antioxidant from
data. Rosmarinus officinalis, in rats, Plant Foods Hum. Nutr. 66 (2011) 196–202.
580 S. Bahri et al. / Biomedicine & Pharmacotherapy 84 (2016) 569–582

[7] M. Romo Vaquero, R. García Villalba, M. Larrosa, M.J. Yáñez-Gascón, E. activity of carnosic acid-rich rosemary extract and the 19-nor Gemini vitamin
Fromentin, J. Flanagan, M. Roller, F.A. Tomás-Barberán, J.C. Espín, M.T. García- D analogue in a mouse model of systemic acute myeloid leukemia, Oncology
Conesa, Bioavailability of the major bioactive diterpenoids in a rosemary 75 (2008) 203–214.
extract: metabolic profile in the intestine, liver, plasma, and brain of Zucker [32] S. Pesakhov, M. Khanin, G.P. Studzinski, M. Danilenko, Distinct combinatorial
rats, Mol. Nutr. Food Res. 57 (2013) 1834–1846. effects of the plant polyphenols curcumin, carnosic acid, and silibinin on
[8] Q.L. Wang, H. Li, X.X. Li, C.Y. Cui, R. Wang, N.X. Yu, L.X. Chen, Acute and 30-day proliferation and apoptosis in acute myeloid leukemia cells, Nutr. Cancer 62
oral toxicity studies of administered carnosic acid, Food Chem. Toxicol. 50 (2010) 811–824.
(2012) 4348–4355. [33] S. Pesakhov, M. Nachliely, Z. Barvish, N. Aqaqe, B. Schwartzman, E. Voronov, Y.
[9] M.V. Barni, M.J. Carlini, E.G. Cafferata, L. Puricelli, S. Moreno, Carnosic acid Sharoni, G.P. Studzinski, D. Fishman, M. Danilenko, Cancer-selective cytotoxic
inhibits the proliferation and migration capacity of human colorectal cancer Ca2+ overload in acute myeloid leukemia cells and attenuation of disease
cells, Oncol. Rep. 27 (2012) 1041–1048. progression in mice by synergistically acting polyphenols curcumin and
[10] D.H. Kim, K.W. Park, I.G. Chae, J. Kundu, E.H. Kim, J.K. Kundu, K.S. Chun, carnosic acid, Oncotarget (2016), doi:http://dx.doi.org/10.18632/
Carnosic acid inhibits STAT3 signaling and induces apoptosis through oncotarget.7240.
generation of ROS in human colon cancer HCT116 cells, Mol. Carcinog. 55 [34] R. Wang, W.H. Cong, G. Guo, X.X. Li, X.L. Chen, X.N. Yu, H. Li, Synergism
(2015) 1096–1110. between carnosic acid and arsenic trioxide on induction of acute myeloid
[11] Y.J. Kim, J.S. Kim, Y.R. Seo, J.H. Park, M.S. Choi, M.K. Sung, Carnosic acid leukemia cell apoptosis is associated with modulation of PTEN/Akt signaling
suppresses colon tumor formation in association with antiadipogenic pathway, Chin. J. Integr. Med. 18 (2012) 934–941.
activity, Mol. Nutr. Food Res. 58 (2014) 2274–2285. [35] X.N. Yu, X.L. Chen, X.X. Li, H.Q. Li, W.R. Jin, Reversion of P-glycoprotein-
[12] I. Borrás-Linares, A. Pérez-Sánchez A, J. Lozano-Sánchez, E. Barrajón-Catalán, mediated multidrug resistance in human leukemic cell line by carnosic acid,
D. Arráez-Román, A. Cifuentes, V. Micol, A.S. Carretero, A bioguided Chin. J. Physiol. 51 (2008) 348–356.
identification of the active compounds that contribute to the [36] L.Q. Wang, R. Wang, X.X. Li, X.N. Yu, X.L. Chen, H. Li, The anti-leukemic effect
antiproliferative/cytotoxic effects of rosemary extract on colon cancer cells, of carnosic acid combined with adriamycin in a K562/A02/SCID leukemia
Food Chem. Toxicol. 80 (2015) 215–222. mouse model, Int. J. Clin. Exp. Med. 8 (2015) 11708–11717.
[13] Q. Gao, H. Liu, Y. Yao, L. Geng, X. Zhang, L. Jiang, B. Shi, F. Yang, Carnosic acid [37] A. López-Jiménez, M. García-Caballero, M.Á. Medina, A.R. Quesada, Anti-
induces autophagic cell death through inhibition of the Akt/mTOR pathway angiogenic properties of carnosol and carnosic acid, two major dietary
in human hepatoma cells, J. Appl. Toxicol. 35 (2015) 485–492. compounds from rosemary, Eur. J. Nutr. 52 (2013) 85–95.
[14] Q. Xiang, Y. Ma, J. Dong, R. Shen, Carnosic acid induces apoptosis associated [38] T. Kayashima, K. Matsubara, Antiangiogenic effect of carnosic acid and
with mitochondrial dysfunction and Akt inactivation in HepG2 cells, Int. J. carnosol, neuroprotective compounds in rosemary leaves, Biosci. Biotechnol.
Food Sci. Nutr. 66 (2015) 76–84. Biochem. 76 (2012) 115–119.
[15] S. Arico, A. Petiot, C. Bauvy, P.F. Dubbelhuis, A.J. Meijer, P. Codogno, E. Ogier- [39] S.Y. Park, H. Song, M.K. Sung, Y.H. Kang, K.W. Lee, J.H. Park, Carnosic acid
Denis, The tumor suppressor PTEN positively regulates macroautophagy by inhibits the epithelial-mesenchymal transition in B16F10 melanoma cells: a
inhibiting the phosphatidylinositol 3-kinase/protein kinase B pathway, J. possible mechanism for the inhibition of cell migration, Int. J. Mol. Sci. 15
Biol. Chem. 2765 (2001) 35243–35246. (2014) 12698–12713.
[16] O. Oral O, Y. Y. Akkoc, O. O. Bayraktar, D. D. Gozuacik, Physiological and [40] S. Kar, S. Palit, W.B. Ball, P.K. Das, Carnosic acid modulates Akt/IKK/NF-kB
pathological significance of the molecular cross-talk between autophagy and signaling by PP2A and induces intrinsic and extrinsic pathway mediated
apoptosis, Histol. Histopathol. 18 (2015) 11714. apoptosis in human prostate carcinoma PC-3 cells, Apoptosis 17 (2012) 735–
[17] A. Thoburn, Apoptosis and autophagy: regulatory connections between two 747.
supposedly different processes, Apoptosis 13 (2008) 1–9. [41] O. Yesil-Celiktas, C. Sevimli, E. Bedir, F. Vardar-Sukan, Inhibitory effects of
[18] J. Thorburn, F. Moore, A. Rao, W.W. Barclay, L.R. Thomas, K.W. Grant, C.D. rosemary extracts, carnosic acid and rosmarinic acid on the growth of various
Cramer, A. Thorburn, Selective inactivation of a FADD-dependent apoptosis human cancer cell lines, Plant Foods Hum. Nutr. 65 (2010) 158–163.
and autophagy pathway in immortal epithelial cells, Mol. Biol. Cell 16 (2005) [42] L.S. Einbond, H.A. Wu, R. Kashiwazaki, K. He, M. Roller, T. Su, X. Wang, S.
1189–1199. Goldsberry, Carnosic acid inhibits the growth of ER-negative human breast
[19] J.O. Pyo, M.H. Jang, Y.K. Kwon, H.J. Lee, J.I. Jun, H.N. Woo, D.H. Cho, B. Choi, H. cancer cells and synergizes with curcumin, Fitoterapia 83 (2012) 1160–
Lee, J.H. Kim, N. Mizushima, Y. Oshumi, Y.K. Jung, Essential roles of Atg5 and 1168.
FADD in autophagic cell death: dissection of autophagic cell death into [43] Q. Xiang, Z. Liu, Y. Wang, H. Xiao, W. Wu, C. Xiao, X. Liu, Carnosic acid
vacuole formation and cell death, J. Biol. Chem. 280 (2005) 20722–20729. attenuates lipopolysaccharide-induced liver injury in rats via fortifying
[20] A.P. Martin, C. Mitchell, M. Rahmani, K.P. Nephew, S. Grant, P. Dent, Inhibition cellular antioxidant defense system, Food Chem. Toxicol. 53 (2013) 1–9.
of MCL-1 enhances lapatinib toxicity and overcomes lapatinib resistance via [44] X. Tian, Y. Hu, M. Li, K. Xia, J. Yin, J. Chen, Z. Liu, Carnosic acid attenuates acute
BAK-dependent autophagy, Cancer Biol. Ther. 8 (2009) 2084–2096. ethanol-induced liver injury via a SIRT1/p66Shc-mediated mitochondrial
[21] S. Yousefi, R. Perozzo, I. Schmid, A. Ziemiecki, T. Schaffner, L. Scapozza, T. pathway, Can. J. Physiol. Pharmacol. 22 (2015) 1–10.
Brunner, H.U. Simon, Calpain-mediated cleavage of Atg5 switches autophagy [45] Q. Guo, Z. Shen, H. Yu, G. Lu, Y. Yu, X. Liu, P. Zheng, Carnosic acid protects
to apoptosis, Nat. Cell Biol. 8 (2006) 1124–1132. against acetaminophen-induced hepatotoxicity by potentiating Nrf2-
[22] S. Pattingre, A. Tassa, X. Qu, R. Garuti, X.H. Liang, N. Mizushima, M. Packer, M. mediated antioxidantcapacity in mice, Korean J. Physiol. Pharmacol. 20
D. Schneider, B. Levine, Bcl-2 antiapoptotic proteins inhibit beclin 1- (2016) 15–23.
dependent autophagy, Cell 122 (2005) 927–939. [46] W. Shan, L. Gao, W. Zeng, Y. Hu, G. Wang, M. Li, J. Zhou, X. Ma, X. Tian, J. Yao,
[23] S. Costa, A. Utan, E. Speroni, R. Cervellati, G. Piva, A. Prandini, M.C. Guerra, Activation of the SIRT1/p66shc antiapoptosis pathway via carnosic acid-
Carnosic acid from rosemary extracts: a potential chemoprotective agent induced inhibition of miR-34a protects rats against nonalcoholic fatty liver
against aflatoxin B1. An in vitro study, J. Appl. Toxicol. 27 (2007) 152–159. disease, Cell Death Dis. 6 (2015) e1833.
[24] K.J. Min, K.J. Jung, T.K. Kwon, Carnosic acid induces apoptosis through [47] L. Gao, W. Shan, W. Zeng, Y. Hu, G. Wang, X. Tian, N. Zhang, X. Shi, Y. Zhao, C.
reactive oxygen species-mediated endoplasmic reticulum stress induction in Ding, F. Zhang, K. Liu, J. Yao, Carnosic acid alleviates chronic alcoholic liver
human renal carcinoma caki cells, J. Cancer Prev. 19 (2014) 170–178. injury by regulating the SIRT1/ChREBP and SIRT1/p66shc pathways in rats,
[25] K.J. Jung, K.J. Min, J.H. Bae, T.K. Kwon, Carnosic acid sensitized TRAIL- Mol. Nutr. Food Res. (2016), doi:http://dx.doi.org/10.1002/mnfr.201500878.
mediated apoptosis through down-regulation of c-FLIP and Bcl-2 expression [48] Y. Hu, N. Zhang, Q. Fan, M. Lin, C. Zhang, G. Fan, X. Zhai, F. Zhang, Z. Chen, J. Yao,
at the post translational levels and CHOP-dependent up-regulation of DR5, Protective efficacy of carnosic acid against hydrogen peroxide induced
Bim, and PUMA expression in human carcinoma caki cells, Oncotarget 6 oxidative injury in HepG2 cells through the SIRT1 pathway, Can. J. Physiol.
(2015) 1556–1568. Pharmacol. 93 (2015) 625–631.
[26] K. Kosaka, J. Mimura, K. Itoh, T. Satoh, Y. Shimojo, C. Kitajima, A. Maruyama, [49] T. Wang, Y. Takikawa, Carnosic acid protects normal mouse hepatocytes
M. Yamamoto, T. Shirasawa, Role of Nrf2 and p62/ZIP in the neurite against H2 O2 -induced cytotoxicity via sirtuin 1-mediated signaling,
outgrowth by carnosic acid in PC12h cells, J. Biochem. 147 (2010) 73–81. Hepatol. Res. 46 (2016) 239–246.
[27] M.R. de Oliveira, G.C. Ferreira, P.F. Schuck, S.M. Dal Bosco, Role for the PI3 K/ [50] B.D. Sahu, U.K. Putcha, M. Kuncha, S.S. Rachamalla, R. Sistla, Carnosic acid
Akt/Nrf2 signaling pathway in the protective effects of carnosic acid against promotes myocardial antioxidant response and prevents isoproterenol-
methylglyoxal-induced neurotoxicity in SH-SY5Y neuroblastoma cells, induced myocardial oxidative stress and apoptosis in mice, Mol. Cell
Chem. Biol. Interact. 242 (2015) 396–406. Biochem. 394 (2014) 163–176.
[28] C.W. Tsai, C.Y. Lin, H.H. Lin, J.H. Chen, Carnosic acid, a rosemary phenolic [51] C. Kocak, F.E. Kocak, R. Akcilar, O.O. Isiklar, H. Kocak, Z. Bayat, H. Simsek, F.
compound, induces apoptosis through reactive oxygen species-mediated Taser, I. Altuntas;1, Molecular and biochemical evidence on the protective
p38 activation in human neuroblastoma IMR-32 cells, Neurochem. Res. 36 effects of embelin and carnosic acid in isoproterenol-induced acute
(2011) 2442–2451. myocardial injury in rats, Life Sci. 147 (2016) 15–23.
[29] L.H. Ren, J.J. Chen, H.P. An, Enhancement of differentiation induction of HL-60 [52] Y.M. Yu, H.C. Lin, W.C. Chang, Carnosic acid prevents the migration of human
cells by 1,25-dihydroxyvitamin D3 in combination with carnosic acid, aortic smooth muscle cells by inhibiting the activation and expression of
Zhongguo Dang Dai Er Ke Za Zhi. 10 (2008) 55–59. matrix metalloproteinase-9, Br. J. Nutr. 100 (2008) 731–738.
[30] M. Danilenko, Q. Wang, X. Wang, J. Levy, Y. Sharoni, G.P. Studzinski, Carnosic [53] Y.M. Yu, C.H. Lin, H.C. Chan, H.D. Tsai, Carnosic acid reduces cytokine-induced
acid potentiates the antioxidant and prodifferentiation effects of 1a, 25- adhesion molecules expression and monocyte adhesion to endothelial cells,
dihydroxyvitamin D3 in leukemia cells but does not promote elevation of Eur. J. Nutr. 48 (2009) 101–106.
basal levels of intracellular calcium, Cancer Res. 63 (2003) 1325–1332. [54] P. Meng, H. Yoshida, T. Matsumiya, T. Imaizumi, K. Tanji, F. Xing, R. Hayakari, J.
[31] A. Shabtay, H. Sharabani, Z. Barvish, M. Kafka, D. Amichay, J. Levy, Y. Sharoni, Dempoya, T. Tatsuta, T. Aizawa-Yashiro, J. Mimura, K. Kosaka, K. Itoh, K. Satoh,
M.R. Uskokovic, G.P. Studzinski, M. Danilenko, Synergistic antileukemic Carnosic acid suppresses the production of amyloid-b 1–42 by inducing the
S. Bahri et al. / Biomedicine & Pharmacotherapy 84 (2016) 569–582 581

metalloprotease gene TACE/ADAM17 in SH-SY5Y human neuroblastoma [77] D.M. Miller, I.N. Singh, J.A. Wang, E.D. Hall, Administration of the Nrf2-ARE
cells, Neurosci. Res. 75 (2013) 94–102. activators sulforaphane and carnosic acid attenuates 4-hydroxy-2-nonenal-
[55] N. Azad, H. Rasoolijazi, M.T. Joghataie, S. Soleimani, Neuroprotective effects of induced mitochondrial dysfunction ex vivo, Free Radic. Biol. Med. 57 (2013)
carnosic acid in an experimental model of Alzheimer's disease in rats, Cell J. 1–9.
Spring. 13 (2011) 39–44. [78] D.M. Miller, I.N. Singh, J.A. Wang, E.D. Hall, Nrf2-ARE activator carnosic acid
[56] H. Rasoolijazi, N. Azad, M.T. Joghataei, M. Kerdari, F. Nikbakht, M. Soleimani, decreases mitochondrial dysfunction, oxidative damage and neuronal
The protective role of carnosic acid against beta-amyloid toxicity in rats, Sci. cytoskeletal degradation following traumatic brain injury in mice, Exp.
World J. (2013) 917082, doi:http://dx.doi.org/10.1155/2013/917082. Neurol. 264 (2014) 103–110.
[57] P. Meng, H. Yoshida, T. Matsumiya, T. Imaizumi, K. Tanji, F. Xing, R. Hayakari, J. [79] K. Kosaka, J. Mimura, K. Itoh, T. Satoh, Y. Shimojo, C. Kitajima, 1A. Maruyama,
Dempoya, T. Tatsuta, T. Aizawa-Yashiro, J. Mimura, K. Kosaka, K. Itoh, K. Satoh, M. Yamamoto, T. Shirasawa, Role of Nrf2 and p62/ZIP in the neurite
Carnosic acid suppresses the production of amyloid-b 1–42 by inducing the outgrowth by carnosic acid in PC12 h cells, J. Biochem. 147 (2010) 73–81.
metalloprotease gene TACE/ADAM17 in SH-SY5Y human neuroblastoma [80] J. Mimura, K. Kosaka, A. Maruyama, T. Satoh, N. Harada, H. Yoshida, K. Satoh,
cells, Neurosci. Res. 75 (2013) 94–102. M. Yamamoto, K. Itoh, Nrf2 regulates NGF mRNA induction by carnosic acid in
[58] P. Meng, H. Yoshida, K. Tanji, T. Matsumiya, F. Xing, R. Hayakari, L. Wang, K. T98G glioblastoma cells and normal human astrocytes, J. Biochem. 150 (2011)
Tsuruga, H. Tanaka, J. Mimura, K. Kosaka, K. Itoh, I. Takahashi, S. Kawaguchi, T. 209–217.
Imaizumi, Carnosic acid attenuates apoptosis induced by amyloid-b 1–42 or [81] S.R. Vaka, S.N. Murthy, M.A. Repka, T. Nagy, Upregulation of endogenous
1–43 in SH-SY5Y human neuroblastoma cells, Neurosci. Res. 94 (2015) 1–9. neurotrophin levels in the brain by intranasal administration of carnosic acid,
[59] M.R. de Oliveira, G.C. Ferreira, P.F. Schuck, S.M. Dal Bosco, Role for the PI3 K/ J. Pharm. Sci. 100 (2011) 3139–3145.
Akt/Nrf2 signaling pathway in the protective effects of carnosic acid against [82] S.D. Chen, B.B. Ji, Y.X. Yan, X. He, K.Y. Han, Q.X. Dai, M.X. Zhang, Y.C. Mo, J.L.
methylglyoxal-induced neurotoxicity in SH-SY5Y neuroblastoma cells, Wang, Carnosic acid attenuates neuropathic pain in rat through the
Chem. Biol. Interact. 242 (2015) 396–406. activation of spinal sirtuin1 and down-regulation of p66shc expression,
[60] A.G. Kanthasamy, M. Kitazawa, Y. Yang, V. Anantharam, A. Kanthasamy, Neurochem. Int. 93 (2016) 95–102.
Environmental neurotoxin dieldrin induces apoptosis via caspase-3- [83] T. Takahashi, T. Tabuchi, Y. Tamaki, K. Kosaka, Y. Takikawa, T. Satoh, Carnosic
dependent proteolytic activation of protein kinase C delta (PKCdelta): acid and carnosol inhibit adipocyte differentiation in mouse 3T3-L1 cells
implications for neurodegeneration in Parkinson’s disease, Mol. Brain 1 through induction of phase2 enzymes and activation of glutathione
(2008) 12. metabolism, Biochem. Biophys. Res. Commun. 382 (2009) 549–554.
[61] L.E. Gee, N. Chen, A. Ramirez-Zamora, D.S. Shin, J.G. Pilitsis, The effects of [84] M. Gaya, V. Repetto, J. Toneatto, C. Anesini, G. Piwien-Pilipuk, S. Moreno,
subthalamic deep brain stimulation on mechanical and thermal thresholds in Antiadipogenic effect of carnosic acid, a natural compound present in
6OHDA-lesioned rats, Eur. J. Neurosci. 42 (2015) 2061–2069. Rosmarinus officinalis, is exerted through the C/EBPs and PPARg pathways at
[62] D. Zhang, B. Lee, A. Nutter, P. Song, N. Dolatabadi, J. Parker, S. Sanz-Blasco, T. the onset of the differentiation program, Biochim. Biophys. Acta 1830 (2013)
Newmeyer, R. Ambasudhan, S.R. McKercher, E. Masliah, S.A. Lipton, 3796–3806.
Protection from cyanide-induced brain injury by the Nrf2 transcriptional [85] M.Y. Park, M.K. Sung, Carnosic acid inhibits lipid accumulation in 3T3-L1
activator carnosic acid, J. Neurochem. 133 (2015) 898–908. adipocytes through attenuation of fatty acid desaturation, J. Cancer Prev. 20
[63] R. Filograna, V.K. Godena, A. Sanchez-Martinez, E. Ferrari, L. Casella, M. (2015) 41–49.
Beltramini, L. Bubacco, A.J. Whitworth, M. Bisaglia, SOD-mimetic M40403 is [86] T. Wang, Y. Takikawa, T. Tabuchi, T. Satoh, K. Kosaka, K. Suzuki, Carnosic acid
protective in cell and fly models of paraquat toxicity: implications for (CA) prevents lipid accumulation in hepatocytes through the EGFR/MAPK
Parkinson disease, J. Biol. Chem. 291 (2016) 9257–9267. pathway, J. Gastroenterol. 47 (2012) 805–813.
[64] J.A. Park, S. Kim, S.Y. Lee, C.S. Kim, K. Kim do, S.J. Kim, H.S. Chun, Beneficial [87] L.J. Dickmann, B.M. VandenBrink, Y.S. Lin, In vitro hepatotoxicity and
effects of carnosic acid on dieldrin-induced dopaminergic neuronal cell cytochrome P450 induction and inhibition characteristics of carnosic acid, a
death, Neuroreport 19 (2008) 1301–1304. dietary supplement with antiadipogenic properties, Drug Metab. Dispos. 40
[65] J.H. Chen, H.P. Ou, C.Y. Lin, F.J. Lin, C.R. Wu, S.W. Chang, C.W. Tsai, Carnosic acid (2012) 1263–1267.
prevents 6-hydroxydopamine-induced cell death in SH-SY5Y cells via [88] K. Ninomiya, H. Matsuda, H. Shimoda, N. Nishida, N. Kasajima, T. Yoshino, T.
mediation of glutathione synthesis, Chem. Res. Toxicol. 25 (2012) 1893–1901. Morikawa, M. Yoshikawa, Carnosic acid, a new class of lipid absorption
[66] C.Y. Lin, J.H. Chen, R.H. Fu, C.W. Tsai, Induction of Pi form of glutathione S- inhibitor from sage, Bioorg. Med. Chem. Lett. 14 (2004) 1943–1946.
transferase by carnosic acid is mediated through PI3 K/Akt/NF-kB pathway [89] A. Ibarra, J. Cases, M. Roller, A. Chiralt-Boix, A. Coussaert, C. Ripoll, Carnosic
and protects against neurotoxicity, Chem. Res. Toxicol. 27 (2015) 1958–1966. acid-rich rosemary (Rosmarinus officinalis L.) leaf extract limits weight gain
[67] C.Y. Lin, C.W. Tsai, C.W. Tsai, Carnosic acid protects SH-SY5Y cells against 6- and improves cholesterol levels and glycaemia in mice on a high-fat diet, Br. J.
hydroxydopamine-induced cell death through upregulation of parkin Nutr. 106 (2011) 1182–1189.
pathway, Neuropharmacology 3908 (2016) 30157–30165. [90] M. Romo Vaquero, M.J. Yáñez-Gascón, R. García Villalba, M. Larrosa, E.
[68] D. Zhang, B. Lee, A. Nutter, P. Song, N. Dolatabadi, J. Parker, S. Sanz-Blasco, T. Fromentin, A. Ibarra, M. Roller, F. Tomás-Barberán, J.C. Espín de Gea, M.T.
Newmeyer, R. Ambasudhan, S.R. McKercher, E. Masliah, S.A. Lipton, García-Conesa, Inhibition of gastric lipase as a mechanism for body weight
Protection from cyanide-induced brain injury by the Nrf2 transcriptional and plasma lipids reduction in Zucker rats fed a rosemary extract rich in
activator carnosic acid, J. Neurochem. 133 (2015) 898–908. carnosic acid, PLoS One 7 (2012) e39773.
[69] M.R. de Oliveira, G.C. Ferreira, P.F. Schuck, Protective effect of carnosic acid [91] M. Romo-Vaquero, M. Larrosa, M.J. Yáñez-Gascón, N. Issaly, J. Flanagan, M.
against paraquat-induced redox impairment and mitochondrial dysfunction Roller, F.A. Tomás-Barberán, J.C. Espín, M.T. Garcia-Conesa, A rosemary
in SH-SY5Y cells: role for PI3 K/Akt/Nrf2 pathway, Toxicol. In Vitro 32 (2016) extract enriched in carnosic acid improves circulating adipocytokines and
41–54. modulates key metabolic sensors in lean Zucker rats: critical and
[70] C.R. Wu, C.W. Tsai, S.W. Chang, C.Y. Lin, L.C. Huang, C.W. Tsai, Carnosic acid contrasting differences in the obese genotype, Mol. Nutr. Food Res. 58
protects against 6-hydroxydopamine-induced neurotoxicity in vivo and in (2014) 942–953.
vitro model of Parkinson’s disease: involvement of antioxidative enzymes [92] M.Y. Park, M.K. Sung, Carnosic acid attenuates obesity-induced glucose
induction, Chem. Biol. Interact. 225 (2015) 40–46. intolerance and hepatic fat accumulation by modulating genes of lipid
[71] T. Rezaie, S.R. McKercher, K. Kosaka, M. Seki, L. Wheeler, V. Viswanath, T. metabolism in C57BL/6J-ob/ob mice, J. Sci. Food Agric. 95 (2015) 828–835.
Chun, R. Joshi, M. Valencia, S. Sasaki, T. Tozawa, T. Satoh, S.A. Lipton, [93] Y. Zhao, R. Sedighi, P. Wang, H. Chen, Y. Zhu, S. Sang, Carnosic acid as a major
Protective effect of carnosic acid, a pro-electrophilic compound, in models of bioactive component in rosemary extract ameliorates high-fat-diet-induced
oxidative stress and light-induced retinal degeneration, Invest. Ophthalmol. obesity and metabolic syndrome in mice, J. Agric. Food Chem. 63 (2015)
Vis. Sci. 53 (2012) 7847–7854. 4843–4852.
[72] K. Kang, M.J. Tarchick, X. Yu, C. Beight, P. Bu, M. Yu, Carnosic acid slows [94] M. Romo-Vaquero, M.V. Selma, M. Larrosa, M. Obiol, R. García-Villalba, R.
photoreceptor degeneration in the Pde6b(rd10) mouse model of retinitis González-Barrio, N. Issaly, J. Flanagan, M. Roller, F.A. Tomás-Barberán, M.T.
pigmentosa, Sci. Rep. 6 (2016) 22632. García-Conesa, A rosemary extract rich in carnosic acid selectively modulates
[73] E. Kansanen, S.M. Kuosmanen, H. Leinonen, A.L. Levonen, The Keap1Nrf2 caecum microbiota and inhibits b-glucosidase activity, altering fiber and
pathway: mechanisms of activation and dysregulation in cancer, Redox Biol. 1 short chain fatty acids fecal excretion in lean and obese female rats, PLoS One
(2013) 45–49. 9 (2014) e94687.
[74] T. Satoh, K. Kosaka, K. Itoh, A. Kobayashi, M. Yamamoto, Y. Shimojo, C. [95] H. Yan, W. He, X. Li, C. Nie, L. Wang, X. Li, L. Wu, P. Tu, Determination of
Kitajima, J. Cui, J. Kamins, S. Okamoto, M. Izumi, T. Shirasawa, S.A. Lipton, carnosic acid in rat stomach and intestine by high performance liquid
Carnosic acid, a catechol-type electrophilic compound, protects neurons chromatography method, Zhongguo Zhong Yao Za Zhi. 34 (2009) 766–769.
both in vitro and in vivo through activation of the Keap1/Nrf2 pathway via S- [96] M.W. Pertino, C. Theoduloz, J.A. Rodríguez, T. Yáñez, V. Lazo, G. Schmeda-
alkylation of targeted cysteines on Keap1, J. Neurochem. 104 (2008) 1116– Hirschmann, Gastroprotective effect of carnosic acid gamma-lactone
1131. derivatives, J. Nat. Prod. 73 (2010) 639–643.
[75] T. Satoh, M. Izumi, Y. Inukai, Y. Tsutsumi, N. Nakayama, K. Kosaka, Y. Shimojo, [97] C. Theoduloz, M.W. Pertino, J.A. Rodríguez, G. Schmeda-Hirschmann,
C. Kitajima, K. Itoh, T. Yokoi, T. Shirasawa, Carnosic acid protects neuronal Gastroprotective effect and cytotoxicity of carnosic acid derivatives, Planta
HT22 cells through activation of the antioxidant-responsive element in free Med. 77 (2011) 882–887.
carboxylic acid- and catechol hydroxyl moieties-dependent manners, [98] G.J. Fisher, S.C. Datta, H.S. Talwar, Z.Q. Wang, J. Varani, S. Kang, J.J. Voorhees,
Neurosci. Lett. 434 (2008) 260–265. Molecular basis of sun-induced premature skin ageing and retinoid
[76] Y. Tamaki, T. Tabuchi, T. Takahashi, K. Kosaka, T. Satoh, Activated glutathione antagonism, Nature 379 (1996) 335–339.
metabolism participates in protective effects of carnosic acid against [99] D. Peus, R.A. Vasa, A. Meves, A. Beyerle, M.R. Pittelkow, UVB-induced
oxidative stress in neuronal HT22 cells, Planta Med. 76 (2010) 683–688. epidermal growth factor receptor phosphorylation is critical for downstream
582 S. Bahri et al. / Biomedicine & Pharmacotherapy 84 (2016) 569–582

signaling and keratinocyte survival, Photochem. Photobiol. 72 (2000) 135– [109] M. Merad, W. Soufi, S. Ghalem, F. Boukli, M.H. Baig, K. Ahmad, M.A. Kamal,
140. Molecular interaction of acetylcholinesterase with carnosic acid derivatives:
[100] E.A. Offord, J.C. Gautier, O. Avanti, C. Scaletta, F. Runge, K. Krämer, L.A. a neuroinformatics study, CNS Neurol. Disord. Drug Targets 13 (2014) 440–
Applegate, Photoprotective potential of lycopene, beta-carotene, vitamin E, 446.
vitamin C and carnosic acid in UVA-irradiated human skin fibroblasts, Free [110] M. Petersen, M.S. Simmonds, Rosmarinic acid, Phytochemistry 62 (2003)
Radic. Biol. Med. 32 (2002) 1293–1303. 121–125.
[101] M. Park, J. Han, C.S. Lee, B.H. Soo, K.M. Lim, H. Ha, Carnosic acid, a phenolic [111] H.P. Bais, T.S. Walker, H.P. Schweizer, Root specific elicitation and
diterpene from rosemary, prevents UV-induced expression of matrix antimicrobial activity of rosmarinic acid in hairy root cultures of Ocimum
metalloproteinases in human skin fibroblasts and keratinocytes, Exp. basilicum, Plant Physiol. Biochem. 40 (2002) 983–995.
Dermatol. 22 (2013) 336–341. [112] N. Osakabe, H. Takano, C. Sanbongi, A. Yasuda, R. Yanagisawa, K. Inoue, T.
[102] J. Oh, T. Yu, S.J. Choi, Y. Yang, H.S. Baek, S.A. An, L.K. Kwon, J. Kim, H.S. Rho, S.S. Yoshikawa, Anti-inflammatory and anti-allergic effect of rosmarinic acid (RA)
Shin, W.S. Choi, S. Hong, J.Y. Cho, Syk/Src pathway-targeted inhibition of skin inhibition of seasonal allergic rhinoconjunctivitis (SAR) and its mechanism,
inflammatory responses by carnosic acid, Mediators Inflamm. (2012) 781375, Biol. Factors 21 (2004) 127–131.
doi:http://dx.doi.org/10.1155/2012/781375. [113] S.S. Huang, R.L. Zheng, Rosmarinic acid inhibits angiogenesis and its
[103] J.J. Lee, Y.R. Jin, J.H. Lee, J.Y. Yu, X.H. Han, K.W. Oh, J.T. Hong, T.J. Kim, Y.P. Yun, mechanism of action in vitro, Cancer Lett. 239 (2006) 271–280.
Antiplatelet activity of carnosic acid, a phenolic diterpene from Rosmarinus [114] X. Sui, T. Liu, C. Ma, L. Yang, Y. Zu, L. Zhang, H. Wang, Microwave irradiation to
officinalis, Planta Med. 73 (2007) 121–127. pretreat rosemary (Rosmarinus officinalis L.) for maintaining antioxidant
[104] H.B. Shin, M.S. Choi, B. Ryu, N.R. Lee, H.I. Kim, H.E. Choi, J. Chang, K.T. Lee, D.S. content during storage and to extract essential oil simultaneously, Food
Jang, K.S. Inn, Antiviral activity of carnosic acid against respiratory syncytial Chem. 131 (2012) 1399–1405.
virus, Virol. J. 10 (2013) 303. [115] D.S. Kim, H.R. Kim, E.R. Woo, S.T. Hong, H.J. Chae, S.W. Chae, Inhibitory effects
[105] B.D. Sahu, K.K. Rentam, U.K. Putcha, M. Kuncha, G.M. Vegi, R. Sistla, Carnosic of rosmarinic acid on adriamycin-induced apoptosis in H9c2 cardiac muscle
acid attenuates renal injury in an experimental model of rat cisplatin- cells by inhibiting reactive oxygen species and the activations of c-Jun N-
induced nephrotoxicity, Food Chem. Toxicol. 49 (2011) 3090–3097. terminal kinase and extracellular signal-regulated kinase, Biochem. Pharm.
[106] C. Lipina, H.S. Hundal, Carnosic acid stimulates glucose uptake in skeletal 70 (2005) 1066–1078.
muscle cells via a PME-1/PP2A/PKB signalling axis, Cell Signal. 26 (2014) [116] G.S. Li, W.L. Jiang, J.W. Tian, G.W. Qu, H.B. Zhu, F.H. Fu, In vitro and in vivo
2343–2349. antifibrotic effects of rosmarinic acid on experimental liver fibrosis,
[107] C.W. Tsai, K.L. Liu, Y.R. Lin, W.C. Kuo, The mechanisms of carnosic acid Phytomedicine 17 (2010) 282–288.
attenuates tumor necrosis factor-a-mediated inflammation and insulin
resistance in 3T3-L1 adipocytes, Mol. Nutr. Food Res. 58 (2013) 654–664.
[108] B. Fuhrman, N. Volkova, M. Rosenblat, M. Aviram, Lycopene synergistically
inhibits LDL oxidation in combination with vitamin E, glabridin, rosmarinic
acid, carnosic acid, or garlic, Antioxid. Redox Signal. 2 (2002) 491–506.

You might also like