Access to this full-text is provided by Frontiers.
Content available from Frontiers in Immunology
This content is subject to copyright.
July 2016 | Volume 7 | Article 2781
REVIEW
published: 22 July 2016
doi: 10.3389/fimmu.2016.00278
Frontiers in Immunology | www.frontiersin.org
Edited by:
Nurit Hollander,
Tel Aviv University, Israel
Reviewed by:
Robert Weissert,
University of Regensburg, Germany
Helmut Jonuleit,
University of Mainz, Germany
*Correspondence:
Azzam A. Maghazachi
amagazachi@sharjah.ac.ae
Specialty section:
This article was submitted to
Immunotherapies and Vaccines,
a section of the journal
Frontiers in Immunology
Received: 30May2016
Accepted: 06July2016
Published: 22July2016
Citation:
Al-JaderiZ and MaghazachiAA
(2016) Utilization of Dimethyl
Fumarate and Related Molecules for
Treatment of Multiple Sclerosis,
Cancer, and Other Diseases.
Front. Immunol. 7:278.
doi: 10.3389/fimmu.2016.00278
Utilization of Dimethyl Fumarate and
Related Molecules for Treatment of
Multiple Sclerosis, Cancer, and Other
Diseases
Zaidoon Al-Jaderi and Azzam A. Maghazachi*
Department of Clinical Sciences, College of Medicine and Sahrjah Institute for Medical Research, University of Sharjah,
Sharjah, United Arab Emirates
Several drugs have been approved for treatment of multiple sclerosis (MS). Dimethyl
fumarate (DMF) is utilized as an oral drug to treat this disease and is proven to be potent
with less side effects than several other drugs. On the other hand, monomethyl fumarate
(MMF), a related compound, has not been examined in greater details although it has
the potential as a therapeutic drug for MS and other diseases. The mechanism of action
of DMF or MMF is related to their ability to enhance the antioxidant pathways and to
inhibit reactive oxygen species. However, other mechanisms have also been described,
which include effects on monocytes, dendritic cells, T cells, and natural killer cells. It is
also reported that DMF might be useful for treating psoriasis, asthma, aggressive breast
cancers, hematopoeitic tumors, inflammatory bowel disease, intracerebral hemorrhage,
osteoarthritis, chronic pancreatitis, and retinal ischemia. In this article, we will touch on
some of these diseases with an emphasis on the effects of DMF and MMF on various
immune cells.
Keywords: NK cells, dimethyl fumarate, cancer, multiple sclerosis, monomethyl fumarate
INTRODUCTION
Twenty years ago, there was no treatment for multiple sclerosis (MS). Today, there are wide varieties
of immunomodulatory drugs, which have been licensed to treat MS patients. For relapsing-remitting
MS (RRMS) patients, several drugs have been approved. During the 1990s, beta interferon and
glatiramer acetate were the only drugs available. Other drugs, such as natalizumab, teriunomide,
and ngolimod, were later approved. ese drugs modify the immune system to slow disease pro-
gression, decrease attacks, and reduce the development of new brain lesions.
Dimethyl fumarate (DMF) has been lately approved by the US Food and Drug Administration
(FDA) as an oral drug for MS patients. is drug was rst used to treat inammatory skin diseases,
such as psoriasis. e benecial eects of this medication corroborated with regulating CD4+ 1
cell dierentiation. In clinical trials, it showed positive benets for MS patients by lowering risk of
relapse and reducing the number of brain lesions (1–6).
e mechanism of action is not fully known. Aer oral intake, DMF is completely absorbed in the
small intestine, and only small amounts are excreted in the feces and urine (7). DMF possesses a short
half-life of ~12min inside the body (8). Aer absorption, DMF is rapidly hydrolyzed by esterases
to monomethyl fumarate (MMF) (9), which has a short half-life of 36h. is molecule interacts
FIGURE 1 | Chemical structures of DMF and MMF. Also shown is the structure of fumaric acid, the precursor molecule.
2
Al-Jaderi and Maghazachi New Therapeutic Modalities for Cancer
Frontiers in Immunology | www.frontiersin.org July 2016 | Volume 7 | Article 278
with the immune cells in the blood circulation and crosses the
blood–brain barrier (BBB) to the central nervous system (CNS)
(10). Figure1 shows the structures of DMF and MMF.
Dimethyl fumarate is an α, β-unsaturated carboxylic acid
ester. It is demonstrated that DMF by activating nuclear factor
erythroid 2-related factor (Nrf2), stimulated the production of
glutathione (GSH), the cells most important scavenger of reac-
tive oxygen species (ROS) (11), hence, protecting against ROS-
induced cytotoxicity. Further studies demonstrate that DMF
downregulated nuclear factor kappa B (NF-κB) in cells, inhibited
the anti-apoptotic protein Bcl-2, and induced apoptosis. Cells
challenged with oxidative stressors increase their antioxidant
capacity as a response to increase ROS production and maintain
homeostasis. Nrf2 acts as a key control of the redox gene tran-
scription; under oxidative stress, the Nrf2 signaling is activated
to enhance the expression of a large number of antioxidants and
enzymes that restore redox homeostasis. Nrf2 interacts with
the cysteine-rich protein Kelch-like ECH-associated protein 1
(Keap1) and acts as an adaptor protein for the Cul3-dependent
E3 (Cul3) ubiquitin ligase complex. In normal conditions, Keap1
promotes ubiquitination and repeatedly eliminates Nrf2 within
a half-life of 13–21min (12, 13). Keap1 possesses many cysteine
residues in the amino acid terminal that act as sensors detecting
changes in cellular redox state. During cellular stress, Keap1 is less
eective at promoting Nrf2 degradation (12, 14).
Under normal conditions, Nrf2 is sequestered in the cytoplasm
via binding to its inhibitory molecule Keap1. ROS/stress causes
dissociation of Nrf2-Keap1 complex, leading to activation of Nrf2
and its translocation into the nucleus. In the nucleus, Nrf2 het-
erodimerizes with other transcription factors, such as MAF, and
consequently, binds the antioxidant responsive elements (ARE)
in the target genes. Nrf2 promotes transcriptional activation of
antioxidants and detoxifying enzymes. At the same time, phos-
phorylation of the repressor molecule IκB by ROS/stress causes
activation of NF-κB, leading to activating gene transcription
encoding inammatory mediators. Studies have shown that Nrf2
and NF-κB pathways have inhibitory inuence on one another.
EFFECTS OF DMF ON THE INNATE
IMMUNE SYSTEM
e main components of the innate immune system are epithelial
barriers, leukocytes, dendritic cells (DCs), and natural killer (NK)
cells. NK cells are large granular lymphocytes that spontaneously
lyse target cells and are important for defending against viral
infections as well as controlling tumor growth. NK cells have also
immunoregulatory role by secretion of cytokines, chemokines,
as well as cell-to-cell cross-talk (15). ese cells express several
activating and inhibitory receptors that detect target cells and
control NK cell activity. In human, NK cells are divided by the
expression of CD56 molecule into CD56dim and CD56bright subsets
(15, 16).
e ow cytometric analysis of peripheral blood immune
cells in 41 DMF-treated MS patients shows that these patients
had signicantly fewer circulating CD8+ T cells, CD4+ T cells,
CD56dim NK cells, CD19+ B cells, and plasmacytoid DCs (17).
Furthermore, the expression of CXCR3+ (a potential marker for
1) and CCR6+ (a potential marker for 17) was reduced,
while the number of regulatory T cells (Treg) was unchanged.
Interestingly, DMF did not aect circulating CD56bright NK cells,
CD14+ monocytes, or myeloid DCs. However, DMF-treated
patients had signicantly fewer CD56dim NK cells when compared
with healthy controls (17). A clinical study of 35 RRMS patients
at baseline, 3months, 6months, and 12months aer initiation
of DMF treatment shows that total leukocyte and lymphocyte
counts diminished aer 6months, whereas aer 12months of
DMF therapy total T cells counts decreased by 44%, CD8+ T cell
3
Al-Jaderi and Maghazachi New Therapeutic Modalities for Cancer
Frontiers in Immunology | www.frontiersin.org July 2016 | Volume 7 | Article 278
counts declined by 54.6%, and CD4+ T cell counts decreased by
39% (18). CD19+ B cell counts were also reduced by 37.5%, and
eosinophils counts were decreased by 54%, whereas the percent-
ages of neutrophils, monocytes, basophils, and NK cells were not
signicantly altered (18).
It has been previously demonstrated that MMF is a potent
agonist of hydroxycarboxylic acid receptor 2 “(HCA2), a G
protein coupled receptor also known as GPR109A” (19). It is
reported that HCA2 mediates the therapeutic eects of DMF or
MMF in experimental autoimmune encephalomyelitis (EAE)
mouse model (20). Recently, we observed that MMF enhanced
primary non-activated human CD56+ NK cell lysis of leukemic
cell line K562 and B-cell lymphoma RAJI cells in vitro (21).
Furthermore, MMF upregulated NKp46 expression on the
surface of CD56+ NK cells, an activity correlated with upregula-
tion of CD107a expression and the release of Granzyme B from
CD56+ NK cells (21).
Moreover, MMF delayed EAE clinical score in SJL/J mice and
prevented the disease progression in treated mice. ese results
are linked to enhanced NK cells lysis of DCs isolated from the
same mice (22). To correlate these ndings with human settings,
we recently observed that human NK cells incubated with various
concentrations of DMF or MMF robustly lysed immature DCs
in vitro (manuscript in preparation). ese ndings suggest
that one mechanism of action for these “drugs” is plausibly due
to activating NK cells to lyse DCs, and consequently, impeding
antigen presentation to autoreactive T cells.
Dendritic cells represent key links between the innate and
adaptive immune system (23). T cells and NK cells are stimulated
through direct contact with activated DCs (24). DCs play a major
role in regulating the immune response by releasing cytokines
and expressing co-stimulatory molecules. ey are capable
of processing both exogenous and endogenous antigens and
present them in the context of MHC class I or II molecules. It
has been reported that DMF inhibited DCs maturation through
a reduction in the release of the inammatory cytokines IL-6
and IL-12. Furthermore, DMF activated type II DCs, which have
anti-inammatory eects and suppressed type I DCs, which are
inammatory (25, 26). DMF induced type II DCs by regulating
GSH depletion, followed by increased heme oxygenase-1 (HO-1)
expression and suppressing STAT1 phosphorylation in DCs (26).
is, combined with the reduction in the inammatory cytokines
by nuclear translocation of NF-κB, resulted in inhibiting CD1a,
CD40, CD80, CD86, and HLA-DR expression (27). Consequently,
the capacity of DCs to stimulate allogeneic 1 and 17 cells is
reduced. It was also determined that increased production of the
2 cytokines and increased expression of IL-10, instead of IL-12
and IL-23 by DCs, enhanced the development of T regulatory
(Treg) cells (28).
We reported that monocyte-derived DCs isolated from
EAE mice treated with MMF did not increase the expression of
CD80 molecule (22). Interestingly, E-cadherin expression was
upregulated in EAE mice, and MMF reversed this upregula-
tion. Increased E-cadherin expression suggests a shiing of
the immune system toward inammatory 1/17 response.
ese results support previous study showing that inammatory
E-cadherin+ bone marrow-derived DCs isolated from animals
with colitis promoted 17 response (29). e study also dem-
onstrates that E-cadherin+ DCs enhanced 1 cell responses (29).
Furthermore, E-cadherin+ DCs increased the number of IFN-γ+
CD4+ T cells and decreased the number of IL-4+ CD4+ T cells
(30). MMF by decreasing E-cadherin expression on DCs may
decrease inammatory 1/17 proliferation and may enhance
the anti-inammatory 2 cells.
Although DMF but not MMF induced apoptosis in iDCs and
moderately inhibited the ability of DCs to induce proliferation of
allogeneic T cells, it is reported that MMF aected the polariza-
tion but not maturation of monocyte-derived DCs, resulting in
downregulating 1 lymphocyte responses (31). In vitro study
for the eects of MMF on DCs dierentiation shows that MMF
inhibited monocyte-derived DCs dierentiation in response to
LPS, resulting in cells that are incapable of appropriately mature
to DCs. In addition, MMF did not decrease the capacity of DCs to
capture antigens, but MMF/DCs interaction resulted in produc-
ing low levels of IL-12, IL-10, and TNF-α, whereas IL-8 produc-
tion was not altered (32). Consequently, MMF/DCs interaction
partially aected IFN-γ production by naive T cells, whereas the
production of IL-4 and IL-10 was not inuenced by MMF (32).
Another study demonstrates that DMF inhibited DCs matura-
tion by reducing the production of the inammatory cytokines
IL-6 and IL-12 as well as the expression of MHC class II, CD80,
and CD86 (27). Furthermore, immature DCs activated fewer
Tcells characterized by low IFN-γ and IL-17 production (27).
In contrast, de Jong etal. (33) demonstrate that MMF increased
the production of IL-4 and IL-5 without altering the production
of IL-2 and interferon-γ in stimulated peripheral blood mononu-
clear cells challenged with bacterial antigens.
TREATMENT OF PSORIASIS AND OTHER
SKIN DISEASES WITH DMF
Psoriasis is a type-1 cytokine-mediated chronic autoimmune skin
disease aided by the inltration of 1/17 cells into the skin
(34–36). DMF is utilized to treat psoriasis in European countries
for more than 30years. Fumaric acid was rst used for treatment
of psoriasis by the German chemist Walter Schweckendiek in
1959. In 1994, DMF was licensed in Germany under the trade
name Fumaderm for the treatment of psoriasis. DMF inhibited
Janus kinas (JAK) signaling and interfered with intracellular
proteins tracking and consequently, inhibited the release of
pro-inammatory cytokines, such as IL-12, IL-23, and TNF,
whereas the release of anti-inammatory cytokines, such as
IL-10, was increased. DMF also inhibited the production of IFN-
γ and enhanced the production of IL-10 in the culture of psoriatic
keratinocytes (37).
Previous experimental and clinical studies were focused on
the mechanism of action for DMF that could aect the immune
system. e immunohistochemical studies of psoriatic plaques
indicate that DMF has several anti-inammatory eects via a
number of pathways, leading to reduction in the levels of several
inammatory T cell subsets (38, 39) and decreased recruitment
4
Al-Jaderi and Maghazachi New Therapeutic Modalities for Cancer
Frontiers in Immunology | www.frontiersin.org July 2016 | Volume 7 | Article 278
of inammatory cells (40). e ability of DMF or MMF to induce
apoptosis of CD4+ and CD8+ T cells and in vitro switching the
immune system toward a 2 anti-inammatory type response in
psoriasis patients could be through impaired DCs maturation and
induction of apoptosis. In addition, DMF inhibited the formation
of new blood vessels, a process that is involved in the formation
of psoriatic plaques (41).
Clinical studies demonstrate that DMF reduced CD4+ T cells
and CD8+ T cells by inducing apoptotic cell death (42). In vivo
studies indicate that DMF inhibited T cell mediated organ rejec-
tion in a rat model (43). A study of allergic contact dermatitis,
a skin disorder in which an exaggerated T cell response occurs,
shows that DMF suppressed allergen-induced T cell proliferation,
corroborated with modulating cytokines/chemokines expression
by reducing the levels of IFN-γ but not IL-5 and downregulating
CXCR3 but not CCR4 expression (44).
TREATMENT OF MULTIPLE SCLEROSIS
PATIENTS WITH DMF
Multiple sclerosis is a chronic inammatory autoimmune
disease of the CNS in which the insulating myelin sheaths of
nerve cell axons in the brain and spinal cord are attacked by the
immune system (45). e principal mechanism responsible for
this disease is still incompletely understood. e consensus is
that activated T cells attack oligodendrocytes, leading to destruc-
tion of myelin sheaths (demyelination). Furthermore, the pres-
ence of inammatory T cells in the CNS triggers recruitment
of more T cells, Bcells, dendritic cells, microglia cells, and NK
cells (46). Due to the progressive neurodegenerative nature of
MS, therapeutic modalities that exhibit direct neuroprotective
eects are needed. A phase 3 clinical trial study of 2667 RRMS
patients demonstrates the ecacy and safety of DMF in MS (1).
In vitro study indicates that DMF increased the frequency of the
multipotent neurospheres resulting in the survival of mouse and
rat neural stem progenitor cells (NPCs) following oxidative stress
with hydrogen peroxide (H2O2) treatment (47). Using motor
neuron survival assay, DMF signicantly promoted survival
of motor neurons under oxidative stress. Furthermore, DMF
increased the expression of Nrf2 at both RNA and protein levels
in the NPC cultures (47).
ere is agreement that antioxidants reduce the risk of certain
pathological conditions, such as neurodegenerative diseases.
Invivo animal studies have shown that DMF or MMF inhibited
the disease course in the EAE model (48). It is also demonstrated
that MMF crossed the BBB, indicating it may have a direct
cytoprotective function in the CNS (49). e detoxication
capabilities of DMF or MMF reduced the production and release
of inammatory molecules, such as TNF-α, IL-1β, and IL-6 as
well as nitric oxide from microglia and astrocytes activated with
LPS in v itro (50, 51). DMF or MMF increased the production
of detoxication enzymes, such as nicotinamide adenine dinu-
cleotide phosphate quinone reductase 1 (NQO-1), HO-1, and
cellular glutathione, abolishing NF-kB translocation into the
nucleus (52). NQO-1 is also detected in the liver and in the CNS
of DMF-treated animals. is results in decreased expression of
NF-kB-dependent genes that regulate the expression of inam-
matory cytokines, chemokines, and adhesion molecules, and
consequently, reduced the damage to CNS cells. Reducing the
expression of adhesion molecules in the BBB represents a critical
step in the transmigration of immune cells into the CNS. DMF
inhibited TNF-α-induced expression of intracellular adhesion
molecule-1 (ICAM-1), E selection, and the vascular cell adhe-
sions molecule-1 (VCAM-1) in endothelial cells invitro (53, 54).
is is correlated with activating Nrf2 (55–58), which is released
from the Keap-1complex via the activity of fumarates (see above).
is may lead to reducing free radicals, preventing the synthesis
of reactive nitrogen species, and thus protecting the CNS from
degeneration and axonal loss (59, 60). ese immunomodulatory
activities of DMF or MMF, which constitute inhibiting cytokine
production and nitric oxide synthesis, are important for the
protection of oligodendrocytes against ROS-induced cytotoxicity
and consequently, oligodendrocytes survival during an oxidative
attack is augmented.
Multiple sclerosis animal models, such as EAE, are induced by
immunization with dierent myelin antigens, such as proteolipid
peptide (PLP139–151) in SJL/J mice, an animal model disease that
may represent relapsing-remitting form of MS, or C57BL/6J mice
immunized with MOG35–55, a model closely resembles chronic
progressive MS. ese models are characterized by inammation,
demyelination, and axonal lose. Treatment of EAE mice with
DMF reduced macrophage-induced inammation in the spinal
cord (48). DMF suppressed 1 and 17 cell dierentiation as
well as expression of pro-inammatory cytokines IFN-γ, TNF-α,
and IL-17 (61). e drug also promoted 2 cells that produce
IL-4, IL-5, and IL-10 (33). In chronic MS, microglia cells are
activated and released pro-inammatory cytokines and stress-
associated molecules leading to neurodegeneration and alteration
of synaptic transmission (62). Modulation of microglia activation
toward an alternatively activated phenotype can modify the out-
come of some experimental models of neurological diseases. e
study on EAE demonstrates that exposure to MMF switched the
molecular and functional phenotype of activated microglia from
pro-inammatory type to neuroprotective eect (49). is switch
in activity may occur through activation of HCAR2. MMF bind-
ing to HCAR2 triggered a pathway driven by the AMPK/Sirt axis
resulting in inhibition of NF-κB and reducing pro-inammatory
cytokine production (49).
EFFECTS OF DMF ON THE CENTRAL
NERVOUS SYSTEM
A recent study reports that administration of DMF protected
claudin-5 expression in the BBB along with reduced brain edema
formation in C57BL/6 mice undergoing experimental ischemia
reperfusion injury (63). Using the immortalized murine brain
endothelial cell line bEND.3, a preservation of zonula occludens-1
(ZO-1) and VE-cadherin localization in oxygen–glucose deprived
cells in the presence of DMF was observed. Reduced transen-
dothelial migration of the human monocyte cell line THP-1
toward CCL2 chemokine in the lower chamber of a transwell
system aer pretreatment of the bEND.3 cells with DMF was also
5
Al-Jaderi and Maghazachi New Therapeutic Modalities for Cancer
Frontiers in Immunology | www.frontiersin.org July 2016 | Volume 7 | Article 278
noted. Further observations demonstrate decreased ICAM-1,
VCAM-1, and E-selectin mRNA expression in bEND.3 cells aer
treatment with DMF for 6h (54).
In vitro human umbilical vein endothelium examination
indicates that DMF or MMF modulated pro-inammatory
intracellular signaling and T-cell adhesiveness of human brain
microvascular endothelial cells (64). Neither DMF nor MMF
reduced the basal expression of ICAM-1 under inammatory
condition or blocked NF-κB in human brain microvascular
endothelial cells compared to solvent control. Hence, it is sug-
gested that brain endothelial cells do not directly mediate a
potential blocking eect of fumaric acid esters on the inltration
of inammatory T cells into the CNS (64). It is also determined
that DMF ameliorated inammation, reduced BBB permeability
and improved neurological outcomes by casein kinase 2 and Nrf2
signaling pathways in experimental intracerebral hemorrhage
(ICH) mouse model (65).
Evidence from clinical and animal studies suggests that
inammation and oxidative stress, which occur aer hematoma
formation, are involved in ICH-induced secondary brain injury
and neurological dysfunction (66). VCAM-1 and ICAM-1 are
adhesion molecules expressed in the endothelium important dur-
ing inammation and aer tissue injury. Both are increased upon
activation of NF-κB-mediated TNF-α signaling pathway. TNF-α
increases early onset endothelial adhesion by protein kinase
C-dependent upregulation of ICAM-1 expression, which exacer-
bates ICH. Investigating the experimental autoimmune neuritis
indicates that DMF treatment reduced the neurological decits by
ameliorating inammatory cell inltration and demyelination of
sciatic nerves. In addition, DMF treatment decreased the level of
pro-inammatory M1 macrophages, while increasing the number
of anti-inammatory M2 macrophages in the spleens and sciatic
nerves of EAN rats (67, 68). In RAW 264.7 macrophage cell line,
a shi in macrophage polarization from M1 to M2 phenotype
was demonstrated to be dependent on DMF application. In sciatic
nerves, DMF treatment elevated the level of Nrf2 and its target
HO-1, which may facilitate macrophage polarization toward M2
type (68). In addition, by reducing NF-kB in astrocytes, DMF
inhibited the degradation of IkBa and reduced the expression
of nitric oxide synthase (69). Moreover, DMF improved the
inammatory milieu in the spleens of EAN rats, characterized by
downregulating mRNA for IFN-γ, TNF-α, IL-6, and IL-17 and
upregulating mRNA level for IL-4 and IL-10 (68).
EFFECTS OF DMF ON TUMOR
DEVELOPMENT
It has been reported that fumarase is involved in DNA repair
(70). By studying yeast cells, it is observed that cytosolic fumarase
plays a role in detecting and repairing DNA damage, particularly
double-stranded DNA breaks. According to this theory, if the
cells lack the fumarase, they may need to repair damaged DNA
and are most likely prone to develop tumors. Further study on
the role of redox demonstrates that high levels of ROS are harm-
ful to normal cells and may lead to development of tumor by
inducing DNA damage. Malignant transformation also increases
cellular stress, leading to high ROS levels. On the other hand,
Keap1-Nrf2 system protects cells from the eects of oxidants by
regulating the expression of cytoprotective proteins (71). In vivo
evidence indicates that Nrf2 has a protective role against tumor
development in mouse models and in prostate cancer in humans
(72). e mechanism by which Nrf2 is protective against tumor
development has been attributed to the ability of Nrf2 to reduce
the amount of ROS and DNA damages in cells.
It has also been demonstrated that DMF inhibited the prolif-
eration of A375 and M24met cell lines and reduced melanoma
growth and metastasis in experimental melanoma mouse models
(73). Furthermore, DMF arrested the cell cycle at the G2-M
boundary and was pro-apoptotic, inhibiting tumor cell growth.
On the other hand, MMF increased primary human CD56+
NK cell lysis of K562 and RAJI tumor cells, suggesting that this
molecule may have insitu antitumor activity (21).
ROLE OF DMF IN GASTROINTESTINAL
ULCERATION
It has been demonstrated that stress can play a pathogenic role
for gastrointestinal ulceration, by disrupting gastric mucosal
defensive barrier (74). Activators of stress give rise to the release
of corticotropin-releasing hormone (CRH). CRH acts on the
pituitary gland and stimulates the secretion of ACTH, which
promotes glucocorticoids release from the adrenal cortex (75).
Glucocorticoids not only interfere with tissue repair, elevate levels
of gastric acids, and pepsin but also reduce the secretion of gastric
mucus and eventually impair gastric mucosal barrier resulting in
peptic ulcer. Low daily oral doses of MMF may prevent the chronic
foot-shock stress-induced gastric ulcers and may associate with dif-
ferential hormonal and oxidative processes (76). MMF suppressed
the stress-induced elevation in adrenal gland corticosterone level
and modulated the oxidative stress responses. Interestingly, DMF
did not inhibit the eect of innate defense against microorgan-
ism. Treatment of monocytes and neutrophils with DMF aer
stimulation with Staphylococcus aureus, Escherichia coli, or the
yeast Candida albicans in addition to zymosan particles or the
tripeptide fMLP resulted in increased production of superoxide
anion, which exerts anti-microbial eects (77).
EFFECTS OF DMF ON COLLAGEN
TYPE II DEGRADATION
In vivo study of collagen type II degradation suggests that DMF
ameliorated the disease by inhibiting the expression of metallo-
proteinase (MMP)-1, MMP-3, and MMP-13 that are induced by
TNF-α (78). DMF may attenuate MMPs expression by suppress-
ing JAK1 and JAK2/STAT3 pathways and by blocking TNF-α-
induced STAT3 phosphorylation and DNA-binding activity (79).
In vivo mice study on renal brosis, where TGF-β plays a key role
in the development of the disease, demonstrates that DMF treat-
ment may prevent renal brosis via Nrf2-mediated suppression
of TGF-β signaling (80).
6
Al-Jaderi and Maghazachi New Therapeutic Modalities for Cancer
Frontiers in Immunology | www.frontiersin.org July 2016 | Volume 7 | Article 278
CONCLUSION AND FUTURE DIRECTIONS
Dimethyl fumarate was originally used for treatment of psoriasis.
Its success in treating RRMS patients led for its approval as an
oral drug to treat MS patients. One mechanism of action that our
group pursued is that DMF might enhance natural killer cell lysis
of dendritic cells, hence, impeding presenting encephalitogen to
autoreactive T cells. Further investigations suggest that DMF
may also be used in the oncology eld due to its ability to sup-
press the growth of melanoma cells. On the other hand, it appears
that related molecules, such as MMF, may have even more potent
antitumor activity than DMF. Although the association among
DMF and MMF is at present conjectural, it is documented that
MMF has robust antitumor activity by activating natural killer
cells to kill tumor cells. is new mechanism of action for MMF
should provide imputes for investigating this molecule not
only as a therapeutic tool for autoimmune diseases but also for
cancer and immunodecient diseases. Tab l e 1 shows the current
knowledge regarding the eects of DMF and MMF on various
immune cells.
AUTHOR CONTRIBUTIONS
Both authors contributed to writing this review article.
TABLE 1 | Immunoregulatory effects of DMF and/or MMF on various immune cells.
Cell type Molecule Cytokine/other molecules involved Effect(s) Reference
T cells DMF/MMF ↓IFN-γ, ↓TNF-α, ↓IL-17, ↑IL-4, ↑IL-5, ↑IL-10, ↓CXCR3,
↓CCR6
↓Bcl-2, ↑Apoptosis, ↓Th1, ↓Th17, ↑Th2, ↓CD4, ↓CD8,
↑Tre g
(17, 18, 27, 32,
42, 44, 61)
B cells DMF ↑Nrf2→↑GSH→↓ROS, ↓NF-kB ↓Bcl-2, ↑Apoptosis, ↓CD19 B cells (17, 18)
Monocytes DMF ↑Nrf2, ↓NF-kB No effect on cell numbers, ↑Antioxidant response (18, 77)
DCs DMF/MMF ↓GSH→↑HO-1, ↓NF-kB, ↓IL-6, ↓IL-12, ↑IL-10, ↓TNF-α↓,
E-cadherin
↑Apoptosis, ↓plasmacytoid DCs, ↓DC maturation, ↓type I
DCs, ↑type II DCs
(17, 22, 25–27,
31, 32)
NK cells DMF/MMF ↑NKp46, ↑CD107, ↑Granzyme B ↓CD56dim NK cells, No effect on CD56bright numbers,
↑CD56bright NK cells lysis of tumor cells, ↑Lysis of DCS
(17, 18, 21, 22)
Macrophages DMF ↑Nrf2, ↓mRNA of IFN-γ, ↓mRNA of TNF-α, ↓mRNA of IL-6,
↓mRNA of IL-17, ↑mRNA of IL-4, ↑mRNA of IL-10
↓M1 macrophages, ↑M2 macrophages (68)
Neutrophils DMF/MMF ↓HCA2 ↓Number of infiltrating neutrophils (20)
Keratinocytes DMF ↓IL-12, ↓IL-23, ↓TNF, ↓IFN-γ, ↑IL-10, ↓IL-6, ↓TGF-α ↓Proliferation of keratinocytes (37)
Endothelial cells DMF ↓TNF-α, ↓ICAM-1, ↓VCAM-1, ↓E-selection, ↑Nrf2 ↓BBB permeability→↓Immune cell migration (41, 54, 55, 65)
Microglia DMF/MMF ↓IL-1, ↓IL-6, ↓TNF-α, ↓NO, ↑Nrf2→↑GSH→↓ROS,
↓NF-kB, ↑NQO-1, ↑HO-1, ↑HCAR2
↑Antioxidant response, switching activated microglia from
pro-inflammatory to neuroprotective
(49, 50, 52)
Astrocytes DMF/MMF ↑Nrf2→↑GSH→↓ROS, ↓NF-kB, ↓IL-1, ↓IL-6, ↓TNF-α, ↓NO ↑Antioxidant response (50, 52, 58)
Neurons DMF ↑Nrf2→↑GSH→↓ROS ↓Apoptosis, ↑Neurons survival under oxidative stress (47, 58)
Tumor cells DMF Arrest the cell cycle at G2-M, ↓pro-apoptotic ↓Proliferation of melanoma cells, ↓Proliferation of tumor
cells, ↑Apoptosis
(73)
↓, decreased; ↑, increased.
REFERENCES
1. Gold R, Kappos L, Arnold DL, Bar-Or A, Giovannoni G, Selmaj K, et al.
Placebo-controlled phase 3 study of oral BG-12 for relapsing multiple sclero-
sis. N Engl J Med (2012) 367:1098–107. doi:10.1056/NEJMoa1114287
2. Kappos L, Gold R, Miller DH, MacManus DG, Havrdova E, Limmroth V, etal.
Ecacy and safety of oral fumarate in patients with relapsing-remitting multiple
sclerosis: a multicentre, randomised, double-blind, placebo-controlled phase
IIb study. Lancet (2008) 372:1463–72. doi:10.1016/S0140-6736(08)61619-0
3. Stangel M, Linker RA. Dimethyl fumarate (BG-12) for the treatment
of multiple sclerosis. Expert Rev Clin Pharmacol (2013) 6:355–62.
doi:10.1586/17512433.2013.811826
4. Marziniak M. [Multiple sclerosis: new treatment options]. MMW Fortschr
Med (2014) 156(Spec No 1):69–73. doi:10.1007/s15006-014-2549-1
5. Moharregh-Khiabani D, Linker RA, Gold R, Stangel M. Fumaric acid and
its esters: an emerging treatment for multiple sclerosis. Curr Neuropharmacol
(2009) 7:60–4. doi:10.2174/157015909787602788
6. Linker RA, Gold R. Dimethyl fumarate for treatment of multiple sclerosis:
mechanism of action, eectiveness, and side eects. Curr Neurol Neurosci Rep
(2013) 13:394–8. doi:10.1007/s11910-013-0394-8
7. Werdenberg D, Joshi R, Wolram S, Merkle HP, Langguth P. Presystemic
metabolism and intestinal absorption of antipsoriatic fumaric acid esters.
Biopharm Drug Dispos (2003) 24:259–73. doi:10.1002/bdd.364
8. Mrowietz U, Christophers E, Altmeyer P. Treatment of severe psoriasis with
fumaric acid esters: scientic background and guidelines for therapeutic use.
e German Fumaric Acid Ester Consensus Conference. Br J Dermatol (1999)
141:424–9. doi:10.1046/j.1365-2133.1999.03034.x
9. Nibbering PH, io B, Zomerdijk TP, Bezemer AC, Beijersbergen RL, VanFR .
Eects of monomethylfumarate on human granulocytes. J Invest Dermatol
(1993) 101:37–42. doi:10.1111/1523-1747.ep12358715
10. Litjens NH, Burggraaf J, van SE, van GC, Mattie H, Schoemaker RC, etal.
Pharmacokinetics of oral fumarates in healthy subjects. Br J Clin Pharmacol
(2004) 58:429–32. doi:10.1111/j.1365-2125.2004.02145.x
11. Mrowietz U, Asadullah K. Dimethylfumarate for psoriasis: more than a dietary
curiosity. Trends Mol Med (2005) 11:43–8. doi:10.1016/j.molmed.2004.11.003
12. Hong F, Sekhar KR, Freeman ML, Liebler DC. Specic patterns of electrophile
adduction trigger Keap1 ubiquitination and Nrf2 activation. J Biol Chem
(2005) 280:31768–75. doi:10.1074/jbc.M503346200
13. Kobayashi A, Kang MI, Watai Y, Tong KI, Shibata T, Uchida K, et al.
Oxidative and electrophilic stresses activate Nrf2 through inhibition of
ubiquitination activity of Keap1. Mol Cell Biol (2006) 26:221–9. doi:10.1128/
MCB.26.1.221-229.2006
14. Kobayashi A, Kang MI, Okawa H, Ohtsuji M, Zenke Y, Chiba T, et al.
Oxidative stress sensor Keap1 functions as an adaptor for Cul3-based E3 ligase
to regulate proteasomal degradation of Nrf2. Mol Cell Biol (2004) 24:7130–9.
doi:10.1128/MCB.24.16.7130-7139.2004
7
Al-Jaderi and Maghazachi New Therapeutic Modalities for Cancer
Frontiers in Immunology | www.frontiersin.org July 2016 | Volume 7 | Article 278
15. Maghazachi AA. Insights into seven and single transmembrane-spanning
domain receptors and their signaling pathways in human natural killer cells.
Pharmacol Rev (2005) 57:339–57. doi:10.1124/pr.57.3.5
16. Maghazachi AA. Role of chemokines in the biology of natural killer cells. Curr
Top Microbiol Immunol (2010) 341:37–58. doi:10.1007/82_2010_20
17. Longbrake EE, Ramsbottom MJ, Cantoni C, Ghezzi L, Cross AH, Piccio L.
Dimethyl fumarate selectively reduces memory T cells in multiple sclerosis
patients. Mult Scler (2015) 22(8):1061–70. doi:10.1177/1352458515608961
18. Spencer CM, Crabtree-Hartman EC, Lehmann-Horn K, Cree BA, Zamvil SS.
Reduction of CD8(+) T lymphocytes in multiple sclerosis patients treated
with dimethyl fumarate. Neurol Neuroimmunol Neuroinamm (2015) 2:e76.
doi:10.1212/NXI.0000000000000076
19. Tang H, Lu JY, Zheng X, Yang Y, Reagan JD. e psoriasis drug monomethyl
fumarate is a potent nicotinic acid receptor agonist. Biochem Biophys Res
Commun (2008) 375:562–5. doi:10.1016/j.bbrc.2008.08.041
20. Chen H, Assmann JC, Krenz A, Rahman M, Grimm M, Karsten CM, etal.
Hydroxycarboxylic acid receptor 2 mediates dimethyl fumarate’s protective
eect in EAE. J Clin Invest (2014) 124:2188–92. doi:10.1172/JCI72151
21. Vego H, Sand KL, Hoglund RA, Fallang LE, Gundersen G, Holmoy T, et al.
Monomethyl fumarate augments NK cell lysis of tumor cells through degran-
ulation and the upregulation of NKp46 and CD107a. Cel l Mol Immunol (2016)
13:57–64. doi:10.1038/cmi.2014.114
22. Al-Jaderi Z, Maghazachi AA. Vitamin D3 and monomethyl fumarate enhance
natural killer cell lysis of dendritic cells and ameliorate the clinical score in
mice suering from experimental autoimmune encephalomyelitis. Toxins
(Basel) (2015) 7:4730–44. doi:10.3390/toxins7114730
23. Steinman RM. e dendritic cell system and its role in immunogenicity. Annu
Rev Immunol (1991) 9:271–96. doi:10.1146/annurev.iy.09.040191.001415
24. Fernandez NC, Lozier A, Flament C, Ricciardi-Castagnoli P, Bellet D, SuterM,
et al. Dendritic cells directly trigger NK cell functions: cross-talk relevant
in innate anti-tumor immune responses invivo. Nat Med (1999) 5:405–11.
doi:10.1038/7403
25. Ghoreschi K, Bruck J, Kellerer C, Deng C, Peng H, Rothfuss O, etal. Fumarates
improve psoriasis and multiple sclerosis by inducing type II dendritic cells.
J Exp Med (2011) 208:2291–303. doi:10.1084/jem.20100977
26. Zhu K, Mrowietz U. Inhibition of dendritic cell dierentia-
tion by fumaric acid esters. J Invest Dermatol (2001) 116:203–8.
doi:10.1046/j.1523-1747.2001.01159.x
27. Peng H, Guerau-de-Arellano M, Mehta VB, Yang Y, Huss DJ, PapenfussTL,
etal. Dimethyl fumarate inhibits dendritic cell maturation via nuclear factor
kappa B (NF-kappa B) and extracellular signal-regulated kinase 1 and 2
(ERK1/2) and mitogen stress-activated kinase 1 (MSK1) signaling. J Biol
Chem (2012) 287:28017–26. doi:10.1074/jbc.M112.383380
28. Moore KW, de Waal MR, Coman RL, O’Garra A. Interleukin-10 and the
interleukin-10 receptor. Annu Rev Immunol (2001) 19:683–765. doi:10.1146/
annurev.immunol.19.1.683
29. Siddiqui KR, Laont S, Powrie F. E-cadherin marks a subset of inamma-
tory dendritic cells that promote T cell-mediated colitis. Immunity (2010)
32:557–67. doi:10.1016/j.immuni.2010.03.017
30. Zhang Y, Hu X, Hu Y, Teng K, Zhang K, Zheng Y, etal. Anti-CD40-induced
inammatory E-cadherin+ dendritic cells enhance T cell responses and
antitumour immunity in murine Lewis lung carcinoma. J Exp Clin Cancer Res
(2015) 34:11. doi:10.1186/s13046-015-0126-9
31. Litjens NH, Rademaker M, Ravensbergen B, Rea D, van der Plas MJ, io B,
etal. Monomethylfumarate aects polarization of monocyte-derived dendritic
cells resulting in down-regulated 1 lymphocyte responses. Eur J Immunol
(2004) 34:565–75. doi:10.1002/eji.200324174
32. Litjens NH, Rademaker M, Ravensbergen B, io HB, van Dissel JT,
Nibbering PH. Eects of monomethylfumarate on dendritic cell dier-
entiation. Br J Dermatol (2006) 154:211–7. doi:10.1111/j.1365-2133.2005.
07002.x
33. de Jong R, Bezemer AC, Zomerdijk TP, Pouw-Kraan T, Ottenho TH,
Nibbering PH. Selective stimulation of T helper 2 cytokine responses by the
anti-psoriasis agent monomethylfumarate. Eur J Immunol (1996) 26:2067–74.
doi:10.1002/eji.1830260916
34. Nestle FO, Turka LA, Nickolo BJ. Characterization of dermal dendritic cells
in psoriasis. Autostimulation of T lymphocytes and induction of 1 type
cytokines. J Clin Invest (1994) 94:202–9. doi:10.1172/JCI117308
35. Gudjonsson JE, Johnston A, Sigmundsdottir H, Valdimarsson H.
Immunopathogenic mechanisms in psoriasis. Clin Exp Immunol (2004)
135:1–8. doi:10.1111/j.1365-2249.2004.02310.x
36. Valdimarsson H, Bake BS, Jonsdotdr I, Fry L. Psoriasis: a disease of abnormal
Keratinocyte proliferation induced by T lymphocytes. Immunol Today (1986)
7:256–9. doi:10.1016/0167-5699(86)90005-8
37. Ockenfels HM, Schultewolter T, Ockenfels G, Funk R, Goos M. e antip-
soriatic agent dimethylfumarate immunomodulates T-cell cytokine secretion
and inhibits cytokines of the psoriatic cytokine network. Br J Dermatol (1998)
139:390–5. doi:10.1046/j.1365-2133.1998.02400.x
38. Basavaraj KH, Ashok NM, Rashmi R, Praveen TK. e role of drugs in the
induction and/or exacerbation of psoriasis. Int J Dermatol (2010) 49:1351–61.
doi:10.1111/j.1365-4632.2010.04570.x
39. Bacharach-Buhles M, Pawlak FM, Matthes U, Joshi RK, Altmeyer P. Fumaric
acid esters (FAEs) suppress CD 15- and ODP 4-positive cells in psoriasis. Acta
Derm Venereol Suppl (Stockh) (1994) 186:79–82.
40. Rubant SA, Ludwig RJ, Diehl S, Hardt K, Kaufmann R, Pfeilschier JM, etal.
Dimethylfumarate reduces leukocyte rolling in vivo through modulation
of adhesion molecule expression. J Invest Dermatol (2008) 128:326–31.
doi:10.1038/sj.jid.5700996
41. Garcia-Caballero M, Mari-Bea M, Medina MA, Quesada AR.
Dimethylfumarate inhibits angiogenesis invitro and invivo: a possible role
for its antipsoriatic eect? J Invest Dermatol (2011) 131:1347–55. doi:10.1038/
jid.2010.416
42. Treumer F, Zhu K, Glaser R, Mrowietz U. Dimethylfumarate is a potent
inducer of apoptosis in human T cells. J Invest Dermatol (2003) 121:1383–8.
doi:10.1111/j.1523-1747.2003.12605.x
43. Lehmann M, Risch K, Nizze H, Lutz J, Heemann U, Volk HD, etal. Fumaric
acid esters are potent immunosuppressants: inhibition of acute and chronic
rejection in rat kidney transplantation models by methyl hydrogen fumarate.
Arch Dermatol Res (2002) 294:399–404.
44. Moed H, Stoof TJ, Boorsma DM, von Blomberg BM, Gibbs S, Bruynzeel DP,
etal. Identication of anti-inammatory drugs according to their capacity to
suppress type-1 and type-2 T cell proles. Clin Exp Allergy (2004) 34:1868–75.
doi:10.1111/j.1365-2222.2004.02124.x
45. Hestvik ALK. e double-edged sword of autoimmunity: lessons from multi-
ple sclerosis. Toxins (2010) 2:856–77. doi:10.3390/toxins2040856
46. Hoglund RA, Maghazachi AA. Multiple sclerosis and the role of immune cells.
World J Exp Med (2014) 4:27–37. doi:10.5493/wjem.v4.i3.27
47. Wang Q, Chuikov S, Taitano S, Wu Q, Rastogi A, Tuck SJ, etal. Dimethyl fuma-
rate protects neural stem/progenitor cells and neurons from oxidative damage
through Nrf2-ERK1/2 MAPK pathway. Int J Mol Sci (2015) 16:13885–907.
doi:10.3390/ijms160613885
48. Schilling S, Goelz S, Linker R, Luehder F, Gold R. Fumaric acid esters are
eective in chronic experimental autoimmune encephalomyelitis and
suppress macrophage inltration. Clin Exp Immunol (2006) 145:101–7.
doi:10.1111/j.1365-2249.2006.03094.x
49. Parodi B, Rossi S, Morando S, Cordano C, Bragoni A, Motta C, etal. Fumarates
modulate microglia activation through a novel HCAR2 signaling pathway
and rescue synaptic dysregulation in inamed CNS. Acta Neuropathol (2015)
130:279–95. doi:10.1007/s00401-015-1422-3
50. Giulian D, Corpuz M. Microglial secretion products and their impact on the
nervous system. Adv Neurol (1993) 59:315–20.
51. Loewe R, Holnthoner W, Groger M, Pillinger M, Gruber F, MechtcheriakovaD,
etal. Dimethylfumarate inhibits TNF-induced nuclear entry of NF-kappa B/
p65 in human endothelial cells. J Immunol (2002) 168:4781–7. doi:10.4049/
jimmunol.168.9.4781
52. Wierinckx A, Breve J, Mercier D, Schultzberg M, Drukarch B, Van Dam AM.
Detoxication enzyme inducers modify cytokine production in rat mixed glial
cells. J Neuroimmunol (2005) 166:132–43. doi:10.1016/j.jneuroim.2005.05.013
53. Asadullah K, Schmid H, Friedrich M, Randow F, Volk HD, Sterry W, et al.
Inuence of monomethylfumarate on monocytic cytokine formation–expla-
nation for adverse and therapeutic eects in psoriasis? Arch Dermatol Res
(1997) 289:623–30. doi:10.1007/s004030050251
54. Vandermeeren M, Janssens S, Borgers M, Geysen J. Dimethylfumarate is an
inhibitor of cytokine-induced E-selectin, VCAM-1, and ICAM-1 expression
in human endothelial cells. Biochem Biophys Res Commun (1997) 234:19–23.
doi:10.1006/bbrc.1997.6570
8
Al-Jaderi and Maghazachi New Therapeutic Modalities for Cancer
Frontiers in Immunology | www.frontiersin.org July 2016 | Volume 7 | Article 278
55. Linker RA, Lee DH, Stangel M, Gold R. Fumarates for the treatment of mul-
tiple sclerosis: potential mechanisms of action and clinical studies. Expert Rev
Neurother (2008) 8:1683–90. doi:10.1586/14737175.8.11.1683
56. Gold R, Linker RA, Stangel M. Fumaric acid and its esters: an emerging
treatment for multiple sclerosis with antioxidative mechanism of action. Clin
Immunol (2012) 142:44–8. doi:10.1016/j.clim.2011.02.017
57. Lee DH, Gold R, Linker RA. Mechanisms of oxidative damage in multi-
ple sclerosis and neurodegenerative diseases: therapeutic modulation
via fumaric acid esters. Int J Mol Sci (2012) 13:11783–803. doi:10.3390/
ijms130911783
58. Scannevin RH, Chollate S, Jung MY, Shackett M, Patel H, Bista P, et al.
Fumarates promote cytoprotection of central nervous system cells against
oxidative stress via the nuclear factor (erythroid-derived 2)-like 2 pathway.
J Pharmacol Exp er (2012) 341:274–84. doi:10.1124/jpet.111.190132
59. Itoh K, Tong KI, Yamamoto M. Molecular mechanism activating Nrf2-Keap1
pathway in regulation of adaptive response to electrophiles. Free Radic Biol
Med (2004) 36:1208–13. doi:10.1016/j.freeradbiomed.2004.02.075
60. Li W, Kong AN. Molecular mechanisms of Nrf2-mediated antioxidant
response. Mol Carcinog (2009) 48:91–104. doi:10.1002/mc.20465
61. Schimrigk S, Brune N, Hellwig K, Lukas C, Bellenberg B, Rieks M, et al.
Oral fumaric acid esters for the treatment of active multiple sclerosis: an
open-label, baseline-controlled pilot study. Eur J Neurol (2006) 13:604–10.
doi:10.1111/j.1468-1331.2006.01292.x
62. Lull ME, Block ML. Microglial activation and chronic neurodegeneration.
Neurotherapeutics (2010) 7:354–65. doi:10.1016/j.nurt.2010.05.014
63. Kunze R, Urrutia A, Homann A, Liu H, Helluy X, Pham M, et al.
Dimethyl fumarate attenuates cerebral edema formation by protecting the
blood-brain barrier integrity. Exp Neurol (2015) 266:99–111. doi:10.1016/j.
expneurol.2015.02.022
64. Haarmann A, Nehen M, Deiss A, Buttmann M. Fumaric acid esters do not
reduce inammatory NF-kappaB/p65 nuclear translocation, ICAM-1 expres-
sion and T-cell adhesiveness of human brain microvascular endothelial cells.
Int J Mol Sci (2015) 16:19086–95. doi:10.3390/ijms160819086
65. Iniaghe LO, Kra PR, Klebe DW, Omogbai EK, Zhang JH, Tang J. Dimethyl
fumarate confers neuroprotection by casein kinase 2 phosphorylation of
Nrf2 in murine intracerebral hemorrhage. Neurobiol Dis (2015) 82:349–58.
doi:10.1016/j.nbd.2015.07.001
66. Aronowski J, Hall CE. New horizons for primary intracerebral hemorrhage
treatment: experience from preclinical studies. Neurol Res (2005) 27:268–79.
doi:10.1179/016164105X25225
67. Javaid K, Rahman A, Anwar KN, Frey RS, Minshall RD, Malik AB. Tumor
necrosis factor-alpha induces early-onset endothelial adhesivity by protein
kinase Czeta-dependent activation of intercellular adhesion molecule-1. Circ
Res (2003) 92:1089–97. doi:10.1161/01.RES.0000072971.88704.CB
68. Han R, Xiao J, Zhai H, Hao J. Dimethyl fumarate attenuates experimental
autoimmune neuritis through the nuclear factor erythroid-derived 2-related
factor 2/hemoxygenase-1 pathway by altering the balance of M1/M2
macrophages. J Neuroinammation (2016) 13:97. doi:10.1186/s12974-016-
0559-x
69. Lin SX, Lisi L, Dello RC, Polak PE, Sharp A, Weinberg G, et al. e
anti-inammatory eects of dimethyl fumarate in astrocytes involve
glutathione and haem oxygenase-1. ASN Neuro (2011) 3:e00055. doi:10.1042/
AN20100033
70. Yogev O, Yogev O, Singer E, Shaulian E, Goldberg M, Fox TD, etal. Fumarase:
a mitochondrial metabolic enzyme and a cytosolic/nuclear component of the
DNA damage response. PLoS Biol (2010) 8:e1000328. doi:10.1371/journal.
pbio.1000328
71. Frohlich DA, McCabe MT, Arnold RS, Day ML. e role of Nrf2 in increased
reactive oxygen species and DNA damage in prostate tumorigenesis. Oncogene
(2008) 27:4353–62. doi:10.1038/onc.2008.79
72. Xu C, Huang MT, Shen G, Yuan X, Lin W, Khor TO, et al. Inhibition of
7,12-dimethylbenz(a)anthracene-induced skin tumorigenesis in C57BL/6
mice by sulforaphane is mediated by nuclear factor E2-related factor 2. Cancer
Res (2006) 66:8293–6. doi:10.1158/0008-5472.CAN-06-0300
73. Loewe R, Valero T, Kremling S, Pratscher B, Kunstfeld R, PehambergerH,
etal. Dimethylfumarate impairs melanoma growth and metastasis. CancerRes
(2006) 66:11888–96. doi:10.1158/0008-5472.CAN-06-2397
74. Brzozowska I, Ptak-Belowska A, Pawlik M, Pajdo R, Drozdowicz D,
KonturekSJ, etal. Mucosal strengthening activity of central and peripheral
melatonin in the mechanism of gastric defense. J Physiol Pharmacol (2009)
60(Suppl 7):47–56.
75. Herman JP, Cullinan WE. Neurocircuitry of stress: central control of the
hypothalamo-pituitary-adrenocortical axis. Trends Neurosci (1997) 20:78–84.
doi:10.1016/S0166-2236(96)10069-2
76. Shakya A, Soni UK, Rai G, Chatterjee SS, Kumar V. Gastro-protective and
anti-stress ecacies of monomethyl fumarate and a fumaria indica extract in
chronically stressed rats. Cell Mol Neurobiol (2015) 36:621–35. doi:10.1007/
s10571-015-0243-1
77. Zhu K, Mrowietz U. Enhancement of antibacterial superoxide-anion genera-
tion in human monocytes by fumaric acid esters. Arch Dermatol Res (2005)
297:170–6. doi:10.1007/s00403-005-0598-0
78. Yik JH, Hu Z, Kumari R, Christiansen BA, Haudenschild DR. Cyclin-
dependent kinase 9 inhibition protects cartilage from the catabolic eects
of proinammatory cytokines. Arthritis Rheumatol (2014) 66:1537–46.
doi:10.1002/art.38378
79. Li Y, Tang J, Hu Y. Dimethyl fumarate protection against collagen II degra-
dation. Biochem Biophys Res Commun (2014) 454:257–61. doi:10.1016/j.
bbrc.2014.10.005
80. Oh CJ, Kim JY, Choi YK, Kim HJ, Jeong JY, Bae KH, etal. Dimethylfumarate
attenuates renal brosis via NF-E2-related factor 2-mediated inhibition of
transforming growth factor-beta/Smad signaling. PLoS One (2012) 7:e45870.
doi:10.1371/journal.pone.0045870
Conict of Interest Statement: e authors declare that the research was con-
ducted in the absence of any commercial or nancial relationships that could be
construed as a potential conict of interest.
Copyright © 2016 Al-Jaderi and Maghazachi. is is an open-access article distrib-
uted under the terms of the Creative Commons Attribution License (CC BY). e
use, distribution or reproduction in other forums is permitted, provided the original
author(s) or licensor are credited and that the original publication in this journal
is cited, in accordance with accepted academic practice. No use, distribution or
reproduction is permitted which does not comply with these terms.
Content uploaded by Zaidon Salim
Author content
All content in this area was uploaded by Zaidon Salim on Oct 15, 2016
Content may be subject to copyright.