ArticlePDF AvailableLiterature Review

Utilization of Dimethyl Fumarate and Related Molecules for Treatment of Multiple Sclerosis, Cancer, and Other Diseases

Frontiers
Frontiers in Immunology
Authors:

Abstract and Figures

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.
Content may be 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: 30May2016
Accepted: 06July2016
Published: 22July2016
Citation:
Al-JaderiZ and MaghazachiAA
(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, teriunomide,
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 inammatory skin diseases,
such as psoriasis. e benecial eects of this medication corroborated with regulating CD4+ 1
cell dierentiation. In clinical trials, it showed positive benets for MS patients by lowering risk of
relapse and reducing the number of brain lesions (16).
e mechanism of action is not fully known. Aer 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 ~12min inside the body (8). Aer absorption, DMF is rapidly hydrolyzed by esterases
to monomethyl fumarate (MMF) (9), which has a short half-life of 36h. 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). Figure1 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 13–21min (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
eective 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 inammatory mediators. Studies have shown that Nrf2
and NF-κB pathways have inhibitory inuence 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 signicantly 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 aect circulating CD56bright NK cells,
CD14+ monocytes, or myeloid DCs. However, DMF-treated
patients had signicantly fewer CD56dim NK cells when compared
with healthy controls (17). A clinical study of 35 RRMS patients
at baseline, 3months, 6months, and 12months aer initiation
of DMF treatment shows that total leukocyte and lymphocyte
counts diminished aer 6months, whereas aer 12months 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
signicantly 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 eects 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 inammatory cytokines IL-6
and IL-12. Furthermore, DMF activated type II DCs, which have
anti-inammatory eects and suppressed type I DCs, which are
inammatory (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 inammatory 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 shiing of
the immune system toward inammatory 1/17 response.
ese results support previous study showing that inammatory
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 inammatory 1/17 proliferation and may enhance
the anti-inammatory 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 aected the polariza-
tion but not maturation of monocyte-derived DCs, resulting in
downregulating 1 lymphocyte responses (31). In vitro study
for the eects of MMF on DCs dierentiation shows that MMF
inhibited monocyte-derived DCs dierentiation 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 aected IFN-γ production by naive T cells, whereas the
production of IL-4 and IL-10 was not inuenced by MMF (32).
Another study demonstrates that DMF inhibited DCs matura-
tion by reducing the production of the inammatory cytokines
IL-6 and IL-12 as well as the expression of MHC class II, CD80,
and CD86 (27). Furthermore, immature DCs activated fewer
Tcells characterized by low IFN-γ and IL-17 production (27).
In contrast, de Jong etal. (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 inltration of 1/17 cells into the skin
(3436). DMF is utilized to treat psoriasis in European countries
for more than 30years. 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 tracking and consequently, inhibited the release of
pro-inammatory cytokines, such as IL-12, IL-23, and TNF,
whereas the release of anti-inammatory 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 aect the immune
system. e immunohistochemical studies of psoriatic plaques
indicate that DMF has several anti-inammatory eects via a
number of pathways, leading to reduction in the levels of several
inammatory 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 inammatory 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-inammatory 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 inammatory 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 inammatory T cells in the CNS triggers recruitment
of more T cells, Bcells, dendritic cells, microglia cells, and NK
cells (46). Due to the progressive neurodegenerative nature of
MS, therapeutic modalities that exhibit direct neuroprotective
eects are needed. A phase 3 clinical trial study of 2667 RRMS
patients demonstrates the ecacy 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 signicantly 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.
Invivo 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 detoxication
capabilities of DMF or MMF reduced the production and release
of inammatory 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 detoxication 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 inam-
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 invitro (53, 54).
is is correlated with activating Nrf2 (5558), 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 dierent 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 inammation,
demyelination, and axonal lose. Treatment of EAE mice with
DMF reduced macrophage-induced inammation in the spinal
cord (48). DMF suppressed 1 and 17 cell dierentiation as
well as expression of pro-inammatory 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-inammatory 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-inammatory type to neuroprotective eect (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-inammatory
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 aer 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 aer
treatment with DMF for 6h (54).
In vitro human umbilical vein endothelium examination
indicates that DMF or MMF modulated pro-inammatory
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 inammatory
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 eect of fumaric acid esters on the inltration
of inammatory T cells into the CNS (64). It is also determined
that DMF ameliorated inammation, 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
inammation and oxidative stress, which occur aer 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 inammation and aer 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 decits by
ameliorating inammatory cell inltration and demyelination of
sciatic nerves. In addition, DMF treatment decreased the level of
pro-inammatory M1 macrophages, while increasing the number
of anti-inammatory 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
inammatory 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 eects 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 insitu 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 eect of innate defense against microorgan-
ism. Treatment of monocytes and neutrophils with DMF aer
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 eects (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 immunodecient diseases. Tab l e 1 shows the current
knowledge regarding the eects 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, 2527,
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, etal.
Ecacy 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, eectiveness, and side eects. Curr Neurol Neurosci Rep
(2013) 13:394–8. doi:10.1007/s11910-013-0394-8
7. Werdenberg D, Joshi R, Wolram 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: scientic 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, VanFR .
Eects 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, etal.
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. Specic 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 Neuroinamm (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, etal.
Hydroxycarboxylic acid receptor 2 mediates dimethyl fumarate’s protective
eect 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 suering 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, SuterM,
et al. Dendritic cells directly trigger NK cell functions: cross-talk relevant
in innate anti-tumor immune responses invivo. Nat Med (1999) 5:405–11.
doi:10.1038/7403
25. Ghoreschi K, Bruck J, Kellerer C, Deng C, Peng H, Rothfuss O, etal. 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 dierentia-
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, PapenfussTL,
etal. 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, Coman 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, Laont S, Powrie F. E-cadherin marks a subset of inamma-
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, etal. Anti-CD40-induced
inammatory 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,
etal. Monomethylfumarate aects 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. Eects of monomethylfumarate on dendritic cell dier-
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, Pfeilschier JM, etal.
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-Bea M, Medina MA, Quesada AR.
Dimethylfumarate inhibits angiogenesis invitro and invivo: a possible role
for its antipsoriatic eect? 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, etal. 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,
etal. Identication of anti-inammatory drugs according to their capacity to
suppress type-1 and type-2 T cell proles. 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, etal. 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
eective in chronic experimental autoimmune encephalomyelitis and
suppress macrophage inltration. 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, etal. Fumarates
modulate microglia activation through a novel HCAR2 signaling pathway
and rescue synaptic dysregulation in inamed 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, MechtcheriakovaD,
etal. 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.
Inuence of monomethylfumarate on monocytic cytokine formation–expla-
nation for adverse and therapeutic eects 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, Homann 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 inammatory 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 Neuroinammation (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-inammatory eects 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, etal. 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, PehambergerH,
etal. Dimethylfumarate impairs melanoma growth and metastasis. CancerRes
(2006) 66:11888–96. doi:10.1158/0008-5472.CAN-06-2397
74. Brzozowska I, Ptak-Belowska A, Pawlik M, Pajdo R, Drozdowicz D,
KonturekSJ, etal. 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 ecacies 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 eects
of proinammatory 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, etal. 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
Conict 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 conict 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.
... Moreover, DMF exerts influence over immune cells, which are involved in the inflammatory response, and which are crucial for nerve myelination [19,[27][28][29]. Its impact on Schwann cells is evident in the preservation and regeneration of myelinated fibers, further contributing to improved outcomes [18,30]. ...
Article
Full-text available
Spinal cord injury results in significant motor and sensory loss. In the experimental ventral root avulsion (VRA) model, the ventral (motor) roots are disconnected from the spinal cord surface, disrupting contact between spinal motoneurons and muscle fibers. Axotomized motoneurons typically degenerate within two to three weeks after avulsion, the situation being exacerbated by an increased glial response and chronic inflammation. Nevertheless, root reimplantation has been observed to stimulate regenerative potential in some motoneurons, serving as a model for CNS/PNS regeneration. We hypothesized that a combination of neuroprotective and immunomodulatory therapies is capable of enhancing regenerative responses following nerve root injury and repair. A heterologous fibrin biopolymer (HFB) was used for surgical repair; dimethyl fumarate (DMF) was used for neuroprotection and immunomodulation; and adipose tissue-derived mesenchymal stem cells (AT-MSCs) were used as a source of trophic factors and cytokines that may further enhance neuronal survival. Thus, adult female Lewis rats underwent unilateral VRA of the L4–L6 roots, followed by reimplantation with HFB, AT-MSCs transplantation, and daily DMF treatment for four weeks, with a 12-week postoperative survival period. An evaluation of the results focused on light microscopy, qRT-PCR, and the Catwalk motor function recovery system. Data were analyzed using one-way or two-way ANOVA (p < 0.05). The results indicate that the combined therapy resulted in a reduced glial response and a 70% improvement in behavioral motor recovery. Overall, the data support the potential of combined regenerative approaches after spinal cord root injury.
... Dimethyl fumarate, a fumaric acid ester (α, β-unsaturated carboxylic acid ester), is an FDA and European Medicines Agency (EMA) approved drug, which is effective in the treatment of relapsing/remitting multiple sclerosis, and is considered a type of indirect antioxidant, as it promotes the activation and stabilization of nuclear factor erythroid 2-related factor 2 (Nrf2) [15], [16]. Dimethyl fumarate enters the body orally and dissociates in the intestine, where it subsequently interacts with immune cells in the blood circulation and crosses the blood-brain barrier into the central nervous system [17]. There are different works involving hepatoprotection, neuroprotection and anti-arthritis protection using daily oral doses between 25 and 100 mg/kg of dimethyl fumarate for several weeks, showing an Nrf2-mediated cytoprotective effect in Wistar rats [18]. ...
Preprint
Full-text available
Purpose Docetaxel is a taxane antineoplastic widely used against different types of cancer. However, its efficacy is limited mainly by its myelotoxicity and peripheral neuropathy with neutropenia and nociceptive alterations as the main clinical signs, respectively. These adverse effects undermine the quality of life of patients leading them to treatment withdrawal. In this study, we set up a unique preclinical scheme for induction of both effects associated with docetaxel administration, in such a way that we can evaluate them in the same animal and carry out future protection trials. Methods Four docetaxel administration schemes were tested varying dose and dosage. Four days after the last dose, behavioral/sensory paw pressure, tail pressure and hot plate tests were conducted. Next, euthanasia was performed, and blood was obtained for total cell count and other toxicological markers. Once the scheme that better showed significant alterations in both nociception and neutropenia was chosen, joint administration with 100 mg/kg/day oral dimethyl fumarate was carried out to evaluate its protective effect. Results A scheme with six doses (5 mg/kg) of docetaxel administered weekly was chosen for protection trials. Dimethyl fumarate showed protection in nociceptive tests compared to the damage group. However, it did not show protection against neutropenia. Conclusion The confirmed experimental model is clinically representative as it was designed for rats through equivalent data obtained from clinical assays. It was useful to evaluate the protective potential of dimethyl fumarate, showing how it could attenuate docetaxel-induced peripheral neuropathy, but not neutropenia.
... One of the drugs previously described to promote effectiveness of oncolytic virotherapy is Dimethyl Fumarate (DMF). DMF is an approved drug for psoriasis taken as an oral therapy, and since 2013 DMF has also been approved for relapsing multiple sclerosis (18). In the context of cancer treatment, DMF has demonstrated anti-tumor properties by reducing tumor growth and metastasis (19,20). ...
Article
Full-text available
Immunotherapy and specifically oncolytic virotherapy has emerged as a promising option for cancer patients, with oncolytic herpes simplex virus-1 (oHSV-1) expressing granulocyte macrophage colony stimulating factor being the first OV to be approved by the FDA for treatment of melanoma. However, not all cancers are sensitive and responsive to oncolytic viruses (OVs). Our group has demonstrated that fumaric and maleic acid esters (FMAEs) are very effective in sensitizing cancer cells to OV infection. Of note, these FMAEs include dimethyl fumarate (DMF, also known as Tecfidera®), an approved treatment for multiple sclerosis and psoriasis. This study aimed to assess the efficacy of DMF in combination with oncolytic HSV-1 in preclinical cancer models. We demonstrate herewith that pre-treatment with DMF or other FMAEs leads to a significant increase in viral growth of oHSV-1 in several cancer cell lines, including melanoma, while decreasing cell viability. Additionally, DMF was able to enhance ex vivo oHSV-1 infection of mouse-derived tumor cores as well as human patient tumor samples but not normal tissue. We further reveal that the increased viral spread and oncolysis of the combination therapy occurs via inhibition of type I IFN production and response. Finally, we demonstrate that DMF in combination with oHSV-1 can improve therapeutic outcomes in aggressive syngeneic murine cancer models. In sum, this study demonstrates the synergistic potential of two approved therapies for clinical evaluation in cancer patients.
... Moreover, it was demonstrated that the expression of Nrf2 and its downstream genes HO-1 and NQO1, at the protein and gene levels, significantly increased in the hippocampus following seizure induction, suggesting that the Nrf2 signaling pathway was activated in the hippocampus after a seizure [27]. DMF was approved by the FDA for the treatment of multiple sclerosis in 2013, and its application in the treatment of various tumors, inflammatory bowel disease, and intracranial hemorrhage has been reported in subsequent studies [28,29]. Although its exact mechanism of action is unknown, orally administered DMF is thought to exert its therapeutic (e.g., neuroprotective, anti-inflammatory) effects via activation of the Nrf2 antioxidant response pathway [30,31]. ...
Article
Full-text available
Background Epilepsy affects over 65 million people worldwide and significantly burdens patients, caregivers, and society. Drug-resistant epilepsy occurs in approximately 30% of patients and growing evidence indicates that oxidative stress contributes to the development of such epilepsies. Activation of the Nrf2 pathway, which is involved in cellular defense, offers a potential strategy for reducing oxidative stress and epilepsy treatment. Dimethyl fumarate (DMF), an Nrf2 activator, exhibits antioxidant and anti-inflammatory effects and is used to treat multiple sclerosis. Methods The expression of Nrf2 and its related genes in vehicle or DMF treated rats were determined via RT-PCR and Western blot analysis. Neuronal cell death was evaluated by immunohistochemical staining. The effects of DMF in preventing the onset of epilepsy and modifying the disease were investigated in the kainic acid-induced status epilepticus model of temporal lobe epilepsy in rats. The open field, elevated plus maze and T-Maze spontaneous alteration tests were used for behavioral assessments. Results We demonstrate that administration of DMF following status epilepticus increased Nrf2 activity, attenuated status epilepticus-induced neuronal cell death, and decreased seizure frequency and the total number of seizures compared to vehicle-treated animals. Moreover, DMF treatment reversed epilepsy-induced behavioral deficits in the treated rats. Moreover, DMF treatment even when initiated well after the diagnosis of epilepsy, reduced symptomatic seizures long after the drug was eliminated from the body. Conclusions Taken together, these findings suggest that DMF, through the activation of Nrf2, has the potential to serve as a therapeutic target for preventing epileptogenesis and modifying epilepsy.
... DMF increases Nrf2 pathway activity, a transcription factor known as a master regulator of antioxidant abilities [48][49][50]. Furthermore, DMF and its biologically active metabolite, monomethyl fumarate (MMF), can cross the blood-brain barrier (BBB) at pharmacologically relevant levels, as indicated by direct effects on tissue and cells for inducing antioxidant effects [51][52][53]. Here, we investigate the effect of DMF on the neuroinflammatory and oxidative stress response at both a sub-chronic (7 weeks) and a chronic time point (16 weeks post-implantation) and MEA recording performance across acute (1-5 weeks), sub-chronic (6-11 weeks), and chronic (12-16 weeks) neuroinflammatory phases. ...
Article
Full-text available
Intracortical microelectrode arrays (MEAs) can be used in a range of applications, from basic neuroscience research to providing an intimate interface with the brain as part of a brain-computer interface (BCI) system aimed at restoring function for people living with neurological disorders or injuries. Unfortunately, MEAs tend to fail prematurely, leading to a loss in functionality for many applications. An important contributing factor in MEA failure is oxidative stress resulting from chronically inflammatory-activated microglia and macrophages releasing reactive oxygen species (ROS) around the implant site. Antioxidants offer a means for mitigating oxidative stress and improving tissue health and MEA performance. Here, we investigate using the clinically available antioxidant dimethyl fumarate (DMF) to reduce the neuroinflammatory response and improve MEA performance in a rat MEA model. Daily treatment of DMF for 16 weeks resulted in a significant improvement in the recording capabilities of MEA devices during the sub-chronic (Weeks 5–11) phase (42% active electrode yield vs. 35% for control). However, these sub-chronic improvements were lost in the chronic implantation phase, as a more exacerbated neuroinflammatory response occurs in DMF-treated animals by 16 weeks post-implantation. Yet, neuroinflammation was indiscriminate between treatment and control groups during the sub-chronic phase. Although worse for chronic use, a temporary improvement (<12 weeks) in MEA performance is meaningful. Providing short-term improvement to MEA devices using DMF can allow for improved use for limited-duration studies. Further efforts should be taken to explore the mechanism behind a worsened neuroinflammatory response at the 16-week time point for DMF-treated animals and assess its usefulness for specific applications.
Article
Full-text available
Dimethyl fumarate is a widely known therapeutic agent with anti‐inflammatory properties for psoriasis and multiple sclerosis. Despite the current attempts to use dimethyl fumarate for treating various inflammatory diseases, its effects on endometriosis have not been previously reported. Endometriosis is a genital disease that causes various health problems in women, and treatment methods targeting the inflammatory environment are being attempted. Therefore, it is hypothesized that dimethyl fumarate has therapeutic effects on endometriosis through its anti‐inflammatory effects. Dimethyl fumarate exerted remarkable effects on cellular mechanisms, including reactive oxygen species production, activation of mitogen‑activated protein kinase signals, loss of mitochondrial function, and disruption of calcium ion homeostasis in the immortalized human ovarian endometrial stromal cells. In an endometriosis mouse model, dimethyl fumarate downregulated cell cycle‐related genes and induced inhibitory effects on endometriosis lesion growth. In particular, the immune cell population and expression of inflammatory cytokines such as IL‐1β, IL‐6, and IL‐10 are regulated by dimethyl fumarate. These results support its potential as a therapeutic agent to control the excessive inflammatory environment in patients with endometriosis. This study identifies for the first time that dimethyl fumarate, which is already in clinical use, can be used to treat endometriosis.
Article
Full-text available
Introduction Intracranial aneurysm (IA) is a common cerebrovascular disease. Considering the risks and benefits of surgery, a significant proportion of patients with unruptured IA (UIA) choose conservative observation. Previous studies suggest that inflammation of aneurysm wall is a high-risk factor of rupture. Dimethyl fumarate (DMF) acts as an anti-inflammatory agent by activating nuclear factor erythroid 2-related factor 2 (Nrf2) and other pathways. Animal experiments found DMF reduces the formation and rupture of IAs. In this study, DMF will be evaluated for its ability to reduce inflammation of the aneurysm wall in high-resolution vessel wall imaging. Methods and analysis This is a multi-centre, randomised, controlled, double-blind clinical trial. Three hospitals will enrol a total of 60 patients who have UIA with enhanced wall. Participants will be assigned randomly in a 1:1 proportion, taking either 240 mg DMF or placebo orally every day for 6 months. As the main result, aneurysm wall enhancement will be measured by the signal intensity after 6 months of DMF treatment. Secondary endpoints include morphological changes of aneurysms and factors associated with inflammation. This study will provide prospective data on the reduction of UIA wall inflammation by DMF. Ethics and dissemination This study has been approved by Medical Ethics Committee of the Beijing Tiantan Hospital, Capital Medical University (approval no: KY2022-064-02). We plan to disseminate our research findings through peer-reviewed journal publication and relevant academic conferences. Trial registration number NCT05959759.
Article
Full-text available
Neurodegenerative and neuropsychiatric disorders are two broad categories of neurological disorders characterized by progressive impairments in movement and cognitive functions within the central and peripheral nervous systems, and have emerged as a significant cause of mortality. Oxidative stress, neuroinflammation, and neurotransmitter imbalances are recognized as prominent pathogenic factors contributing to cognitive deficits and neurobehavioral anomalies. Consequently, preventing neurodegenerative and neuropsychiatric diseases has surfaced as a pivotal challenge in contemporary public health. This review explores the investigation of neurodegenerative and neuropsychiatric disorders using both synthetic and natural bioactive compounds. A central focus lies on melatonin, a neuroregulatory hormone secreted by the pineal gland in response to light-dark cycles. Melatonin, an amphiphilic molecule, assumes multifaceted roles, including scavenging free radicals, modulating energy metabolism, and synchronizing circadian rhythms. Noteworthy for its robust antioxidant and antiapoptotic properties, melatonin exhibits diverse neuroprotective effects. The inherent attributes of melatonin position it as a potential key player in the pathophysiology of neurological disorders. Preclinical and clinical studies have demonstrated melatonin’s efficacy in alleviating neuropathological symptoms across neurodegenerative and neuropsychiatric conditions (depression, schizophrenia, bipolar disorder, and autism spectrum disorder). The documented neuroprotective prowess of melatonin introduces novel therapeutic avenues for addressing neurodegenerative and psychiatric disorders. This comprehensive review encompasses many of melatonin’s applications in treating diverse brain disorders. Despite the strides made, realizing melatonin’s full neuroprotective potential necessitates further rigorous clinical investigations. By unravelling the extended neuroprotective benefits of melatonin, future studies promise to deepen our understanding and augment the therapeutic implications against neurological deficits.
Article
Full-text available
Background Guillain–Barré syndrome (GBS) is an acute, post-infectious, immune-mediated, demyelinating disease of peripheral nerves and nerve roots. Dimethyl fumarate (DMF), a fumaric acid ester, exhibits various biological activities, including multiple immunomodulatory and neuroprotective effects. However, the potential mechanism underlying the effect of DMF in GBS animal model experimental autoimmune neuritis (EAN) is unclear. Methods Using EAN, an established GBS model, we investigated the effect of DMF by assessing clinical score, histological staining and electrophysiological studies. Then, we further explored the potential mechanism by Western blot analysis, flow cytometry, fluorescence immunohistochemistry, PCR, and ELISA analysis. The Mann–Whitney U test was used to compare differences between control group and treatment groups where appropriate. ResultsDMF treatment reduced the neurological deficits by ameliorating inflammatory cell infiltration and demyelination of sciatic nerves. In addition, DMF treatment decreased the level of pro-inflammatory M1 macrophages while increasing the number of anti-inflammatory M2 macrophages in the spleens and sciatic nerves of EAN rats. In RAW 264.7, a shift in macrophage polarization from M1 to M2 phenotype was demonstrated to be depended on DMF application. In sciatic nerves, DMF treatment elevated the level of the antioxidant transcription factor nuclear factor erythroid-derived 2-related factor 2 (Nrf2) and its target gene hemoxygenase-1 (HO-1) which could facilitate macrophage polarization toward M2 type. Moreover, DMF improved the inflammatory milieu in spleens of EAN rats, characterized by downregulation of messenger RNA (mRNA) of IFN-γ, TNF-α, IL-6, and IL-17 and upregulation of mRNA level of IL-4 and IL-10. Conclusions Taken together, our data demonstrate that DMF can effectively suppress EAN, and the mechanism involves altering the balance of M1/M2 macrophages and attenuating inflammation.
Article
Full-text available
Experimental autoimmune encephalomyelitis (EAE) is a CD4⁺ T cell mediated inflammatory demyelinating disease that is induced in mice by administration of peptides derived from myelin proteins. We developed EAE in SJL mice by administration of PLP139-151 peptide. The effect of treating these mice with 1α,25-Dihydroxyvitamin D₃ (vitamin D₃), or with monomethyl fumarate (MMF) was then examined. We observed that both vitamin D₃ and MMF inhibited and/or prevented EAE in these mice. These findings were corroborated with isolating natural killer (NK) cells from vitamin D₃-treated or MMF-treated EAE mice that lysed immature or mature dendritic cells. The results support and extend other findings indicating that an important mechanism of action for drugs used to treat multiple sclerosis (MS) is to enhance NK cell lysis of dendritic cells.
Article
Full-text available
Background: Dimethyl fumarate (DMF) alters the phenotype of circulating immune cells and causes lymphopenia in a subpopulation of treated multiple sclerosis (MS) patients. Objective: To phenotypically characterize circulating leukocytes in DMF-treated MS patients. Methods: Cross-sectional observational comparisons of peripheral blood from DMF-treated MS patients (n = 17 lymphopenic and n = 24 non-lymphopenic), untreated MS patients (n = 17) and healthy controls (n = 23); immunophenotyped using flow cytometry. Longitudinal samples were analyzed for 13 DMF-treated patients. Results: Lymphopenic DMF-treated patients had significantly fewer circulating CD8(+) and CD4(+) T cells, CD56(dim) natural killer (NK) cells, CD19(+) B cells and plasmacytoid dendritic cells when compared to controls. CXCR3(+) and CCR6(+) expression was disproportionately reduced among CD4(+) T cells, while the proportion of T-regulatory (T-reg) cells was unchanged. DMF did not affect circulating CD56(hi) NKcells, monocytes or myeloid dendritic cells. Whether lymphopenic or not, DMF-treated patients had a lower proportion of circulating central and effector memory T cells and concomitant expansion of naïve T cells compared to the controls. Conclusions: DMF shifts the immunophenotypes of circulating T cells, causing a reduction of memory cells and a relative expansion of naïve cells, regardless of the absolute lymphocyte count. This may represent one mechanism of action of the drug. Lymphopenic patients had a disproportionate loss of CD8(+) T-cells, which may affect their immunocompetence.
Article
Full-text available
Multiple sclerosis (MS) is the most common multifocal inflammatory demyelinating disease of the central nervous system (CNS). Due to the progressive neurodegenerative nature of MS, developing treatments that exhibit direct neuroprotective effects are needed. Tecfidera™ (BG-12) is an oral formulation of the fumaric acid esters (FAE), containing the active metabolite dimethyl fumarate (DMF). Although BG-12 showed remarkable efficacy in lowering relapse rates in clinical trials, its mechanism of action in MS is not yet well understood. In this study, we reported the potential neuroprotective effects of dimethyl fumarate (DMF) on mouse and rat neural stem/progenitor cells (NPCs) and neurons. We found that DMF increased the frequency of the multipotent neurospheres and the survival of NPCs following oxidative stress with hydrogen peroxide (H2O2) treatment. In addition, utilizing the reactive oxygen species (ROS) assay, we showed that DMF reduced ROS production induced by H2O2. DMF also decreased oxidative stress-induced apoptosis. Using motor neuron survival assay, DMF significantly promoted survival of motor neurons under oxidative stress. We further analyzed the expression of oxidative stress-induced genes in the NPC cultures and showed that DMF increased the expression of transcription factor nuclear factor-erythroid 2-related factor 2 (Nrf2) at both levels of RNA and protein. Furthermore, we demonstrated the involvement of Nrf2-ERK1/2 MAPK pathway in DMF-mediated neuroprotection. Finally, we utilized SuperArray gene screen technology to identify additional anti-oxidative stress genes (Gstp1, Sod2, Nqo1, Srxn1, Fth1). Our data suggests that analysis of anti-oxidative stress mechanisms may yield further insights into new targets for treatment of multiple sclerosis (MS).
Article
Full-text available
Dimethyl fumarate (DMF) is approved for disease-modifying treatment of patients with relapsing-remitting multiple sclerosis. Animal experiments suggested that part of its therapeutic effect is due to a reduction of T-cell infiltration of the central nervous system (CNS) by uncertain mechanisms. Here we evaluated whether DMF and its primary metabolite monomethyl fumarate (MMF) modulate pro-inflammatory intracellular signaling and T-cell adhesiveness of nonimmortalized single donor human brain microvascular endothelial cells at low passages. Neither DMF nor MMF at concentrations of 10 or 50 µM blocked the IL-1β-induced nuclear translocation of NF-κB/p65, whereas the higher concentration of DMF inhibited the nuclear entry of p65 in human umbilical vein endothelium cultured in parallel. DMF and MMF also did not alter the IL-1β-stimulated activation of p38 MAPK in brain endothelium. Furthermore, neither DMF nor MMF reduced the basal or IL-1β-inducible expression of ICAM-1. In accordance, both fumaric acid esters did not reduce the adhesion of activated Jurkat T cells to brain endothelium under basal or inflammatory conditions. Therefore, brain endothelial cells probably do not directly mediate a potential blocking effect of fumaric acid esters on the inflammatory infiltration of the CNS by T cells.
Article
Full-text available
Results of the very first experiments conducted to evaluate therapeutic potentials of a fumarate containing Fumaria indica extract and of fairly low daily oral doses of monomethyl fumarate for prevention of chronic unavoidable foot-shock stress-induced gastric ulcers, and possible involvement of diverse neuro-hormonal and oxidative process in their stress response desensitizing effects are reported and discussed in this article. Preventive effects of 21 daily oral 60, 120, and 240 mg/kg doses of a standardized 50 % methanolic F. indica extract (MFI) and 1.25, 2.50, and 5.00 mg/kg/day of pure monomethyl fumarate (MMF) were compared in rats subjected to one hour daily unavoidable foot-shocks. A pharmaceutically well-standardized Withania somnifera (WS) root extract was used as a reference herbal anti-stress agent in all experiments. Effects of the treatments on stress-induced alterations in body weight, adrenal and spleen weights, gastric ulcer and ulcer index, weight of glandular stomach, protective mucosal glycoprotein content, cellular proliferation, oxidative stress on stomach fundus, and brain tissues of male rats were quantified. Other parameters quantified were plasma corticosterone levels, brain monoamine levels, and expressions of the cytokines TNF-α, IL-10, and IL-1β in blood and brain of stressed and treated rats. Most but not every observed stress-induced anomalies were suppressed or completely prevented by both MFI and pure MMF treatments in dose-dependent manner. Qualitatively, the observed activity profiles of both of them were similar to those of WS dose tested. These results reveal that both MFI and MMF are potent gastro-protective agents against chronic unavoidable stress-induced ulcers and strongly suggest that they act as regulators or modulators of monoamine, corticosterone, and cytokine homeostasis.
Article
Full-text available
Dimethyl fumarate (DMF), recently approved as an oral immunomodulatory treatment for relapsing-remitting multiple sclerosis (MS), metabolizes to monomethyl fumarate (MMF) which crosses the blood–brain barrier and has demonstrated neuroprotective effects in experimental studies. We postulated that MMF exerts neuroprotective effects through modulation of microglia activation, a critical component of the neuroinflammatory cascade that occurs in neurodegenerative diseases such as MS. To ascertain our hypothesis and define the mechanistic pathways involved in the modulating effect of fumarates, we used real-time PCR and biochemical assays to assess changes in the molecular and functional phenotype of microglia, quantitative Western blotting to monitor activation of postulated pathway components, and ex vivo whole-cell patch clamp recording of excitatory post-synaptic currents in corticostriatal slices from mice with experimental autoimmune encephalomyelitis (EAE), a model for MS, to study synaptic transmission. We show that exposure to MMF switches the molecular and functional phenotype of activated microglia from classically activated, pro-inflammatory type to alternatively activated, neuroprotective one, through activation of the hydroxycarboxylic acid receptor 2 (HCAR2). We validate a downstream pathway mediated through the AMPK–Sirt1 axis resulting in deacetylation, and thereby inhibition, of NF-κB and, consequently, of secretion of pro-inflammatory molecules. We demonstrate through ex vivo monitoring of spontaneous glutamate-mediated excitatory post-synaptic currents of single neurons in corticostriatal slices from EAE mice that the neuroprotective effect of DMF was exerted on neurons at pre-synaptic terminals by modulating glutamate release. By exposing control slices to untreated and MMF-treated activated microglia, we confirm the modulating effect of MMF on microglia function and, thereby, its indirect neuroprotective effect at post-synaptic level. These findings, whereby DMF-induced activation of a new HCAR2-dependent pathway on microglia leads to the modulation of neuroinflammation and restores synaptic alterations occurring in EAE, represent a possible novel mechanism of action for DMF in MS. Electronic supplementary material The online version of this article (doi:10.1007/s00401-015-1422-3) contains supplementary material, which is available to authorized users.
Article
Full-text available
To evaluate the influence of dimethyl fumarate (DMF, Tecfidera) treatment of multiple sclerosis (MS) on leukocyte and lymphocyte subsets. Peripheral blood leukocyte and lymphocyte subsets, including CD3(+), CD4(+), and CD8(+) T cells; CD19(+) B cells; and CD56(+) natural killer (NK) cells, were obtained at baseline and monitored at 3 months, 6 months, and 12 months after initiation of DMF treatment. Total leukocyte and lymphocyte counts diminished after 6 months of DMF therapy. At 12 months, lymphocyte counts had decreased by 50.1% (p < 0.0001) and were below the lower limit of normal (LLN) in one-half of patients. CD3(+) T lymphocyte counts fell by 44.2% (p < 0.0001). Among subsets, CD8(+) T cell counts declined by 54.6% (p < 0.0001), whereas CD4(+) T cell counts decreased by 39.2% (p = 0.0006). This disproportionate reduction of CD8(+) T cells relative to CD4(+) T cells was significant (p = 0.007) and was reflected by a 35.5% increase in the CD4/CD8 ratio (p = 0.007). A majority of CD8(+) T cell counts, but not CD4(+) T cell counts, were below the LLN even when total lymphocyte counts were greater than 500 cells/μL. CD19(+) B cell counts were reduced by 37.5% (p = 0.035). Eosinophil levels decreased by 54.1% (p = 0.006), whereas levels of neutrophils, monocytes, basophils, and NK cells were not significantly altered. Subsets of peripheral blood leukocytes and lymphocytes are differentially affected by DMF treatment of MS. Reduction of CD8(+) T cells is more pronounced than that of CD4(+) T cells. These findings may have implications for cell-mediated antiviral immunity during DMF treatment.
Article
Edema formation, inflammation and increased blood-brain barrier permeability contribute to poor outcomes after intracerebral hemorrhage (ICH). This study examined the therapeutic effect of Dimethyl fumarate (DMF), a fumaric acid ester that activates nuclear factor erythroid-2 related factor 2 (Nrf2) and Nrf2 heterodimerization effector musculo-aponeurotic fibrosacoma-G (MAFG) in a murine ICH model. Male CD-1 mice (n=176) were subjected to intrastriatal infusion of bacterial collagenase (n=120), autologous blood (n=18) or sham surgery (n=30). After ICH, animals either received vehicle, Dimethyl fumarate (10mg or 100mg/kg) or casein kinase 2 inhibitor (E)-3-(2,3,4,5-tetrabromophenyl)acrylic acid (TBCA). Thirty-eight mice also received scrambled siRNA or MAFG siRNA 24hours before ICH. Brain water content and neurological function were evaluated. Dimethyl fumarate reduced Evans blue extravasation, decreased brain water content, and improved neurological deficits at 24 and 72hours after ICH. Casein kinase 2 inhibitor TBCA and MAFG siRNA prevented the effect of Dimethyl fumarate on brain edema and neurological function. After ICH, ICAM-1 levels increased and Casein kinase 2 levels decreased. Dimethyl fumarate reduced ICAM-1 but enhanced Casein kinase 2 levels. Again, Casein kinase 2 inhibitor TBCA and MAFG siRNA abolished the effect of Dimethyl fumarate on ICAM-1 and Casein kinase 2. Dimethyl fumarate preserved pNrf2 and MAFG expression in the nuclear lysate after ICH and the effect of Dimethyl fumarate was abolished by Casein kinase 2 inhibitor TBCA and MAFG siRNA. Dimethyl fumarate reduced microglia activation in peri-hematoma areas after ICH. The protective effect of Dimethyl fumarate on brain edema and neurological function was repeated in a blood injection mouse model. Dimethyl fumarate ameliorated inflammation, reduced blood barrier permeability, and improved neurological outcomes by Casein kinase 2 and Nrf2 signaling pathways after experimental ICH in mice. Copyright © 2015. Published by Elsevier Inc.