Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Inflammation ; 46(1): 453-467, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36195817

ABSTRACT

This study aimed to clarify the nephroprotective effect of dimethyl fumarate (DMF) against Di (2-ethylhexyl) phthalate (DEHP)-induced nephrotoxicity in both in vitro and in vivo models. The HEK-293 cells were exposed to different concentrations of DMF plus IC50 concentration of monoethylhexyl phthalate (MEHP) (the main metabolite of DEHP). Then, some of the oxidative stress parameters including ROS, MDA, and GSH, and cytotoxicity (MTT assay) were determined in treated cells. For in vivo evaluation, rats were divided into 7 groups (n = 6 per group). Corn oil group (gavage), DEHP group (200 mg/kg dissolved in corn oil, gavage), DMF (15, 30, and 60 mg/kg, gavage) plus DEHP (200 mg/kg) groups, DMF (60 mg/kg, gavage) alone, and vitamin E (20 mg/kg, intraperitoneal (IP)) plus DEHP (200 mg/kg) group. This treatment continued for 45 days. Then, BUN and creatinine were evaluated by a commercial kit based on the urease enzymatic method and the Jaffe method, respectively. Mitochondrial oxidative stress and mitochondrial dysfunction parameters were evaluated using appropriate reagents, and gene expression of the p65 nuclear factor kappa B (NF-κB), tumor necrosis factor alpha (TNFα), nuclear factor E2-related factor 2 (Nrf2), and heme oxygenase-1 (HO-1) were evaluated by real-time PCR method. High concentrations of DMF significantly increased cell viability, and GSH content and significantly decreased ROS and MDA levels compared with the MEHP group in HEK-293 cells. DMF (60 mg/kg) significantly decreased BUN and creatinine levels compared with the DEHP group. Mitochondrial function and mitochondrial swelling were significantly improved in DMF group (60 mg/kg) compared with the DEHP group. DMF (30 and 60 mg/kg) significantly improved MMP collapse compared with the DEHP group. DMF (30 and 60 mg/kg) significantly decreased ROS levels compared with the DEHP group in isolated kidney mitochondria. DMF (60 mg/kg) significantly decreased MDA levels and significantly increased GSH content compared with DEHP group in isolated kidney mitochondria. The mRNA expression levels of Nrf2 and HO-1 were significantly reduced in the DEHP group compared to the control group and were significantly increased in the DMF group compared to the DEHP group. p65NF-κB and TNFα mRNA expression levels were significantly increased in the DEHP group compared to the control group. However, DMF significantly decreased p65NF-κB and TNFα mRNA expression compared to the DEHP group. DMF can act as a nephroprotective agent against DEHP partly through modulation of oxidative stress, mitochondrial function, and inflammation.


Subject(s)
Diethylhexyl Phthalate , NF-kappa B , Rats , Humans , Animals , NF-kappa B/metabolism , Diethylhexyl Phthalate/toxicity , Dimethyl Fumarate/pharmacology , Dimethyl Fumarate/therapeutic use , NF-E2-Related Factor 2/metabolism , Reactive Oxygen Species/pharmacology , Heme Oxygenase-1/metabolism , Corn Oil/pharmacology , Creatinine , HEK293 Cells , Tumor Necrosis Factor-alpha/metabolism , Oxidative Stress , Signal Transduction , RNA, Messenger/metabolism
2.
Oxid Med Cell Longev ; 2020: 4678252, 2020.
Article in English | MEDLINE | ID: mdl-33123312

ABSTRACT

Nrf2 is a master regulator of antioxidant cellular defence, and agents activating the Nrf2 pathway have been tested in various diseases. However, unexpected side effects of cardiovascular nature reported for bardoxolone methyl in patients with type 2 diabetes mellitus and stage 4 chronic kidney disease (the BEACON trial) still have not been fully explained. Here, we aimed to characterize the effects of bardoxolone methyl compared with other Nrf2 activators-dimethyl fumarate and L-sulforaphane-on human microvascular endothelium. Endothelial toxicity, bioenergetics, mitochondrial membrane potential, endothelin-1 (ET-1) release, endothelial permeability, Nrf2 expression, and ROS production were assessed in human microvascular endothelial cells (HMEC-1) incubated for 3 and 24 hours with 100 nM-5 µM of either bardoxolone methyl, dimethyl fumarate, or L-sulforaphane. Three-hour incubation with bardoxolone methyl (100 nM-5 µM), although not toxic to endothelial cells, significantly affected endothelial bioenergetics by decreasing mitochondrial membrane potential (concentrations ≥ 3 µM), decreasing spare respiratory capacity (concentrations ≥ 1 µM), and increasing proton leak (concentrations ≥ 500 nM), while dimethyl fumarate and L-sulforaphane did not exert such actions. Bardoxolone methyl at concentrations ≥ 3 µM also decreased cellular viability and induced necrosis and apoptosis in the endothelium upon 24-hour incubation. In turn, endothelin-1 decreased permeability in endothelial cells in picomolar range, while bardoxolone methyl decreased ET-1 release and increased endothelial permeability even after short-term (3 hours) incubation. In conclusion, despite that all three Nrf2 activators exerted some beneficial effects on the endothelium, as evidenced by a decrease in ROS production, bardoxolone methyl, the most potent Nrf2 activator among the tested compounds, displayed a distinct endothelial profile of activity comprising detrimental effects on mitochondria and cellular viability and suppression of endothelial ET-1 release possibly interfering with ET-1-dependent local regulation of endothelial permeability.


Subject(s)
Endothelin-1/metabolism , Oleanolic Acid/analogs & derivatives , Permeability/drug effects , Cell Line , Cell Survival/drug effects , Dimethyl Fumarate/pharmacology , Down-Regulation/drug effects , Endothelial Cells/cytology , Endothelial Cells/metabolism , Gene Expression/drug effects , Humans , Isothiocyanates/pharmacology , Membrane Potential, Mitochondrial/drug effects , Microvessels/cytology , Mitochondria/drug effects , Mitochondria/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Oleanolic Acid/pharmacology , Reactive Oxygen Species/metabolism , Sulfoxides/pharmacology
3.
Biochem Biophys Res Commun ; 532(3): 475-481, 2020 11 12.
Article in English | MEDLINE | ID: mdl-32892947

ABSTRACT

Dimethyl fumarate (DMF) has emerged as a first-line treatment for the relapsing-remitting multiple sclerosis (RRMS) subtype. It is hypothesized that DMF has anti-inflammatory and antioxidant effects although mechanisms are not fully understood. This study used RNA-seq to profile gene expression responses to DMF in cultured astrocytes. Responses were compared with those of isosorbide di-(methyl fumarate) (IDMF), a newly designed fumarate that may partially replicate DMF activity with fewer adverse effects. Both compounds altered the expression of MS-associated genes, including those near MS susceptibility loci and genes dysregulated in MS patient astrocytes. The shared DMF/IDMF transcriptome response involved altered expression of antioxidant genes (e.g., HMOX1) and genes linked to extracellular matrix integrity (TIMP3, MMP9) and migration of pro-inflammatory cells into CNS (CCL2). IDMF-specific transcriptome responses included down-regulation of mitotic genes associated with a proliferative reactive astrocyte phenotype (ICAM1) and repression of genes encoding NF-kappaB subunits (NFKB2, RELA, RELB) and NF-kappaB targets (NCAPG, CXCL1, OAS3). Overall, these results identify astrocyte-centered mechanisms that may contribute to the established efficacy of DMF as an RRMS treatment. Furthermore, our findings support a rationale for further studies of IDMF as a novel fumarate, which may have unique suppressive effects on astrocyte reactivity and glial scar formation. [200 words].


Subject(s)
Astrocytes/drug effects , Dimethyl Fumarate/analogs & derivatives , Astrocytes/metabolism , Cells, Cultured , Dimethyl Fumarate/pharmacology , Down-Regulation/drug effects , Drug Evaluation, Preclinical , Genetic Predisposition to Disease , Humans , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase Inhibitors/pharmacology , Mitosis/drug effects , Mitosis/genetics , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Phenotype , Protein Biosynthesis/drug effects , Transcriptome/drug effects
4.
Mol Neurobiol ; 57(1): 105-117, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31494826

ABSTRACT

Recently, dimethyl fumarate (DMF) and Korean red ginseng (ginseng), based on their purported antioxidative and anti-inflammatory properties, have exhibited protective potential in various neurological conditions. Their effects on cerebral ischemia and underlying mechanisms remain inconclusive; however, increasing evidence indicates the involvement of the transcriptional factor Nrf2. This study evaluated the preventive effects of DMF and ginseng on hippocampal neuronal damage following hypoxia-ischemia (HI) and assessed the contributions of reactive gliosis and the Nrf2 pathway. Adult wild type (WT) and Nrf2-/- mice were pretreated with DMF or ginseng for 7 days prior to HI. At 24 h after HI, DMF or ginseng significantly reduced infarct volume (52.5 ± 12.3% and 47.8 ± 10.7%), brain edema (61.5 ± 17.4% and 39.3 ± 12.8%), and hippocampal CA1 neuronal degeneration, and induced expressions of Nrf2 target proteins in WT, but not Nrf2-/-, mice. Such hippocampal neuroprotective benefits were also observed at 6 h and 7 days after HI. The dynamic attenuation of reactive gliosis in microglia and astrocytes correlated well with this sustained neuroprotection in an Nrf2-dependent manner. In both early and late stages of HI, astrocytic dysfunctions in extracellular glutamate clearance and water transport, as indicated by glutamine synthetase and aquaporin 4, were also attenuated after HI in WT, but not Nrf2-/-, mice treated with DMF or ginseng. Together, DMF and ginseng confer robust and prolonged Nrf2-dependent neuroprotection against ischemic hippocampal damage. The salutary Nrf2-dependent attenuation of reactive gliosis may contribute to this neuroprotection, offering new insight into the cellular basis of an Nrf2-targeting strategy for stroke prevention or treatment.


Subject(s)
Antioxidants , Brain Ischemia , NF-E2-Related Factor 2 , Neuroprotective Agents , Panax , Animals , Antioxidants/pharmacology , Astrocytes/drug effects , Astrocytes/metabolism , Brain Ischemia/drug therapy , Dimethyl Fumarate/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Mice, Knockout , Microglia/drug effects , Microglia/metabolism , Neuroprotection/drug effects , Neuroprotective Agents/pharmacology , NF-E2-Related Factor 2/metabolism
5.
Free Radic Biol Med ; 131: 98-114, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30458277

ABSTRACT

The transcriptional factor Nrf2, a master regulator of oxidative stress and inflammation that are tightly linked to the development and progression of cerebral ischemia pathology, plays a vital role in inducing the endogenous neuroprotective process. Here, hypoxic-ischemia (HI) was performed in adult Nrf2 knockout and wildtype mice that were orally pretreated either with standardized Korean red ginseng extract (Ginseng) or dimethyl fumarate (DMF), two candidate Nrf2 inducers, to determine whether the putative protection was through an Nrf2-dependent mechanism involving the attenuation of reactive gliosis. Results show that Nrf2 target cytoprotective genes were distinctly elevated following HI. Pretreatment with Ginseng or DMF elicited robust neuroprotection against the deterioration of acute cerebral ischemia damage in an Nrf2-dependent manner as revealed by the reductions of neurological deficits score, infarct volume and brain edema, as well as enhanced expression levels of Nrf2 target antioxidant proteins and anti-inflammation mediators. In both ischemic striatum and cortex, the dynamic pattern of attenuated reactive gliosis in astrocytes and microglia, including affected astrocytic dysfunction in glutamate metabolism and water homeostasis, correlated well with the Nrf2-dependent neuroprotection by Ginseng or DMF. Furthermore, such neuroprotective benefits extended to the late phase of ischemic brain damage after HI, as evidenced by improvements in neurobehavioral outcomes, infarct volume and brain edema. Overall, pretreatment with Ginseng or DMF identically attenuates reactive gliosis and confers long-lasting neuroprotective efficacy against ischemic brain damage through an Nrf2-dependent mechanism. This study also provides new insight into the profitable contribution of reactive gliosis in the Nrf2-dependent neuroprotection in acute brain injury.


Subject(s)
Dimethyl Fumarate/pharmacology , Gliosis/drug therapy , Hypoxia-Ischemia, Brain/drug therapy , NF-E2-Related Factor 2/genetics , Neuroprotective Agents/pharmacology , Panax/chemistry , Animals , Aquaporin 4/genetics , Aquaporin 4/metabolism , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Carotid Arteries/surgery , Cerebral Cortex/blood supply , Cerebral Cortex/metabolism , Cerebral Cortex/physiopathology , Cerebrovascular Disorders/surgery , Corpus Striatum/blood supply , Corpus Striatum/metabolism , Corpus Striatum/physiopathology , Disease Models, Animal , Gene Expression Regulation , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Gliosis/genetics , Gliosis/metabolism , Gliosis/physiopathology , Humans , Hypoxia-Ischemia, Brain/genetics , Hypoxia-Ischemia, Brain/metabolism , Hypoxia-Ischemia, Brain/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Microglia/drug effects , Microglia/metabolism , Microglia/pathology , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/deficiency , Plant Extracts/pharmacology
6.
Nat Commun ; 9(1): 4344, 2018 10 19.
Article in English | MEDLINE | ID: mdl-30341347

ABSTRACT

Dimethyl fumarate (DMF) has been applied for decades in the treatment of psoriasis and now also multiple sclerosis. However, the mechanism of action has remained obscure and involves high dose over long time of this small, reactive compound implicating many potential targets. Based on a 1.9 Å resolution crystal structure of the C-terminal kinase domain of the mouse p90 Ribosomal S6 Kinase 2 (RSK2) inhibited by DMF we describe a central binding site in RSKs and the closely related Mitogen and Stress-activated Kinases (MSKs). DMF reacts covalently as a Michael acceptor to a conserved cysteine residue in the αF-helix of RSK/MSKs. Binding of DMF prevents the activation loop of the kinase from engaging substrate, and stabilizes an auto-inhibitory αL-helix, thus pointing to an effective, allosteric mechanism of kinase inhibition. The biochemical and cell biological characteristics of DMF inhibition of RSK/MSKs are consistent with the clinical protocols of DMF treatment.


Subject(s)
Dimethyl Fumarate/pharmacology , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Animals , Binding Sites , Binding, Competitive , Crystallography, X-Ray , Cysteine/chemistry , Dimethyl Fumarate/chemistry , HEK293 Cells , Humans , Mice , Models, Molecular , Mutation , Ribosomal Protein S6 Kinases, 90-kDa/chemistry , Ribosomal Protein S6 Kinases, 90-kDa/physiology
7.
Eur J Neurol ; 25(3): 584-e36, 2018 03.
Article in English | MEDLINE | ID: mdl-29316038

ABSTRACT

BACKGROUND AND PURPOSE: Dimethyl fumarate (DMF) is an oral treatment for relapsing-remitting multiple sclerosis (MS) with anti-inflammatory and possible neuroprotective properties. Its effect on white matter and gray matter pathology is still not fully understood. The aim of the study was to characterize the effect of DMF on normal-appearing white matter (NAWM) and thalamic pathology longitudinally. METHODS: In this observational, longitudinal, 24-month magnetic resonance imaging study, 75 patients with relapsing-remitting MS treated with DMF and 40 age- and sex-matched healthy individuals were enrolled. Regional diffusion tensor imaging metrics and tract-based spatial statistics analyses were used to assess differences between groups. Mean diffusivity, axial diffusivity, radial diffusivity and fractional anisotropy were measured in the thalamus and NAWM. Baseline differences and changes over time were evaluated within and between study groups. RESULTS: At baseline, patients with MS showed significantly increased diffusivity and decreased fractional anisotropy in the thalamus (P < 0.001 for mean diffusivity, axial diffusivity and radial diffusivity) and NAWM (all P < 0.016) compared with healthy individuals. No significant within-group difference was found in diffusion tensor imaging measures over 24 months in either group. Healthy individuals showed a significantly greater rate of increased diffusivity parameters in the thalamus and NAWM compared with patients with MS, over 24 months (P < 0.05). CONCLUSIONS: The lack of changes in diffusion tensor imaging metrics in patients with MS over 24 months possibly indicates a neuroprotective role of DMF. These findings provide additional evidence of the beneficial effect of DMF on MS-related pathology.


Subject(s)
Dimethyl Fumarate/pharmacology , Immunosuppressive Agents/pharmacology , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Multiple Sclerosis, Relapsing-Remitting/pathology , Neuroprotective Agents/pharmacology , Thalamus/pathology , White Matter/pathology , Adult , Diffusion Tensor Imaging , Female , Humans , Longitudinal Studies , Male , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/diagnostic imaging , Single-Blind Method , Thalamus/diagnostic imaging , White Matter/diagnostic imaging
8.
Mult Scler Relat Disord ; 11: 32-39, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28104252

ABSTRACT

INTRODUCTION: Pre-clinical drug discovery for multiple sclerosis (MS) is a labor intensive activity to perform in rodent models. This is owing to the long duration of disease induction and observation of treatment effects in an experimental autoimmune encephalomyelitis (EAE) model. We propose a novel adult zebrafish based model which offers a quick and simple protocol that can used to screen candidates as a step between in vitro experiments and rodent studies. The experiments conducted for this manuscript were to standardize a suitable model of EAE in adult zebrafish and validate it using known modulators. METHODS: The EAE model was developed by disease induction with myelin oligodendrocyte glycoprotein - 35-55 (MOG) and observation of survival, clinical signs and body weight changes. We have validated this model using fingolimod. We have further performed detailed validation using dimethyl fumarate, dexamethasone and SR1001, which are known modulators of rodent EAE. RESULTS: The immunization dose for the disease induction was observed to be 0.6mg/ml of MOG in CFA (Complete Freund's adjuvant), injected subcutaneously (s.c.) near spinal vertebrae. In the validation study with fingolimod, we have demonstrated the modulation of disease symptoms, which were also confirmed by histopathological evaluation. Furthermore, detailed validation with three other known drugs showed that our observations concur with those reported in conventional rodent models. DISCUSSION: We have standardized and validated the adult zebrafish EAE model. This model can help get a quick idea of in vivo activity of drugs in a week using very low quantities of candidate compounds. Further work will be required to define this model particularly as it is found that zebrafish may not express a MOG homologue.


Subject(s)
Drug Evaluation, Preclinical/methods , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Zebrafish , Animals , Dexamethasone/pharmacology , Dimethyl Fumarate/pharmacology , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Fingolimod Hydrochloride/pharmacology , Immunologic Factors/pharmacology , Phenotype , Sulfonamides/pharmacology , Thiazoles/pharmacology , Video Recording
9.
Pharmacol Rep ; 68(2): 404-14, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26922546

ABSTRACT

Several lines of evidence have demonstrated the potential biomedical applications of fumaric acid (FA) and its ester derivatives against many human disease conditions. Fumaric acid esters (FAEs) have been licensed for the systemic treatment of the immune-mediated disease psoriasis. Biogen Idec Inc. announced about the safety and efficacy of the formulation FAE (BG-12) for treating RRMS (relapsing-remitting multiple sclerosis). Another FAE formulation DMF (dimethyl fumarate) was found to be capable of reduction in inflammatory cardiac conditions, such as autoimmune myocarditis and ischemia and reperfusion. DMF has also been reported to be effective as a potential neuroprotectant against the HIV-associated neurocognitive disorders (HAND). Many in vivo studies carried out on rat and mice models indicated inhibitory effects of fumaric acid on carcinogenesis of different origins. Moreover, FAEs has emerged as an important matrix ingredient in the fabrication of biodegradable scaffolds for tissue engineering applications. Drug delivery vehicles composed of FAEs have shown promising results in delivering some leading drug molecules. Apart from these specific applications and findings, many more studies on FAEs have revealed new therapeutic potentials with the scope of clinical applications. However, until now, this scattered vital information has not been written into a collective account and analyzed for minute details. The aim of this paper is to review the advancement made in the biomedical application of FA and FAEs and to focus on the clinical investigation and molecular interpretation of the beneficial effects of FA and FAEs.


Subject(s)
Esters/pharmacology , Esters/therapeutic use , Fumarates/pharmacology , Fumarates/therapeutic use , Animals , Dimethyl Fumarate/pharmacology , Dimethyl Fumarate/therapeutic use , Humans , Multiple Sclerosis, Relapsing-Remitting/drug therapy , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Psoriasis/drug therapy
10.
Exp Dermatol ; 25(2): 124-30, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26513635

ABSTRACT

Fumaric acid esters, dimethyl fumarate (DMF) in particular, have been established for the therapy of psoriasis and, more recently, multiple sclerosis. In the light of therapy-limiting dose-dependent side effects, such as gastrointestinal irritation, reducing the effective doses of FAE is a worthwhile goal. In search of strategies to maintain the anti-inflammatory activity of DMF at reduced concentrations, we found that NF-κB inhibition augmented key anti-inflammatory effects of DMF in two complementary experimental settings in vitro. At non-toxic concentrations, both proteasome inhibition with bortezomib as well as blocking NF-κB activation through KINK-1, a small molecule inhibitor of IKKß-profoundly enhanced DMF-dependent inhibition of nuclear NF-κB translocation in TNFα-stimulated human endothelial cells. This resulted in significant and selective co-operative down-regulation of endothelial adhesion molecules crucial for leucocyte extravasation, namely E-selectin (CD62E), VCAM-1 (CD106) and ICAM-1 (CD54), on both mRNA and protein levels. Functionally, these molecular changes led to synergistically decreased rolling and firm adhesion of human lymphocytes on TNF-activated endothelial cells, as demonstrated in a dynamic flow chamber system. If our in vitro findings can be translated into clinical settings, it is conceivable that anti-inflammatory effects of DMF can be achieved with lower doses than currently used, thus potentially reducing unwanted side effects.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Dimethyl Fumarate/pharmacology , Endothelial Cells/drug effects , NF-kappa B/antagonists & inhibitors , Bortezomib/pharmacology , Cell Adhesion , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Drug Synergism , E-Selectin/biosynthesis , E-Selectin/genetics , Endothelial Cells/metabolism , Gene Expression Regulation/drug effects , Hemorheology , Human Umbilical Vein Endothelial Cells , Humans , In Vitro Techniques , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/genetics , Leukocytes/cytology , Oxazines/pharmacology , Proteasome Endopeptidase Complex/drug effects , Pyridines/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Transcription, Genetic/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Vascular Cell Adhesion Molecule-1/biosynthesis , Vascular Cell Adhesion Molecule-1/genetics
11.
J Biol Chem ; 291(7): 3639-47, 2016 Feb 12.
Article in English | MEDLINE | ID: mdl-26683377

ABSTRACT

In breast tumors, activation of the nuclear factor κB (NFκB) pathway promotes survival, migration, invasion, angiogenesis, stem cell-like properties, and resistance to therapy--all phenotypes of aggressive disease where therapy options remain limited. Adding an anti-inflammatory/anti-NFκB agent to breast cancer treatment would be beneficial, but no such drug is approved as either a monotherapy or adjuvant therapy. To address this need, we examined whether dimethyl fumarate (DMF), an anti-inflammatory drug already in clinical use for multiple sclerosis, can inhibit the NFκB pathway. We found that DMF effectively blocks NFκB activity in multiple breast cancer cell lines and abrogates NFκB-dependent mammosphere formation, indicating that DMF has anti-cancer stem cell properties. In addition, DMF inhibits cell proliferation and significantly impairs xenograft tumor growth. Mechanistically, DMF prevents p65 nuclear translocation and attenuates its DNA binding activity but has no effect on upstream proteins in the NFκB pathway. Dimethyl succinate, the inactive analog of DMF that lacks the electrophilic double bond of fumarate, is unable to inhibit NFκB activity. Also, the cell-permeable thiol N-acetyl l-cysteine, reverses DMF inhibition of the NFκB pathway, supporting the notion that the electrophile, DMF, acts via covalent modification. To determine whether DMF interacts directly with p65, we synthesized and used a novel chemical probe of DMF by incorporating an alkyne functionality and found that DMF covalently modifies p65, with cysteine 38 being essential for the activity of DMF. These results establish DMF as an NFκB inhibitor with anti-tumor activity that may add therapeutic value in the treatment of aggressive breast cancers.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Breast Neoplasms/drug therapy , Dimethyl Fumarate/pharmacology , NF-kappa B/antagonists & inhibitors , Neoplasm Proteins/antagonists & inhibitors , Signal Transduction/drug effects , Transcription Factor RelA/antagonists & inhibitors , Active Transport, Cell Nucleus/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cysteine/chemistry , Dimethyl Fumarate/chemistry , Dimethyl Fumarate/therapeutic use , Female , Gene Expression Regulation, Neoplastic/drug effects , Genes, Reporter/drug effects , Humans , Mice, Nude , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Random Allocation , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL