1
|
Li Y, Wang Y, Wang S, Zhu H. Effects of Oxidative Stress Gene Protein, Expression, and DNA Methylation on Multiple Sclerosis: A Multi-Omics Mendelian Randomized Study. Brain Behav 2025; 15:e70606. [PMID: 40444654 PMCID: PMC12123450 DOI: 10.1002/brb3.70606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/04/2025] [Accepted: 03/31/2025] [Indexed: 06/02/2025] Open
Abstract
BACKGROUND Oxidative stress (OS) is linked to the development of multiple sclerosis (MS), but the causal relationship in terms of genetic pathophysiology remains ambiguous. We employed Mendelian randomization (MR) and colocalization analysis to explore the relationship between OS genes and MS, utilizing an integrative multi-omics approach. METHODS We obtained data from a genome-wide association study (GWAS) of MS from the International Multiple Sclerosis Genetics Consortium (Discovery phase) and the FinnGen study (Replication phase). Mendelian randomization analyses were conducted using summary data to evaluate the association between molecular features of OS-related genes and MS. Additional colocalization analyses were undertaken to ascertain whether the identified signal pairs shared causal genetic variants. RESULTS Integration of multi-omics data, including mQTL-eQTL and eQTL-pQTL, revealed that the STAT3 gene is associated with MS, supported by Level 1 evidence. The CR1 gene shows an association with MS risk, evidenced by Level 3 support. Methylation at cg24718015 and cg17833746 in the STAT3 gene correlates with reduced expression of STAT3. At the protein level, high circulating levels of STAT3 are inversely associated with MS risk (OR: 0.43, 95% CI, 0.33-0.54). Elevated levels of TNFRSF1A are also linked with a decreased risk of MS (OR: 0.21; 95% CI, 0.12-0.37), while higher levels of CR1 are positively associated with an increased risk of MS (OR: 1.17; 95% CI, 1.08-1.27). CONCLUSION This study identifies specific OS genes that are associated with MS and enhances our understanding of its pathogenesis.
Collapse
Affiliation(s)
- Yang Li
- Department of NeurologyThe First Hospital of Jilin UniversityChangchunJilinChina
| | - Yushi Wang
- Department of NeurologyThe First Hospital of Jilin UniversityChangchunJilinChina
| | - Shuning Wang
- Department of NeurologyThe First Hospital of Jilin UniversityChangchunJilinChina
| | - Hui Zhu
- Department of NeurologyThe First Hospital of Jilin UniversityChangchunJilinChina
| |
Collapse
|
2
|
Balzani MFV, Coser LDO, de Oliveira ALR. Immunomodulation by 4-Hydroxy-TEMPO (TEMPOL) and Dimethyl Fumarate (DMF) After Ventral Root Crush (VRC) in C57BL/6J Mice: A Flow Cytometry Analysis. BIOLOGY 2025; 14:473. [PMID: 40427663 PMCID: PMC12109085 DOI: 10.3390/biology14050473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/10/2025] [Accepted: 04/11/2025] [Indexed: 05/29/2025]
Abstract
Spinal motor nerve root lesions can happen after avulsion or crush, generating acute motoneuron death and synaptic loss, consequently, causing motor and sensory dysfunctions. Local response is mediated by astroglial and microglial cells, giving rise to a pro-inflammatory profile. TEMPOL and DMF are drugs that have been studied in our laboratory after spinal cord nerve root injuries and have demonstrated significant results in terms of neuroprotection and immunomodulation, decreasing the inflammation process. In the present work, a flow cytometry approach was used to evaluate cellular responses to injury and immunomodulation. For this, injured animals received TEMPOL, DMF or vehicle once a day for 7, 14 or 28 days of treatment. Flow cytometry multiparametric analysis allowed the quantification of different pro- and anti-inflammatory glial, macrophage and lymphocyte markers. Contrasting with the vehicle treated counterpart, TEMPOL and DMF led to downregulation of pro-inflammatory cytokines in astrocytes and microglia subpopulations, but did not show significant results in increasing anti-inflammatory phenotypes. As for macrophage and lymphocyte subpopulations, both treatments showed a balance between pro- and anti-inflammatory phenotypes. Therefore, it was concluded that both drugs exhibit immunomodulatory action, contributing to a pro-regenerative profile in the tissue.
Collapse
Affiliation(s)
- Maria Fernanda Vannucci Balzani
- Department of Structural and Functional Biology, Laboratory of Nerve Regeneration, Institute of Biology, University of Campinas (UNICAMP), Campinas 13083-862, SP, Brazil; (M.F.V.B.); (L.d.O.C.)
| | - Lilian de Oliveira Coser
- Department of Structural and Functional Biology, Laboratory of Nerve Regeneration, Institute of Biology, University of Campinas (UNICAMP), Campinas 13083-862, SP, Brazil; (M.F.V.B.); (L.d.O.C.)
| | - Alexandre Leite Rodrigues de Oliveira
- Department of Structural and Functional Biology, Laboratory of Nerve Regeneration, Institute of Biology, University of Campinas (UNICAMP), Campinas 13083-862, SP, Brazil; (M.F.V.B.); (L.d.O.C.)
- Center for Gender-Specific Biology and Medicine (CGBM)–Brazil, University of Campinas (UNICAMP), Campinas 13083-862, SP, Brazil
| |
Collapse
|
3
|
Rościszewska-Żukowska I, Biesiadecki M, Mołoń M, Rożek A, Bartosik-Psujek H, Galiniak S. Elevated Oxidative Stress in Patients with Coexisting Multiple Sclerosis and Migraine: A Cross-Sectional Study. Antioxidants (Basel) 2025; 14:511. [PMID: 40427393 PMCID: PMC12108162 DOI: 10.3390/antiox14050511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Revised: 04/12/2025] [Accepted: 04/22/2025] [Indexed: 05/29/2025] Open
Abstract
One potential association that is gaining increasing attention is the link between multiple sclerosis (MS) and migraine, which are suggested to frequently coexist in young patients. This is the first study to analyze the levels of multiple markers of oxidative stress in sociodemographically similar groups of patients with migraine, MS, and both MS and migraine. A single cross-sectional study was conducted at the Department of Neurology, Rzeszów University. The study included 110 participants, comprising 26 healthy controls, 24 subjects with migraines, 30 with MS, and 30 with both MS and migraine. Oxidative stress markers were measured in patients' serum. Patients with MS and migraines had statistically elevated levels of 3-nitrotyrosine, Amadori products, 4-hydroxy-nonenal, and oxidative damage to amino acids. Moreover, we observed reduced levels of thiol groups and total antioxidant capacity in the serum of patients with MS and migraines compared to healthy controls. The co-occurrence of migraines in MS leads to greater oxidative stress than MS alone. The impact of chronic oxidative stress on both MS and migraines may exacerbate symptoms and deteriorate the quality of life.
Collapse
Affiliation(s)
| | - Marek Biesiadecki
- Faculty of Medicine, University of Rzeszów, Warzywna 1a, 35-310 Rzeszów, Poland; (M.B.); (H.B.-P.)
| | - Mateusz Mołoń
- Faculty of Biology and Nature Protection, University of Rzeszów, Zelwerowicza 4, 35-601 Rzeszów, Poland; (M.M.); (A.R.)
| | - Aleksandra Rożek
- Faculty of Biology and Nature Protection, University of Rzeszów, Zelwerowicza 4, 35-601 Rzeszów, Poland; (M.M.); (A.R.)
| | - Halina Bartosik-Psujek
- Faculty of Medicine, University of Rzeszów, Warzywna 1a, 35-310 Rzeszów, Poland; (M.B.); (H.B.-P.)
| | - Sabina Galiniak
- Faculty of Medicine, University of Rzeszów, Warzywna 1a, 35-310 Rzeszów, Poland; (M.B.); (H.B.-P.)
| |
Collapse
|
4
|
Gold R, Barnett M, Chan A, Feng H, Fujihara K, Giovannoni G, Montalbán X, Shi FD, Tintoré M, Xue Q, Yang C, Zhou H. Clinical use of dimethyl fumarate in multiple sclerosis treatment: an update to include China, using a modified Delphi method. Ther Adv Neurol Disord 2023; 16:17562864231180734. [PMID: 37465201 PMCID: PMC10350766 DOI: 10.1177/17562864231180734] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 05/20/2023] [Indexed: 07/20/2023] Open
Abstract
Dimethyl fumarate (DMF) is a widely used oral disease-modifying therapy for multiple sclerosis (MS). Its efficacy and safety profiles are supported by over a decade of experience. Differences exist between Asia and Europe/United States in the prevalence and characteristics of MS; most data for DMF are derived from populations outside Asia. DMF was recently (2021) approved for use in China. The objectives of this review were to evaluate the evidence for DMF's profile, to provide an update to healthcare providers on current knowledge surrounding its use and to assess the relevance of existing data to use in China. This study used a modified Delphi method based on the insights of a scientific Steering Committee (SC), with a structured literature review conducted to assess the data of DMF. The literature review covered all papers in English (from 01 January 2011 to 21 February 2022) that include 'dimethyl fumarate' and 'multiple sclerosis', and their MeSH terms, on PubMed, supplemented by EMBASE and Citeline searches. Papers were categorized by topic and assessed for relevance and quality, before being used to formulate statements summarizing the literature on each subject. SC members voted on/revised statements, requiring ⩾80% agreement and ⩽10% disagreement for inclusion. Statements not reaching this level were discussed further until agreement was reached or until there was agreement to remove the statement. A total of 1030 papers were retrieved and used to formulate the statements and evidence summaries considered by the SC members. A total of 45 statements were agreed by the SC members. The findings support the positive efficacy and safety profile of DMF in treating patients with MS. Limited Chinese patient data are an ongoing consideration; however, based on current evidence, the statements are considered applicable to both the global and Chinese populations. DMF is a valuable addition to address unmet MS treatment needs in China. Registration: Not applicable.
Collapse
Affiliation(s)
- Ralf Gold
- Department of Neurology, Ruhr University Bochum, Bochum 44791, Germany
| | - Michael Barnett
- Brain and Mind Centre, University of Sydney and Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Andrew Chan
- Department of Neurology, Inselspital (Bern University Hospital), University of Bern, Bern, Switzerland
| | - Huiyu Feng
- Department of Neurology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Kazuo Fujihara
- Department of Multiple Sclerosis Therapeutics, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Gavin Giovannoni
- Department of Neurology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Xavier Montalbán
- Neurology Department, Multiple Sclerosis Center of Catalonia (Cemcat), Vall d’Hebron University Hospital, Barcelona, Spain
| | - Fu-Dong Shi
- Department of Neurology, Institute of Neuroimmunology, Tianjin Medical University General Hospital, Tianjin, China
| | - Mar Tintoré
- Neurology Department, Multiple Sclerosis Center of Catalonia (Cemcat), Vall d’Hebron University Hospital, Barcelona, Spain
| | - Qun Xue
- Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chunsheng Yang
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Hongyu Zhou
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
5
|
Möhle L, Stefan K, Bascuñana P, Brackhan M, Brüning T, Eiriz I, El Menuawy A, van Genderen S, Santos-García I, Górska AM, Villa M, Wu J, Stefan SM, Pahnke J. ABC Transporter C1 Prevents Dimethyl Fumarate from Targeting Alzheimer's Disease. BIOLOGY 2023; 12:932. [PMID: 37508364 PMCID: PMC10376064 DOI: 10.3390/biology12070932] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/14/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023]
Abstract
Alzheimer's disease (AD), the leading cause of dementia, is a growing health issue with very limited treatment options. To meet the need for novel therapeutics, existing drugs with additional preferred pharmacological profiles could be recruited. This strategy is known as 'drug repurposing'. Here, we describe dimethyl fumarate (DMF), a drug approved to treat multiple sclerosis (MS), to be tested as a candidate for other brain diseases. We used an APP-transgenic model (APPtg) of senile β-amyloidosis mice to further investigate the potential of DMF as a novel AD therapeutic. We treated male and female APPtg mice through drinking water at late stages of β-amyloid (Aβ) deposition. We found that DMF treatment did not result in modulating effects on Aβ deposition at this stage. Interestingly, we found that glutathione-modified DMF interacts with the ATP-binding cassette transporter ABCC1, an important gatekeeper at the blood-brain and blood-plexus barriers and a key player for Aβ export from the brain. Our findings suggest that ABCC1 prevents the effects of DMF, which makes DMF unsuitable as a novel therapeutic drug against AD. The discovered effects of ABCC1 also have implications for DMF treatment of multiple sclerosis.
Collapse
Affiliation(s)
- Luisa Möhle
- Department of Pathology, Section of Neuropathology/Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Sognsvannsveien 20, 0372 Oslo, Norway
| | - Katja Stefan
- Department of Pathology, Section of Neuropathology/Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Sognsvannsveien 20, 0372 Oslo, Norway
| | - Pablo Bascuñana
- Department of Pathology, Section of Neuropathology/Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Sognsvannsveien 20, 0372 Oslo, Norway
| | - Mirjam Brackhan
- Department of Pathology, Section of Neuropathology/Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Sognsvannsveien 20, 0372 Oslo, Norway
| | - Thomas Brüning
- Department of Pathology, Section of Neuropathology/Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Sognsvannsveien 20, 0372 Oslo, Norway
| | - Ivan Eiriz
- Department of Pathology, Section of Neuropathology/Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Sognsvannsveien 20, 0372 Oslo, Norway
| | - Ahmed El Menuawy
- Department of Pathology, Section of Neuropathology/Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Sognsvannsveien 20, 0372 Oslo, Norway
| | - Sylvie van Genderen
- Department of Pathology, Section of Neuropathology/Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Sognsvannsveien 20, 0372 Oslo, Norway
| | - Irene Santos-García
- Department of Pathology, Section of Neuropathology/Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Sognsvannsveien 20, 0372 Oslo, Norway
| | - Anna Maria Górska
- Department of Pathology, Section of Neuropathology/Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Sognsvannsveien 20, 0372 Oslo, Norway
| | - María Villa
- Department of Pathology, Section of Neuropathology/Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Sognsvannsveien 20, 0372 Oslo, Norway
| | - Jingyun Wu
- Department of Pathology, Section of Neuropathology/Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Sognsvannsveien 20, 0372 Oslo, Norway
| | - Sven Marcel Stefan
- Department of Pathology, Section of Neuropathology/Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Sognsvannsveien 20, 0372 Oslo, Norway
- Pahnke Lab (Drug Development and Chemical Biology), Lübeck Institute of Experimental Dermatology (LIED), University of Lübeck (UzL) and University Medical Center Schleswig-Holstein (UKSH), Ratzeburger Allee 160, 23538 Lübeck, Germany
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Jens Pahnke
- Department of Pathology, Section of Neuropathology/Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Sognsvannsveien 20, 0372 Oslo, Norway
- Pahnke Lab (Drug Development and Chemical Biology), Lübeck Institute of Experimental Dermatology (LIED), University of Lübeck (UzL) and University Medical Center Schleswig-Holstein (UKSH), Ratzeburger Allee 160, 23538 Lübeck, Germany
- Department of Pharmacology, Faculty of Medicine, University of Latvia, Jelgavas iela 3, 1004 Rīga, Latvia
- Department of Neurobiology, The Georg S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
6
|
Evidence for novel cell defense mechanisms sustained by dimethyl fumarate in multiple sclerosis patients: the HuR/SOD2 cascade. Mult Scler Relat Disord 2022; 68:104197. [PMID: 36270254 DOI: 10.1016/j.msard.2022.104197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 09/22/2022] [Accepted: 09/27/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Dimethyl fumarate (DMF) is an effective treatment for relapsing remitting Multiple Sclerosis (MS) and its mechanisms of action encompass immunomodulatory and cytoprotective effects. Despite DMF is known to activate the Nrf2 pathway, Nrf2-independent mechanisms have been also reported and new insights on the underlying molecular mechanisms are still emerging including transcriptional and post-transcriptional events. At this regard, we focused on a small family of RNA-binding proteins, the ELAV-like proteins, that play a pivotal role in post-transcriptional mechanisms and are involved in the pathogenesis of several psychiatric and neurologic disorders. HuR, the ubiquitously expressed member of the family, is implicated in many cellular functions, including survival, inflammation and proper functioning of the immune system. We previously documented the potential entanglement of HuR in MS pathogenesis. In the present work, we explored HuR protein levels in peripheral blood mononuclear cells (PBMCs) from MS patients before and after DMF treatment compared to healthy controls (HC). Considering that HuR may act on various targets, playing a protective role against oxidative stress, our main goals were to evaluate whether manganese-dependent superoxide dismutase transcript (SOD2) could represent a new molecular target of HuR and to study the potential influence of DMF treatment on this interaction. METHODS PBMCs from 20 patients with MS and 20 frequency-matched HC by sex and age were used to evaluate HuR, MnSOD (the protein coded by SOD2) and Nrf2 protein content by Western blot, before and after 12 months of DMF treatment. Immunoprecipitation experiments coupled with RNA extraction in PBMCs were performed to explore whether SOD2 mRNA could be physically bound by HuR and whether the expression of MnSOD protein could be affected by 12 months of DMF treatment. RESULTS In PBMCs, HuR protein binds SOD2 transcript in HC and in MS patients naïve to disease modifying treatment. The expression of MnSOD protein is positively affected by 12 months of DMF treatment. PBMCs from MS patients have a lower HuR and MnSOD protein content compared to matched HC (HuR: p<0.01, MnSOD: p<0.01). Of interest, 12 months of DMF treatment in MS patients restores the amount of both HuR protein and MnSOD enzyme to the levels observed in HC. We also confirmed that Nrf2 is an HuR target, and we report that its levels are significantly increased in MS patients naïve to disease modifying treatment and remain elevated following DMF administration. CONCLUSION SOD2 transcript is a new target of HuR protein. DMF induces an increased expression of HuR protein, which ultimately interacts more strongly with SOD2 transcript promoting the expression of this antioxidant protein. The activation of this molecular cascade can constitute an additional tool that the cells can exploit to counteract the oxidative stress associated with MS development, and can account for the multifaceted molecular mechanisms underlying DMF efficacy in MS.
Collapse
|
7
|
Sun X, Suo X, Xia X, Yu C, Dou Y. Dimethyl Fumarate is a Potential Therapeutic Option for Alzheimer's Disease. J Alzheimers Dis 2021; 85:443-456. [PMID: 34842188 DOI: 10.3233/jad-215074] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Dimethyl fumarate (DMF) has been approved for clinical treatment of multiple sclerosis based on its antioxidant and anti-inflammatory effects by activating the Nrf2 pathway. Since both oxidative stress and inflammation are involved in Alzheimer's disease (AD), DMF is a potential therapeutic option for AD. OBJECTIVE This study aims to test the therapeutic effects of DMF on AD model mice and to reveal its underlying molecular mechanisms. METHODS Cell viability assay and in vitro immunofluorescence imaging were used to evaluate the antioxidant effect of DMF on embryonic mouse hippocampal neurons. Behavioral test and brain magnetic resonance imaging were used to assess the therapeutic effects of DMF on spatial learning and memory as well as hippocampal volume in AD model mice with and without Nrf2 knockdown. Western blotting was used to analyze the expression of antioxidant enzymes and molecules associated with AD-related pathological pathways. RESULTS DMF inhibits reactive oxygen species overproduction and protects neurons without Nrf2 knockdown from death. DMF reduces amyloid-β induced memory impairment and hippocampal atrophy in AD model mice rather than in Nrf2 knockdown AD mice. DMF delays the progression of AD by activating the Nrf2 pathway to enhance the expression of downstream antioxidant enzymes and inhibits lipid peroxidation, apoptosis, inflammation, mitochondrial dysfunction and amyloid-β deposition. CONCLUSION These results indicate that DMF is a potential therapeutic option for AD through its antioxidant, anti-inflammatory, anti-apoptotic, and other anti-AD effects by activating the Nrf2 pathway.
Collapse
Affiliation(s)
- Xiaodi Sun
- Department of Radiology and Tianjin Key Laboratoryof Functional Imaging, Tianjin Medical University General Hospital, Tianjin, P.R. China
| | - Xinjun Suo
- Department of Radiology and Tianjin Key Laboratoryof Functional Imaging, Tianjin Medical University General Hospital, Tianjin, P.R. China
| | - Xianyou Xia
- Department of Cell Biology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Tianjin Medical University, Tianjin, P.R. China
| | - Chunshui Yu
- Department of Radiology and Tianjin Key Laboratoryof Functional Imaging, Tianjin Medical University General Hospital, Tianjin, P.R. China
| | - Yan Dou
- Department of Radiology and Tianjin Key Laboratoryof Functional Imaging, Tianjin Medical University General Hospital, Tianjin, P.R. China
| |
Collapse
|
8
|
Möhle L, Brackhan M, Bascuñana P, Pahnke J. Dimethyl fumarate does not mitigate cognitive decline and β-amyloidosis in female APPPS1 mice. Brain Res 2021; 1768:147579. [PMID: 34233173 DOI: 10.1016/j.brainres.2021.147579] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/14/2021] [Accepted: 06/30/2021] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Alzheimer's disease (AD) is the leading cause of dementia and a major global health issue. Currently, only limited treatment options are available to patients. One possibility to expand the treatment repertoire is repurposing of existing drugs such as dimethyl fumarate (DMF). DMF is approved for treatment of multiple sclerosis and previous animal studies have suggested that DMF may also have a beneficial effect for the treatment of AD. METHODS We used an APPPS1 transgenic model of senile β-amyloidosis and treated female mice orally with DMF in two treatment paradigms (pre and post onset). We quantified learning and memory parameters, β-amyloidosis, and neuroinflammation to determine the potential of DMF as AD therapeutics. RESULTS Treatment with DMF had no influence on water maze performance, β-amyloid accumulation, plaque formation, microglia activation, and recruitment of immune cells to the brain. Compared to vehicle-treated animals, oral DMF treatment could not halt or retard disease progression in the mice. DISCUSSION Our results do not favour the use of DMF as treatment for AD. While our results stand in contrast to previous findings in other models, they emphasize the importance of animal model selection and suggest further studies to elucidate the mechanisms leading to conflicting results.
Collapse
Affiliation(s)
- Luisa Möhle
- Department of Neuro-/Pathology, Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Oslo, Norway.
| | - Mirjam Brackhan
- Department of Neuro-/Pathology, Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Oslo, Norway; LIED, University of Lübeck, Lübeck, Germany
| | - Pablo Bascuñana
- Department of Neuro-/Pathology, Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Oslo, Norway
| | - Jens Pahnke
- Department of Neuro-/Pathology, Translational Neurodegeneration Research and Neuropathology Lab, University of Oslo (UiO) and Oslo University Hospital (OUS), Oslo, Norway; LIED, University of Lübeck, Lübeck, Germany; Department of Pharmacology, Faculty of Medicine, University of Latvia, Rīga, Latvia.
| |
Collapse
|
9
|
Habib AA, Hammad SF, Amer MM, Kamal AH. Stability indicating RP-HPLC method for determination of dimethyl fumarate in presence of its main degradation products: Application to degradation kinetics. J Sep Sci 2020; 44:726-734. [PMID: 33253476 DOI: 10.1002/jssc.202001007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 10/20/2020] [Accepted: 11/25/2020] [Indexed: 02/01/2023]
Abstract
A stability-indicating reverse phase-high-perfromance liquid chromatography method for the quantitative determination of dimethyl fumarate in presence of its main degradation products was developed. The chromatographic conditions were optimized using two-level full factorial design, chromatographic analysis was performed using Inertsil® column (250 × 4.6 mm, 5 μm) maintained at 25°C. Mobile phase was a mixture of water (pH 2.6 adjusted with phosphoric acid) and methanol (40:60, v/v) at a flow rate 1.0 mL/min, detection was performed at 210 nm using diode array detector. Stress degradation of dimethyl fumarate under acidic, alkaline, neutral, oxidative, photolytic, and thermal conditions was carried out, it was found to be very susceptible to hydrolysis under acidic and alkaline conditions; further investigation of degradation kinetics over pH range 1-9 was carried out. The degradation rate constant (K), t1/2 and t90 were calculated. Dimethyl fumarate show decreasing in stability in the following pH order: 7 < 5 < 3 < 1 < 9. The method was validated as per ICH guidelines, the method was found to be linear over concentration range 10-150 μg/mL with coefficient of determination (r2 ) 0.9997. The method was successfully applied for dimethyl fumarate determination in Marclerosis® dosage form within run time less than eight minutes without interference from excipients.
Collapse
Affiliation(s)
- Ahmed A Habib
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Sherin F Hammad
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Mona M Amer
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Amira H Kamal
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
10
|
Mullen L, Mengozzi M, Hanschmann EM, Alberts B, Ghezzi P. How the redox state regulates immunity. Free Radic Biol Med 2020; 157:3-14. [PMID: 31899344 DOI: 10.1016/j.freeradbiomed.2019.12.022] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/05/2019] [Accepted: 12/19/2019] [Indexed: 12/30/2022]
Abstract
Oxidative stress is defined as an imbalance between the levels of reactive oxygen species (ROS) and antioxidant defences. The view of oxidative stress as a cause of cell damage has evolved over the past few decades to a much more nuanced view of the role of oxidative changes in cell physiology. This is no more evident than in the field of immunity, where oxidative changes are now known to regulate many aspects of the immune response, and inflammatory pathways in particular. Our understanding of redox regulation of immunity now encompasses not only increases in reactive oxygen and nitrogen species, but also changes in the activities of oxidoreductase enzymes. These enzymes are important regulators of immune pathways both via changes in their redox activity, but also via other more recently identified cytokine-like functions. The emerging picture of redox regulation of immune pathways is one of increasing complexity and while therapeutic targeting of the redox environment to treat inflammatory disease is a possibility, any such strategy is likely to be more nuanced than simply inhibiting ROS production.
Collapse
Affiliation(s)
- Lisa Mullen
- Brighton and Sussex Medical School, Falmer, Brighton, UK
| | | | - Eva-Maria Hanschmann
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Germany
| | - Ben Alberts
- Brighton and Sussex Medical School, Falmer, Brighton, UK
| | - Pietro Ghezzi
- Brighton and Sussex Medical School, Falmer, Brighton, UK.
| |
Collapse
|
11
|
Takeda T, Tsubaki M, Asano R, Itoh T, Imano M, Satou T, Nishida S. Dimethyl fumarate suppresses metastasis and growth of melanoma cells by inhibiting the nuclear translocation of NF-κB. J Dermatol Sci 2020; 99:168-176. [PMID: 32693971 DOI: 10.1016/j.jdermsci.2020.07.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 07/11/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Malignant melanoma is among the deadliest forms of skin cancers, and its incidence has been increasing over the past decades. In malignant melanoma, activation of the nuclear factor kappa B (NF-κB) promotes survival, migration, and invasion of cancer cells. Anti-NF-κB agents for treating metastatic melanoma would be beneficial, but no such drug is approved as either monotherapy or adjuvant therapy. Dimethyl fumarate (DMF) is an approved anti-inflammatory drug already in clinical use for psoriasis and multiple sclerosis. OBJECTIVE We investigated the anti-tumour effect of DMF treatment in metastatic melanoma in vitro and in vivo. METHODS The cell viability was assessed via trypan blue exclusion assay. The migration and invasion was analyzed in a Boyden chamber assay. The anti-metastatic effects and anti-tumour activity of DMF was determined in an in-vivo model. The expressions of NF-κB pathway and NF-κB regulatory proteins were detected via western blotting. RESULTS DMF decreased the cell viability, migration and invasion in vitro. In addition, DMF inhibited spontaneous metastasis and tumour growth. Mechanistically, DMF prevented the nuclear translocation of NF-κB, whereas no changes were observed in the phosphorylation levels of inhibitor of kappa B (IκB). In addition, DMF inhibited the expression of matrix metalloproteinases (MMPs) and very late antigens (VLAs). Furthermore, DMF treatment decreased the expression of Survivin and Bcl-extra large (Bcl-XL) proteins. CONCLUSION Our results suggest that DMF as a novel inhibitor of NF-κB may be a potential therapeutic agent for metastatic melanoma.
Collapse
Affiliation(s)
- Tomoya Takeda
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Osaka, Japan
| | - Masanobu Tsubaki
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Osaka, Japan
| | - Ryota Asano
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Osaka, Japan
| | - Tatsuki Itoh
- Department of Food Science and Nutrition, Kindai University School of Agriculture, Nara, Japan
| | - Motohiro Imano
- Department of Surgery, Kindai University School of Medicine, Osaka, Japan
| | - Takao Satou
- Department of Pathology, Kindai University School of Medicine, Osaka, Japan
| | - Shozo Nishida
- Division of Pharmacotherapy, Kindai University School of Pharmacy, Osaka, Japan.
| |
Collapse
|
12
|
Serafini MM, Catanzaro M, Fagiani F, Simoni E, Caporaso R, Dacrema M, Romanoni I, Govoni S, Racchi M, Daglia M, Rosini M, Lanni C. Modulation of Keap1/Nrf2/ARE Signaling Pathway by Curcuma- and Garlic-Derived Hybrids. Front Pharmacol 2020; 10:1597. [PMID: 32047434 PMCID: PMC6997134 DOI: 10.3389/fphar.2019.01597] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 12/10/2019] [Indexed: 12/15/2022] Open
Abstract
Nrf2 is a basic leucine zipper transcription factor that binds to the promoter region of the antioxidant response element (ARE), inducing the coordinated up-regulation of antioxidant and detoxification genes. We recently synthesized a set of new molecules by combining the functional moieties of curcumin and diallyl sulfide, both known to induce the expression of antioxidant phase II enzymes by activating Nrf2 pathway. The aim of the study is to investigate the ability of such compounds to activate Keap1/Nrf2/ARE cytoprotective pathway, in comparison with two reference Nrf2-activators: curcumin and dimethyl fumarate, a drug approved for the treatment of relapsing-remitting multiple sclerosis. Furthermore, since Nrf2 pathway is known to be regulated also by epigenetic modifications, including key modifications in microRNA (miRNA) expression, the effects of the hybrids on the expression levels of selected miRNAs, associated with Nrf2 signaling pathway have also been investigated. The results show that compounds exert antioxidant effect by activating Nrf2 signaling pathway and inducing the ARE-regulated expression of its downstream target genes, such as HO-1 and NQO1, with two hybrids to a higher extent than curcumin. In addition, some molecules induce changes in the expression levels of miR-125b-5p, even if to a lesser extent than curcumin. However, no changes have been observed in the expression levels of mRNA coding for glutathione synthetase, suggesting that the modulation of this mRNA is not strictly under the control of miR-125b-5p, which could be influenced by other miRNAs.
Collapse
Affiliation(s)
- Melania Maria Serafini
- Department of Drug Sciences, University of Pavia, Pavia, Italy.,Scuola Universitaria Superiore IUSS, Pavia, Italy
| | | | - Francesca Fagiani
- Department of Drug Sciences, University of Pavia, Pavia, Italy.,Scuola Universitaria Superiore IUSS, Pavia, Italy
| | - Elena Simoni
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Roberta Caporaso
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Marco Dacrema
- Department of Pharmacy, University of Napoli Federico II, Naples, Italy
| | - Irene Romanoni
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Stefano Govoni
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Marco Racchi
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Maria Daglia
- Department of Pharmacy, University of Napoli Federico II, Naples, Italy
| | - Michela Rosini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Cristina Lanni
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
13
|
Nrf2 and oxidative stress. Pathology 2020. [DOI: 10.1016/b978-0-12-815972-9.00007-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
14
|
Sghaier R, Nury T, Leoni V, Caccia C, Pais De Barros JP, Cherif A, Vejux A, Moreau T, Limem K, Samadi M, Mackrill JJ, Masmoudi AS, Lizard G, Zarrouk A. Dimethyl fumarate and monomethyl fumarate attenuate oxidative stress and mitochondrial alterations leading to oxiapoptophagy in 158N murine oligodendrocytes treated with 7β-hydroxycholesterol. J Steroid Biochem Mol Biol 2019; 194:105432. [PMID: 31344443 DOI: 10.1016/j.jsbmb.2019.105432] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 07/01/2019] [Accepted: 07/19/2019] [Indexed: 01/08/2023]
Abstract
Oxidative stress and mitochondrial dysfunction contribute to the pathogenesis of neurodegenerative diseases and favor lipid peroxidation, leading to increased levels of 7β-hydroxycholesterol (7β-OHC) which induces oxiapoptophagy (OXIdative stress, APOPTOsis, autoPHAGY). The cytoprotective effects of dimethylfumarate (DMF), used in the treatment of relapsing remitting multiple sclerosis and of monomethylfumarate (MMF), its main metabolite, were evaluated on murine oligodendrocytes 158 N exposed to 7β-OHC (50 μM, 24 h) with or without DMF or MMF (25 μM). The activity of 7β-OHC in the presence or absence DMF or MMF was evaluated on several parameters: cell adhesion; plasma membrane integrity measured with propidium iodide (PI), trypan blue and fluoresceine diacetate (FDA) assays; LDH activity; antioxidant enzyme activities (superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GPx)); generation of lipid peroxidation products (malondialdehyde (MDA), conjugated dienes (CDs)) and protein oxidation products (carbonylated proteins (CPs)); reactive oxygen species (ROS) overproduction conducted with DHE and DHR123. The effect on mitochondria was determined with complementary criteria: measurement of succinate dehydrogenase activity, evaluation of mitochondrial potential (ΔΨm) and mitochondrial superoxide anions (O2●-) production using DiOC6(3) and MitoSOX, respectively; quantification of mitochondrial mass with Mitotracker Red, and of cardiolipins and organic acids. The effects on mitochondrial and peroxisomal ultrastructure were determined by transmission electron microscopy. Intracellular sterol and fatty acid profiles were determined. Apoptosis and autophagy were characterized by staining with Hoechst 33,342, Giemsa and acridine orange, and with antibodies raised against caspase-3 and LC3. DMF and MMF attenuate 7β-OHC-induced cytotoxicity: cell growth inhibition; decreased cell viability; mitochondrial dysfunction (decrease of succinate dehydrogenase activity, loss of ΔΨm, increase of mitochondrial O2●- production, alteration of the tricarboxilic acid (TCA) cycle, and cardiolipins content); oxidative stress induction (ROS overproduction, alteration of GPx, CAT, and SOD activities, increased levels of MDA, CDs, and CPs); changes in fatty acid and cholesterol metabolism; and cell death induction (caspase-3 cleavage, activation of LC3-I in LC3-II). Ultrastructural alterations of mitochondria and peroxisomes were prevented. These results demonstrate that DMF and MMF prevent major dysfunctions associated with neurodegenerative diseases: oxidative stress, mitochondrial dysfunction, apoptosis and autophagy.
Collapse
Affiliation(s)
- Randa Sghaier
- Univ. Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270 / Inserm, Dijon, France; Univ. Sousse, Laboratory of Biochemistry, Faculty of Medicine, Tunisia; Univ. Monastir, Faculty of Medicine, LR12ES05, Lab-NAFS 'Nutrition - Functional Food & Vascular Health', Monastir; Univ. Manouba, Laboratory of Biotechnology and Valorisation of Bio-Géo Ressources (LR11ES31), Higher Institute of Biotechnology, Sidi Thabet, Tunisia
| | - Thomas Nury
- Univ. Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270 / Inserm, Dijon, France
| | - Valerio Leoni
- Laboratory of Clinical Chemistry, Hospital of Varese, ASST-Settelaghi, Varese, Italy
| | - Claudio Caccia
- Unit of Medical Genetics and Neurogenetics, IRCCS Carlo Besta, Milano, Italy
| | | | - Ameur Cherif
- Univ. Manouba, Laboratory of Biotechnology and Valorisation of Bio-Géo Ressources (LR11ES31), Higher Institute of Biotechnology, Sidi Thabet, Tunisia
| | - Anne Vejux
- Univ. Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270 / Inserm, Dijon, France
| | - Thibault Moreau
- Univ. Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270 / Inserm, Dijon, France; Univ. Hospital, Department of Neurology, Dijon, France
| | - Khalifa Limem
- Univ. Sousse, Laboratory of Biochemistry, Faculty of Medicine, Tunisia
| | - Mohammad Samadi
- LCPMC-A2, ICPM, Dept of Chemistry, Univ. Lorraine, Metz Technopôle, Metz, France
| | - John J Mackrill
- Department of Physiology, Biosciences Institute, University College Cork, Cork, Ireland
| | - Ahmed Slaheddine Masmoudi
- Univ. Manouba, Laboratory of Biotechnology and Valorisation of Bio-Géo Ressources (LR11ES31), Higher Institute of Biotechnology, Sidi Thabet, Tunisia
| | - Gérard Lizard
- Univ. Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA 7270 / Inserm, Dijon, France.
| | - Amira Zarrouk
- Univ. Sousse, Laboratory of Biochemistry, Faculty of Medicine, Tunisia; Univ. Monastir, Faculty of Medicine, LR12ES05, Lab-NAFS 'Nutrition - Functional Food & Vascular Health', Monastir.
| |
Collapse
|
15
|
Tobore TO. On elucidation of the role of mitochondria dysfunction and oxidative stress in multiple sclerosis. ACTA ACUST UNITED AC 2019. [DOI: 10.1111/ncn3.12335] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
16
|
Hoang TT, Johnson DA, Raines RT, Johnson JA. Angiogenin activates the astrocytic Nrf2/antioxidant-response element pathway and thereby protects murine neurons from oxidative stress. J Biol Chem 2019; 294:15095-15103. [PMID: 31431502 DOI: 10.1074/jbc.ra119.008491] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 08/15/2019] [Indexed: 12/30/2022] Open
Abstract
The angiogenin (ANG) gene is mutated frequently in individuals with amyotrophic lateral sclerosis (ALS), a fatal neurodegenerative disease characterized by the progressive loss of motor neurons. Delivering human ANG to mice that display ALS-like symptoms extends their lifespan and improves motor function. ANG is a secretory vertebrate RNase that enters neuronal cells and cleaves a subset of tRNAs, leading to the inhibition of translation initiation and the assembly of stress granules. Here, using murine neuronal and astrocytic cell lines, we find that ANG triggers the activation of the Nrf2 (nuclear factor erythroid 2-related factor 2) pathway, which provides a critical cellular defense against oxidative stress. This activation, which occurred in astrocytes but not in neurons, promoted the survival of proximal neurons that had oxidative injury. These findings extend the role of ANG as a neuroprotective agent and underscore its potential utility in ALS management.
Collapse
Affiliation(s)
- Trish T Hoang
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | - Delinda A Johnson
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Ronald T Raines
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706 .,Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin 53706.,Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139
| | - Jeffrey A Johnson
- School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin 53705
| |
Collapse
|
17
|
Akino N, Wada-Hiraike O, Isono W, Terao H, Honjo H, Miyamoto Y, Tanikawa M, Sone K, Hirano M, Harada M, Hirata T, Hirota Y, Koga K, Oda K, Fujii T, Osuga Y. Activation of Nrf2/Keap1 pathway by oral Dimethylfumarate administration alleviates oxidative stress and age-associated infertility might be delayed in the mouse ovary. Reprod Biol Endocrinol 2019; 17:23. [PMID: 30760288 PMCID: PMC6375213 DOI: 10.1186/s12958-019-0466-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 02/08/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Age-associated infertility is a problem worldwide, and management of oxidative stress is known to be essential. Nuclear factor-E2-related factor 2 (Nrf2)/Kelch-like ECH-associated protein 1 (Keap1)-antioxidant response element (ARE) signaling pathway works as an essential defense mechanism against oxidative stress, and an oral drug Dimethylfumarate (DMF) is known to activate the pathway. METHODS We tested the hypothesis that oral DMF could alleviate oxidative stress in the ovary, resulting in salvation of age-associated infertility in a mouse model of reproductive age, and we examined the effects of DMF administration. 20 mg/kg DMF was administrated to female mice from 32 to 48 weeks, and Nrf2 levels, antioxidant levels, ovarian reserve, DNA damage, and oxidative stress were examined. RESULTS DMF administration resulted in elevated mRNA and protein levels of Nrf2, antioxidants, and telomere, and serum levels of Nrf2 and anti-mullerian hormone were also elevated. Results of TUNEL assay and Immunohistochemistry of mice ovarian tissues showed that DNA damage and oxidative stress were decreased by DMF administration, and significantly more oocytes were collected along with preservation of 60% more primordial follicles. CONCLUSIONS Our data suggest that DMF administration activates the Nrf2/Keap1 pathway, elevate levels of antioxidants, and decrease DNA damage and oxidative stress, resulting in improved ovarian reserve in the mouse ovary.
Collapse
Affiliation(s)
- Nana Akino
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Obstetrics and Gynecology, Graduate school of Medicine, The University of Tokyo, Tokyo, 1138655 Japan
| | - Osamu Wada-Hiraike
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Obstetrics and Gynecology, Graduate school of Medicine, The University of Tokyo, Tokyo, 1138655 Japan
| | - Wataru Isono
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Obstetrics and Gynecology, Graduate school of Medicine, The University of Tokyo, Tokyo, 1138655 Japan
- 0000 0000 9239 9995grid.264706.1Department of Obstetrics and Gynecology, Mizonokuchi Hospital, Teikyo University, Kawasaki, 2138507 Japan
| | - Hiromi Terao
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Obstetrics and Gynecology, Graduate school of Medicine, The University of Tokyo, Tokyo, 1138655 Japan
| | - Harunori Honjo
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Obstetrics and Gynecology, Graduate school of Medicine, The University of Tokyo, Tokyo, 1138655 Japan
| | - Yuichiro Miyamoto
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Obstetrics and Gynecology, Graduate school of Medicine, The University of Tokyo, Tokyo, 1138655 Japan
| | - Michihiro Tanikawa
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Obstetrics and Gynecology, Graduate school of Medicine, The University of Tokyo, Tokyo, 1138655 Japan
| | - Kenbun Sone
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Obstetrics and Gynecology, Graduate school of Medicine, The University of Tokyo, Tokyo, 1138655 Japan
| | - Mana Hirano
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Obstetrics and Gynecology, Graduate school of Medicine, The University of Tokyo, Tokyo, 1138655 Japan
| | - Miyuki Harada
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Obstetrics and Gynecology, Graduate school of Medicine, The University of Tokyo, Tokyo, 1138655 Japan
| | - Tetsuya Hirata
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Obstetrics and Gynecology, Graduate school of Medicine, The University of Tokyo, Tokyo, 1138655 Japan
| | - Yasushi Hirota
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Obstetrics and Gynecology, Graduate school of Medicine, The University of Tokyo, Tokyo, 1138655 Japan
| | - Kaori Koga
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Obstetrics and Gynecology, Graduate school of Medicine, The University of Tokyo, Tokyo, 1138655 Japan
| | - Katsutoshi Oda
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Obstetrics and Gynecology, Graduate school of Medicine, The University of Tokyo, Tokyo, 1138655 Japan
| | - Tomoyuki Fujii
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Obstetrics and Gynecology, Graduate school of Medicine, The University of Tokyo, Tokyo, 1138655 Japan
| | - Yutaka Osuga
- 0000 0001 2151 536Xgrid.26999.3dDepartment of Obstetrics and Gynecology, Graduate school of Medicine, The University of Tokyo, Tokyo, 1138655 Japan
| |
Collapse
|
18
|
|
19
|
Sigfridsson E, Marangoni M, Johnson JA, Hardingham GE, Fowler JH, Horsburgh K. Astrocyte-specific overexpression of Nrf2 protects against optic tract damage and behavioural alterations in a mouse model of cerebral hypoperfusion. Sci Rep 2018; 8:12552. [PMID: 30135571 PMCID: PMC6105641 DOI: 10.1038/s41598-018-30675-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 07/30/2018] [Indexed: 12/19/2022] Open
Abstract
Mouse models have shown that cerebral hypoperfusion causes white matter disruption and memory impairment relevant to the study of vascular cognitive impairment and dementia. The associated mechanisms include inflammation and oxidative stress are proposed to drive disruption of myelinated axons within hypoperfused white matter. The aim of this study was to determine if increased endogenous anti-oxidant and anti-inflammatory signalling in astrocytes was protective in a model of mild cerebral hypoperfusion. Transgenically altered mice overexpressing the transcription factor Nrf2 (GFAP-Nrf2) and wild type littermates were subjected to bilateral carotid artery stenosis or sham surgery. Behavioural alterations were assessed using the radial arm maze and tissue was collected for pathology and transcriptome analysis six weeks post-surgery. GFAP-Nrf2 mice showed less pronounced behavioural impairments compared to wild types following hypoperfusion, paralleled by reduced optic tract white matter disruption and astrogliosis. There was no effect of hypoperfusion on anti-oxidant gene alterations albeit the levels were increased in GFAP-Nrf2 mice. Instead, pro-inflammatory gene expression was determined to be significantly upregulated in the optic tract of hypoperfused wild type mice but differentially affected in GFAP-Nrf2 mice. In particular, complement components (C4 and C1q) were increased in wild type hypoperfused mice but expressed at levels similar to controls in hypoperfused GFAP-Nrf2 mice. This study provides evidence that overexpression of Nrf2 in astrocytes exerts beneficial effects through repression of inflammation and supports the potential use of Nrf2-activators in the amelioration of cerebrovascular-related inflammation and white matter degeneration.
Collapse
Affiliation(s)
- Emma Sigfridsson
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Martina Marangoni
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Jeffrey A Johnson
- Division of Pharmaceutical Sciences, University of Wisconsin, Madison, USA
- Molecular and Environmental Toxicology Center, University of Wisconsin, Madison, USA
- Center for Neuroscience, University of Wisconsin, Madison, USA
- Waisman Center, University of Wisconsin, Madison, USA
| | - Giles E Hardingham
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
- Edinburgh Medical School, University of Edinburgh, Edinburgh, UK
- The UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Jill H Fowler
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK.
| | - Karen Horsburgh
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
20
|
Akino N, Wada-Hiraike O, Terao H, Honjoh H, Isono W, Fu H, Hirano M, Miyamoto Y, Tanikawa M, Harada M, Hirata T, Hirota Y, Koga K, Oda K, Kawana K, Fujii T, Osuga Y. Activation of Nrf2 might reduce oxidative stress in human granulosa cells. Mol Cell Endocrinol 2018; 470:96-104. [PMID: 28986302 DOI: 10.1016/j.mce.2017.10.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 09/08/2017] [Accepted: 10/02/2017] [Indexed: 12/15/2022]
Abstract
Nuclear factor-E2-related factor 2 (Nrf2)/Kelch-like ECH-associated protein 1 (Keap1)-antioxidant response element (ARE) signaling pathway is one of the most important defense mechanisms against oxidative stress (OS). It is well documented that equilibration status of OS plays fundamental roles in human reproductive medicine, and the physiological role of Nrf2 in ovarian granulosa cells (GCs) has not been determined yet. Herein we aimed to study the function of Nrf2 in GCs. Human ovarian tissues were subjected to immunohistochemistry to localize Nrf2 and Keap1 and we detected the expression of Nrf2 and Keap1 in the human GCs. Human luteinized GCs were isolated and cultured, and hydrogen peroxide (H2O2) or Dimethylfumarates (DMF), an activator of Nrf2, were added to GCs to analyze the relationship between Nrf2 and antioxidants by quantitative RT-PCR. The mRNA levels of Nrf2, catalase, superoxide dismutase 1 (SOD1), and 8-Oxoguanine DNA glycosylase (OGG1) were elevated by H2O2, and DMF treatment showed similar but pronounced effects through activation of Nrf2. To determine the relationship of Nrf2 and the generation of antioxidants, siRNAs were used and quantitative RT-PCR were conducted. Decreased expression of Nrf2 resulted in a decreased level of these antioxidant mRNA. Intracellular levels of ROS were investigated by fluorescence of 8-hydroxy-2'-deoxyguanosine and fluorescent dye, 2',7'-dichlorodihydrofluorescein diacetate after H2O2 and/or DMF treatment, and DMF treatment quenched intracellular ROS generation by H2O2. These results show that activation of Nrf2 might lead to alleviate OS in human GCs, and this could provide novel insight to conquer the age-related fertility decline that is mainly attributed to the accumulation of aberrant OS.
Collapse
Affiliation(s)
- Nana Akino
- Department of Obstetrics and Gynecology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113 8655, Japan
| | - Osamu Wada-Hiraike
- Department of Obstetrics and Gynecology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113 8655, Japan.
| | - Hiromi Terao
- Department of Obstetrics and Gynecology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113 8655, Japan
| | - Harunori Honjoh
- Department of Obstetrics and Gynecology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113 8655, Japan
| | - Wataru Isono
- Department of Obstetrics and Gynecology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113 8655, Japan; Department of Physiological Chemistry and Metabolism, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113 8655, Japan
| | - Houju Fu
- Department of Obstetrics and Gynecology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113 8655, Japan
| | - Mana Hirano
- Department of Obstetrics and Gynecology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113 8655, Japan
| | - Yuichiro Miyamoto
- Department of Obstetrics and Gynecology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113 8655, Japan
| | - Michihiro Tanikawa
- Department of Obstetrics and Gynecology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113 8655, Japan
| | - Miyuki Harada
- Department of Obstetrics and Gynecology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113 8655, Japan
| | - Tetsuya Hirata
- Department of Obstetrics and Gynecology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113 8655, Japan
| | - Yasushi Hirota
- Department of Obstetrics and Gynecology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113 8655, Japan
| | - Kaori Koga
- Department of Obstetrics and Gynecology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113 8655, Japan
| | - Katsutoshi Oda
- Department of Obstetrics and Gynecology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113 8655, Japan
| | - Kei Kawana
- Department of Obstetrics and Gynecology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113 8655, Japan
| | - Tomoyuki Fujii
- Department of Obstetrics and Gynecology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113 8655, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113 8655, Japan
| |
Collapse
|
21
|
Omotoso GO, Gbadamosi IT, Afolabi TT, Abdulwahab AB, Akinlolu AA. Ameliorative effects of Moringa on cuprizone-induced memory decline in rat model of multiple sclerosis. Anat Cell Biol 2018; 51:119-127. [PMID: 29984057 PMCID: PMC6026826 DOI: 10.5115/acb.2018.51.2.119] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 01/14/2018] [Accepted: 03/05/2018] [Indexed: 01/03/2023] Open
Abstract
Cuprizone is a neurotoxin with copper-chelating ability used in animal model of multiple sclerosis in which oxidative stress has been documented as one of the cascade in the pathogenesis. Moringa oleifera is a phytomedicinal plant with antioxidant and neuroprotective properties. This study aimed at evaluating the ameliorative capability of M. oleifera in cuprizone-induced behavioral and histopathological alterations in the prefrontal cortex and hippocampus of Wistar rats. Four groups of rats were treated with normal saline, cuprizone, M. oleifera and a combination of M. oleifera and cuprizone, for five weeks. The rats were subjected to Morris water maze and Y-maze to assess long and short-term memory respectively. The animals were sacrificed, and brain tissues were removed for histochemical and enzyme lysate immunosorbent assay for catalase, superoxide dismutase, and nitric oxide. Cuprizone significantly induced oxidative and nitrosative stress coupled with memory decline and cortico-hippocampal neuronal deficits; however, administration of M. oleifera significantly reversed the neuropathological deficits induced by cuprizone.
Collapse
Affiliation(s)
- Gabriel Olaiya Omotoso
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Ismail Temitayo Gbadamosi
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Theresa Titilayo Afolabi
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Ahmad Bolakale Abdulwahab
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Adelaja Abdulazeez Akinlolu
- Department of Anatomy, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| |
Collapse
|
22
|
Zhang Q, Li Z, Wu S, Li X, Sang Y, Li J, Niu Y, Ding H. Myricetin alleviates cuprizone-induced behavioral dysfunction and demyelination in mice by Nrf2 pathway. Food Funct 2018; 7:4332-4342. [PMID: 27713953 DOI: 10.1039/c6fo00825a] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Multiple sclerosis (MS) is a demyelinating disease occurring in the central nervous system. In the present study, we evaluated the function of myricetin on the alleviation of behavioral dysfunction and myelin protection in the cuprizone-induced demyelination model. Mice were daily fed with fodder including 0.2% cuprizone and were administrated myricetin (100 mg kg-1) by gavage administration for 5 weeks. The treatment of myricetin ameliorated hyper-locomotion and behavior impairment induced by cuprizone toxicity. With the administration of myricetin, the demyelinating lesion was lessened via increasing the LFB staining area and myelin phosphatide protein (MBP) expression. In addition, myricetin evidently promoted Nrf2 translocation in the nuclear fraction and enhanced the HO-1 and NQO1 expression levels. Our data revealed that myricetin may be a potential candidate for mitigating motor defects and demyelination in a cuprizone-induced mouse model via activating the Nrf2 pathway.
Collapse
Affiliation(s)
- Qianying Zhang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, P. R. China.
| | - Zhike Li
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, P. R. China.
| | - Shuangchan Wu
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, P. R. China.
| | - Xiaofei Li
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, P. R. China.
| | - Ying Sang
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, P. R. China.
| | - Jian Li
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, P. R. China.
| | - Yunhui Niu
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, P. R. China.
| | - Hong Ding
- Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, Wuhan University School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, P. R. China.
| |
Collapse
|
23
|
Mills EA, Ogrodnik MA, Plave A, Mao-Draayer Y. Emerging Understanding of the Mechanism of Action for Dimethyl Fumarate in the Treatment of Multiple Sclerosis. Front Neurol 2018; 9:5. [PMID: 29410647 PMCID: PMC5787128 DOI: 10.3389/fneur.2018.00005] [Citation(s) in RCA: 157] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 01/03/2018] [Indexed: 01/22/2023] Open
Abstract
Dimethyl fumarate (DMF) is an effective treatment option for relapsing-remitting multiple sclerosis (MS), but its therapeutic mechanism of action has not been fully elucidated. A better understanding of its mechanism will allow for the development of assays to monitor its clinical efficacy and safety in patients, as well as guide the development of the next generation of therapies for MS. In order to build the foundation for determining its mechanism, we reviewed the manner in which DMF alters lymphocyte subsets in MS patients, its impact on clinical efficacy and safety, as well as its molecular effects in cellular and animal models. DMF decreases absolute lymphocyte counts, but does not affect all subsets uniformly. CD8+ T-cells are the most profoundly affected, but reduction also occurs in the CD4+ population, particularly within the pro-inflammatory T-helper Th1 and Th17 subsets, creating a bias toward more anti-inflammatory Th2 and regulatory subsets. Similarly, B-lymphocyte, myeloid, and natural killer populations are also shifted toward a more anti-inflammatory state. In vitro and animal models demonstrate a role for DMF within the central nervous system (CNS) in promoting neuronal survival in an Nrf2 pathway-dependent manner. However, the impact of DMF directly within the CNS of MS patients remains largely unknown.
Collapse
Affiliation(s)
- Elizabeth A Mills
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Magdalena A Ogrodnik
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Andrew Plave
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Yang Mao-Draayer
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, United States.,Graduate Program in Immunology, Program in Biomedical Sciences, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
24
|
Hamada S, Shimosegawa T, Taguchi K, Nabeshima T, Yamamoto M, Masamune A. Simultaneous K-ras activation and Keap1 deletion cause atrophy of pancreatic parenchyma. Am J Physiol Gastrointest Liver Physiol 2018; 314:G65-G74. [PMID: 28971839 DOI: 10.1152/ajpgi.00228.2017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The Kelch-like ECH-associated protein 1 (Keap1)-NF-E2-related factor 2 (Nrf2) system has a wide variety of effects in addition to the oxidative stress response, such as growth promotion and chemoresistance of cancer cells. Nrf2 is constitutively activated in most cancer cells. However, the activation of Nrf2 together with oncogenic mutations does not always result in cancer promotion. K-rasLSL-G12D/+:: p53LSL-R172H/+:: Pdx-1-Cre (KPC) mice are an established model of pancreatic cancer that specifically express mutants of both K-ras and p53 in the pancreas by using Pdx-1-Cre. We here generated Pdx-1-Cre::K-rasLSL-G12D/+:: Keap1fl/fl (KC::Keap1) and KPC:: Keap1fl/fl (KPC::Keap1) mice in which Nrf2 is constitutively activated by Keap1 deletion. KC::Keap1 and KPC::Keap1 mice started to die or showed obvious weakness at approximately around 40 days after birth. Histological examination revealed that KC::Keap1 and KPC::Keap1 mice did not develop pancreatic cancer but, instead, progressive atrophy of the pancreatic parenchyma. In these mice, amylase-positive acinar cells as well as insulin- and glucagon-positive islet cells were decreased and surrounded by fibrotic tissues. KC::Keap1 and KPC::Keap1 mice presented lower body weight and glucose levels than C::Keap1 mice, presumably resulting from pancreatic exocrine insufficiency. Histological changes were not obvious in C::Keap1 and PC::Keap1 mice. The presence of the p53 mutation did not affect the phenotypes in KC::Keap1 mice. Heterologous or homologous Nrf2 deletion ( Nrf2+/- or Nrf2-/-) rescued the pancreatic phenotypes, weight loss, and hypoglycemia in KC::Keap1 mice, suggesting that Nrf2 is a major downstream target of Keap1. In conclusion, simultaneous K-ras activation and Keap1 deletion caused progressive atrophy of the pancreatic parenchyma in mice. NEW & NOTEWORTHY Aberrant activation of the Kelch-like ECH-associated protein 1 (Keap1)-NF-E2-related factor 2 (Nrf2) system usually promotes carcinogenesis, and we assumed that simultaneous activation of K-ras and Nrf2 might promote pancreatic carcinogenesis. Conditional expression of mutant K-ras and Keap1 deletion did not result in pancreatic cancer development. Instead, these mice developed progressive loss of pancreatic parenchyma, accompanied by body weight loss and hypoglycemia, presumably because of pancreatic exocrine insufficiency. Nrf2 activation by Keap1 deletion concomitant with K-ras activation cause pancreatic atrophy.
Collapse
Affiliation(s)
- Shin Hamada
- Division of Gastroenterology, Tohoku University Graduate School of Medicine , Sendai , Japan
| | - Tooru Shimosegawa
- Division of Gastroenterology, Tohoku University Graduate School of Medicine , Sendai , Japan
| | - Keiko Taguchi
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine , Sendai , Japan
| | - Tatsuhide Nabeshima
- Division of Gastroenterology, Tohoku University Graduate School of Medicine , Sendai , Japan
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine , Sendai , Japan
| | - Atsushi Masamune
- Division of Gastroenterology, Tohoku University Graduate School of Medicine , Sendai , Japan
| |
Collapse
|
25
|
Wu Q, Wang Q, Mao G, Dowling CA, Lundy SK, Mao-Draayer Y. Dimethyl Fumarate Selectively Reduces Memory T Cells and Shifts the Balance between Th1/Th17 and Th2 in Multiple Sclerosis Patients. THE JOURNAL OF IMMUNOLOGY 2017; 198:3069-3080. [PMID: 28258191 DOI: 10.4049/jimmunol.1601532] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 02/08/2017] [Indexed: 01/20/2023]
Abstract
Dimethyl fumarate (DMF; trade name Tecfidera) is an oral formulation of the fumaric acid ester that is Food and Drug Administration approved for treatment of relapsing-remitting multiple sclerosis. To better understand the therapeutic effects of Tecfidera and its rare side effect of progressive multifocal leukoencephalopathy, we conducted cross-sectional and longitudinal studies by immunophenotyping cells from peripheral blood (particularly T lymphocytes) derived from untreated and 4-6 and 18-26 mo Tecfidera-treated stable relapsing-remitting multiple sclerosis patients using multiparametric flow cytometry. The absolute numbers of CD4 and CD8 T cells were significantly decreased and the CD4/CD8 ratio was increased with DMF treatment. The proportions of both effector memory T cells and central memory T cells were reduced, whereas naive T cells increased in treated patients. T cell activation was reduced with DMF treatment, especially among effector memory T cells and effector memory RA T cells. Th subsets Th1 (CXCR3+), Th17 (CCR6+), and particularly those expressing both CXCR3 and CD161 were reduced most significantly, whereas the anti-inflammatory Th2 subset (CCR3+) was increased after DMF treatment. A corresponding increase in IL-4 and decrease in IFN-γ and IL-17-expressing CD4+ T cells were observed in DMF-treated patients. DMF in vitro treatment also led to increased T cell apoptosis and decreased activation, proliferation, reactive oxygen species, and CCR7 expression. Our results suggest that DMF acts on specific memory and effector T cell subsets by limiting their survival, proliferation, activation, and cytokine production. Monitoring these subsets could help to evaluate the efficacy and safety of DMF treatment.
Collapse
Affiliation(s)
- Qi Wu
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Qin Wang
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Guangmei Mao
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Catherine A Dowling
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Steven K Lundy
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109; and.,Graduate Program in Immunology, Program in Biomedical Sciences, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Yang Mao-Draayer
- Department of Neurology, University of Michigan Medical School, Ann Arbor, MI 48109; .,Graduate Program in Immunology, Program in Biomedical Sciences, University of Michigan Medical School, Ann Arbor, MI 48109
| |
Collapse
|
26
|
Assessment of Serum Nitrogen Species and Inflammatory Parameters in Relapsing-Remitting Multiple Sclerosis Patients Treated with Different Therapeutic Approaches. BIOMED RESEARCH INTERNATIONAL 2016; 2016:4570351. [PMID: 28078290 PMCID: PMC5204117 DOI: 10.1155/2016/4570351] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Revised: 10/20/2016] [Accepted: 11/08/2016] [Indexed: 11/17/2022]
Abstract
The role of nitric oxide and its reactive derivatives (NO x ) is well known in the pathogenesis of multiple sclerosis, which is an inflammatory disease while NO x seems to be important in coordinating inflammatory response. The purpose of the present study was to assess serum NO x as one of the nitrogen species and inflammatory parameters in relapsing-remitting multiple sclerosis patients and to compare the effectiveness of various types of disease-modifying therapies that reduce nitric oxide and inflammatory biomarkers. Elevated NO x level was observed in patients who received the first-line disease-modifying therapy (interferons beta-1a and beta-1b) in comparison with the subjects treated with the second-line disease-modifying therapy (natalizumab; fingolimod) and healthy controls without significant differences in C-reactive protein and interleukin-1 beta. A negative correlation was observed between serum NO x level and the duration of multiple sclerosis confirmed in the whole study population and in subjects treated with the first-line agents. Only serum NO x , concentration could reveal a potential efficacy of disease-modifying therapy with a better reduction in NO x level due to the second-line agents of disease-modifying therapy.
Collapse
|
27
|
Bhise V, Dhib-Jalbut S. Further understanding of the immunopathology of multiple sclerosis: impact on future treatments. Expert Rev Clin Immunol 2016; 12:1069-89. [PMID: 27191526 DOI: 10.1080/1744666x.2016.1191351] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The understanding of the immunopathogenesis of multiple sclerosis (MS) has expanded with more research into T-cell subtypes, cytokine contributors, B-cell participation, mitochondrial dysfunction, and more. Treatment options have rapidly expanded with three relatively recent oral therapy alternatives entering the arena. AREAS COVERED In the following review, we discuss current mechanisms of immune dysregulation in MS, how they relate to current treatments, and the impact these findings will have on the future of therapy. Expert commentary: The efficacy of these medications and understanding their mechanisms of actions validates the immunopathogenic mechanisms thought to underlie MS. Further research has exposed new targets, while new promising therapies have shed light on new aspects into the pathophysiology of MS.
Collapse
Affiliation(s)
- Vikram Bhise
- a Rutgers Biomedical and Health Sciences - Departments of Pediatrics , Robert Wood Johnson Medical School , New Brunswick , NJ , USA
| | - Suhayl Dhib-Jalbut
- b Rutgers Biomedical and Health Sciences - Departments of Neurology , Robert Wood Johnson Medical School , New Brunswick , NJ , USA
| |
Collapse
|