1
|
Staun-Ram E, Volkowich A, Miller A. Immunotherapy-mediated modulation of the gut microbiota in multiple sclerosis and associations with diet and clinical response-the effect of dimethyl fumarate therapy. Ther Adv Neurol Disord 2025; 18:17562864241306565. [PMID: 40092554 PMCID: PMC11907610 DOI: 10.1177/17562864241306565] [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: 06/17/2024] [Accepted: 11/18/2024] [Indexed: 03/19/2025] Open
Abstract
Background Accumulating evidence supports a role of the microbiota in health and disease, including in multiple sclerosis (MS). How MS drugs affect the microbiota and whether this is part of their mode of action is yet unknown. Objectives To assess how dimethyl fumarate (DMF) affects the gut microbiota and whether the microbiota is associated with clinical response or adverse events (AEs) to DMF or diet. Design An observational cohort study, in which the microbiota from 45 patients with relapsing-remitting MS pre-DMF initiation and following 6 months of DMF therapy, and from 47 matched healthy controls, were compared, and associations with clinical and dietary data assessed. Data sources and methods Microbial DNA was sequenced and analyzed using MicrobiomeAnalyst. The clinical response was assessed after 1-year DMF therapy based upon evidence of disease activity (relapse, ΔEDSS increase >1, or MRI activity compared to pre-treatment). Dietary data were obtained by food questionnaires. Results Alterations in relative abundance of several microbes were identified post 6-month DMF therapy compared to pre-treatment, including an increase in Firmicutes, Lachnospiraceae, and Ruminococcaceae, while reduction in Bacteroidetes and Proteobacteria. Patients who showed disease activity within 1 year from DMF initiation had pre-treatment higher abundance of Proteobacteria, Flavonifractor, and Acidaminococcaceae, while lower abundance of Firmicutes, Ruminococcaceae, Butyricicoccus, and Massiliprevotella massiliensis, compared to patients without disease activity. Patients who discontinued DMF therapy due to AEs had pre-treatment higher abundance of Proteobacteria, Bacteroidetes, Eggerthella, and Lachnoclostridium and lower abundance of Ruminococcaceae, Megamonas, and Holdemanella, among others. Differentially abundant microbes correlated with intake of several nutrients. Conclusion DMF immunotherapy is associated with modifications of the microbiota. The microbiota may affect the severity of AEs and the clinical response to DMF, and is potentially modulated by diet. Microbiota-based, personalized treatment approach, integrating pharmacotherapy with dietary components, carries potential to improved clinical outcome.
Collapse
Affiliation(s)
- Elsebeth Staun-Ram
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Neuroimmunology Unit and Multiple Sclerosis Center, Lady Davis Carmel Medical Center, Haifa, Israel
| | - Anat Volkowich
- Neuroimmunology Unit and Multiple Sclerosis Center, Lady Davis Carmel Medical Center, Haifa, Israel
| | - Ariel Miller
- Neuroimmunology Unit and Multiple Sclerosis Center, Lady Davis Carmel Medical Center, Michal St. 7, Haifa 3436212, Israel
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
- Department of Neurology, Lady Davis Carmel Medical Center, Haifa 3436212, Israel
| |
Collapse
|
2
|
Candan B, Gungor S. Current and Evolving Concepts in the Management of Complex Regional Pain Syndrome: A Narrative Review. Diagnostics (Basel) 2025; 15:353. [PMID: 39941283 PMCID: PMC11817358 DOI: 10.3390/diagnostics15030353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
Background/Objectives: Complex regional pain syndrome (CRPS) is characterized by severe pain and reduced functionality, which can significantly affect an individual's quality of life. The current treatment of CRPS is challenging. However, recent advances in diagnostic and treatment methods show promise for improving patient outcomes. This review aims to place the question of CRPS in a broader context and highlight the objectives of the research for future directions in the management of CRPS. Methods: This study involved a comprehensive literature review. Results: Research has identified three primary pathophysiological pathways that may explain the clinical variability observed in CRPS: inflammatory mechanisms, vasomotor dysfunction, and maladaptive neuroplasticity. Investigations into these pathways have spurred the development of novel diagnostic and treatment strategies focused on N-Methyl-D-aspartate Receptor Antagonists (NMDA), Toll-like receptor 4 (TLR-4), α1 and α2 adrenoreceptors, as well as the identification of microRNA (miRNA) biomarkers. Treatment methods being explored include immune and glial-modulating agents, intravenous immunoglobulin (IVIG) therapy, plasma exchange therapy, and neuromodulation techniques. Additionally, there is ongoing debate regarding the efficacy of other treatments, such as free radical scavengers, alpha-lipoic acid (ALA), dimethyl fumarate (DMF), adenosine monophosphate-activated protein kinase (AMPK) activators such as metformin, and phosphodiesterase-5 inhibitors such as tadalafil. Conclusions: The controversies surrounding the mechanisms, diagnosis, and treatment of CRPS have prompted researchers to investigate new approaches aimed at enhancing understanding and management of the condition, with the goal of alleviating symptoms and reducing associated disabilities.
Collapse
Affiliation(s)
- Burcu Candan
- Department of Anesthesiology and Reanimation, Bahçeşehir University Göztepe Medical Park Hospital, 34732 Istanbul, Türkiye
| | - Semih Gungor
- Division of Musculoskeletal and Interventional Pain Management, Department of Anesthesiology, Critical Care and Pain Management, Hospital for Special Surgery, New York, NY 10021, USA;
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
3
|
Vida H, Sahar M, Nikdouz A, Arezoo H. Chemokines in neurodegenerative diseases. Immunol Cell Biol 2024. [PMID: 39723647 DOI: 10.1111/imcb.12843] [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: 12/04/2023] [Revised: 06/09/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024]
Abstract
Neurodegeneration and neuroinflammation disorders are mainly the result of the deposition of various proteins, such as α-synuclein, amyloid-β and prions, which lead to the initiation and activation of inflammatory responses. Different chemokines are involved in the infiltration and movement of inflammatory leukocytes into the central nervous system (CNS) that express chemokine receptors. Dysregulation of several members of chemokines has been shown in the CNS, cerebrospinal fluid and peripheral blood of patients who have neurodegenerative disorders. Upon infiltration of various cells, they produce many inflammatory mediators such as cytokines. Besides them, some CNS-resident cells, such as neurons and astrocytes, are also involved in the pathogenesis of neurodegeneration by producing chemokines. In this review, we summarize the role of chemokines and their related receptors in the pathogenesis of neurodegeneration and neuroinflammation disorders, including multiple sclerosis, Parkinson's disease and Alzheimer's disease. Therapeutic strategies targeting chemokines or their related receptors are also discussed in this article.
Collapse
Affiliation(s)
- Hashemi Vida
- Medicinal Plants Research Center, Maragheh University of Medical Sciences, Maragheh, Iran
| | - Mehranfar Sahar
- Cellular and Molecular Medicine Research Institute, Cellular and Molecular Research Center, Urmia University of Medical Sciences, Urmia, Iran
- Department of Immunology and Genetics, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
- Urmia University of Medical Sciences, Urmia, Iran
| | - Amin Nikdouz
- Department of Translational Medicine, Universita degli Studi del Piemonte Orientale Amedeo Avogadro, Vercelli, Italy
| | - Hosseini Arezoo
- Cellular and Molecular Medicine Research Institute, Cellular and Molecular Research Center, Urmia University of Medical Sciences, Urmia, Iran
- Department of Immunology and Genetics, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
4
|
Mohammed EMA. Understanding Multiple Sclerosis Pathophysiology and Current Disease-Modifying Therapies: A Review of Unaddressed Aspects. FRONT BIOSCI-LANDMRK 2024; 29:386. [PMID: 39614433 DOI: 10.31083/j.fbl2911386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/31/2024] [Accepted: 08/13/2024] [Indexed: 12/01/2024]
Abstract
Multiple sclerosis (MS) is a complex autoimmune disorder of the central nervous system (CNS) with an unknown etiology and pathophysiology that is not completely understood. Although great strides have been made in developing disease-modifying therapies (DMTs) that have significantly improved the quality of life for MS patients, these treatments do not entirely prevent disease progression or relapse. Identifying the unaddressed pathophysiological aspects of MS and developing targeted therapies to fill in these gaps are essential in providing long-term relief for patients. Recent research has uncovered some aspects of MS that remain outside the scope of available DMTs, and as such, yield only limited benefits. Despite most MS pathophysiology being targeted by DMTs, many patients still experience disease progression or relapse, indicating that a more detailed understanding is necessary. Thus, this literature review seeks to explore the known aspects of MS pathophysiology, identify the gaps in present DMTs, and explain why current treatments cannot entirely arrest MS progression.
Collapse
Affiliation(s)
- Eiman M A Mohammed
- Kuwait Cancer Control Centre, Department of Medical Laboratory, Molecular Genetics Laboratory, Ministry of Health, 13001 Shuwaikh, Kuwait
| |
Collapse
|
5
|
Xu D, Wang M, Wang L. Simvastatin alleviates experimental autoimmune encephalomyelitis through regulating the balance of Th17 and Treg in mice. Allergol Immunopathol (Madr) 2024; 52:36-43. [PMID: 39278849 DOI: 10.15586/aei.v52i5.1100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/22/2024] [Indexed: 09/18/2024]
Abstract
The aim of this study was to elucidate the therapeutic effect of simvastatin on experimental autoimmune encephalomyelitis (EAE) by regulating the balance between Th17 and Treg cells in mice. C57BL/6 mice were randomly divided into four groups: normal group, EAE group, simvastatin (2 and 10 mg/kg) group, and AG490 group (with AG490 serving as the positive control). Neurological function scores of mice were assessed daily. The four groups received treatments of normal saline, normal saline, and simvastatin (2 and 10 mg/kg), respectively. In the AG490 group, mice were injected intraperitoneally with AG490 (1 mg) every other day, and treatment was halted after 3 weeks. The spinal cord was stained with hematoxylin and eosin (H&E), and immunohistochemical staining for retinoic acid receptor-related orphan receptor γ(RORγ) and Foxp3 (Foxp3) was performed. Spleen samples were taken for Th17 and Treg analysis using flow cytometry. The levels of interleukin-17 and transforming growth factor-β (TGF-β) were detected using enzyme-linked immunosorbent assay (ELISA). In the simvastatin and AG490 groups, recovery from neurological impairment was earlier compared to the EAE group, and the symptoms were notably improved. Both simvastatin and AG490 reduced focal inflammation, decreased RORγ-positive cell infiltration, and significantly increased the number of FOXP3-positive cells. The number of Th17 cells and the level of IL-17 in the spleen were decreased in the simvastatin and AG490 treatment groups, while the number of Treg cells and TGF-β levels were significantly increased across all treatment groups. Simvastatin exhibits anti-inflammatory and immunomodulatory effects, potentially alleviating symptoms of neurological dysfunction of EAE. Regulating the balance between Th17 and Treg may represent a therapeutic mechanism for simvastatin in treating EAE.
Collapse
MESH Headings
- Animals
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Th17 Cells/immunology
- Th17 Cells/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/drug effects
- Simvastatin/pharmacology
- Simvastatin/administration & dosage
- Mice
- Mice, Inbred C57BL
- Female
- Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism
- Interleukin-17/metabolism
- Forkhead Transcription Factors/metabolism
- Spinal Cord/immunology
- Spinal Cord/drug effects
- Spinal Cord/pathology
- Humans
- Transforming Growth Factor beta/metabolism
- Disease Models, Animal
Collapse
Affiliation(s)
- Dongsheng Xu
- Department of Neurology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Manxia Wang
- Department of Neurology, The Second Hospital of Lanzhou University, Lanzhou, China;
| | - Lijuan Wang
- Department of Neurology, The Second Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
6
|
Wang J, Yuan Y, Zhang S, Lu S, Han G, Bian M, huang L, Meng D, Su D, Xiao L, Xiao Y, Zhang J, Gong N, Jiang L. Remodeling of the Intra-Conduit Inflammatory Microenvironment to Improve Peripheral Nerve Regeneration with a Neuromechanical Matching Protein-Based Conduit. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2302988. [PMID: 38430538 PMCID: PMC11077661 DOI: 10.1002/advs.202302988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 12/22/2023] [Indexed: 03/04/2024]
Abstract
Peripheral nerve injury (PNI) remains a challenging area in regenerative medicine. Nerve guide conduit (NGC) transplantation is a common treatment for PNI, but the prognosis of NGC treatment is unsatisfactory due to 1) neuromechanical unmatching and 2) the intra-conduit inflammatory microenvironment (IME) resulting from Schwann cell pyroptosis and inflammatory-polarized macrophages. A neuromechanically matched NGC composed of regenerated silk fibroin (RSF) loaded with poly(3,4-ethylenedioxythiophene): poly(styrene sulfonate) (P:P) and dimethyl fumarate (DMF) are designed, which exhibits a matched elastic modulus (25.1 ± 3.5 MPa) for the peripheral nerve and the highest 80% elongation at break, better than most protein-based conduits. Moreover, the NGC can gradually regulate the intra-conduit IME by releasing DMF and monitoring sciatic nerve movements via piezoresistive sensing. The combination of NGC and electrical stimulation modulates the IME to support PNI regeneration by synergistically inhibiting Schwann cell pyroptosis and reducing inflammatory factor release, shifting macrophage polarization from the inflammatory M1 phenotype to the tissue regenerative M2 phenotype and resulting in functional recovery of neurons. In a rat sciatic nerve crush model, NGC promoted remyelination and functional and structural regeneration. Generally, the DMF/RSF/P:P conduit provides a new potential therapeutic approach to promote nerve repair in future clinical treatments.
Collapse
Affiliation(s)
- Jia‐Yi Wang
- Department of Orthopaedic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Ya Yuan
- Department of Orthopaedic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
- Department of RehabilitationZhongshan HospitalFudan UniversityShanghai200032China
| | - Shu‐Yan Zhang
- The Key Laboratory for Ultrafine Materials of Ministry of Education, Engineering Research Centre for Biomedical Materials of Ministry of EducationFrontiers Science Center for Materiobiology and Dynamic ChemistrySchool of Materials Science and EngineeringEast China University of Science and TechnologyShanghai200237China
| | - Shun‐Yi Lu
- Department of Orthopaedic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Guan‐Jie Han
- Department of Orthopaedic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Meng‐Xuan Bian
- Department of Orthopaedic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Lei huang
- Department of Orthopaedic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - De‐Hua Meng
- Department of Orthopaedic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Di‐Han Su
- Department of Orthopaedic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Lan Xiao
- School of MechanicalMedical and Process EngineeringCentre for Biomedical TechnologiesQueensland University of TechnologyBrisbane4059Australia
- Australia‐China Centre for Tissue Engineering and Regenerative MedicineQueensland University of TechnologyBrisbane4059Australia
| | - Yin Xiao
- School of MechanicalMedical and Process EngineeringCentre for Biomedical TechnologiesQueensland University of TechnologyBrisbane4059Australia
- Australia‐China Centre for Tissue Engineering and Regenerative MedicineQueensland University of TechnologyBrisbane4059Australia
- School of Medicine and Dentistry & Menzies Health Institute QueenslandGriffith UniversityGold Coast4222Australia
| | - Jian Zhang
- Department of Orthopaedic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| | - Ning‐Ji Gong
- Department of EmergencyDepartment of OrthopedicsThe Second HospitalCheeloo College of MedicineShandong UniversityJinanShandong250033China
| | - Li‐Bo Jiang
- Department of Orthopaedic SurgeryZhongshan HospitalFudan UniversityShanghai200032China
| |
Collapse
|
7
|
Zhang R, Zhang Q, Cui Z, Huang B, Ma H. Dimethyl fumarate restores Ca 2+ dyshomeostasis through activation of the SIRT1 signal to treat nonalcoholic fatty liver disease. Life Sci 2024; 341:122505. [PMID: 38364937 DOI: 10.1016/j.lfs.2024.122505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 02/09/2024] [Accepted: 02/12/2024] [Indexed: 02/18/2024]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is characterized by an excessive lipid accumulation in the liver, with a global prevalence of approximately 25 %. While early-stage steatosis is reversible and can be intervened upon, it has the potential to progress to some serious complications, including cirrhosis and even liver cancer. Dimethyl fumarate (DMF), a derivative of fumaric acid shows promise in intervening in certain diseases. However, the precise effect and underlying mechanism of DMF on hepatic steatosis remain unclear. In this study, we demonstrated that DMF mitigates hepatic steatosis in mice subjected to high-fat/high-cholesterol (HFHC) diets. Meanwhile, our in vivo and in vitro results showed that DMF relieves lipid accumulation, oxidative stress, and endoplasmic reticulum (ER) stress. Mechanically, our findings revealed that the effect of DMF on reducing lipid accumulation is linked to the restoration of Ca2+ homeostasis. Furthermore, we found that activation of the SIRT1 signal by DMF plays an important role in correcting the mishandling of the Ca2+ signal, and knockdown of SIRT1 expression reverses the beneficial role of DMF PA-incubated AML12 cells. In conclusion, our results suggested DMF's amelioration of hepatic steatosis is related to the activation of SIRT1-mediated Ca2+ signaling.
Collapse
Affiliation(s)
- Rui Zhang
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Quanwei Zhang
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - ZiYi Cui
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - BenZeng Huang
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Haitian Ma
- Key Laboratory of Animal Physiology and Biochemistry, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China; MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China.
| |
Collapse
|
8
|
Manai F, Zanoletti L, Arfini D, Micco SGD, Gjyzeli A, Comincini S, Amadio M. Dimethyl Fumarate and Intestine: From Main Suspect to Potential Ally against Gut Disorders. Int J Mol Sci 2023; 24:9912. [PMID: 37373057 DOI: 10.3390/ijms24129912] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/04/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Dimethyl fumarate (DMF) is a well-characterized molecule that exhibits immuno-modulatory, anti-inflammatory, and antioxidant properties and that is currently approved for the treatment of psoriasis and multiple sclerosis. Due to its Nrf2-dependent and independent mechanisms of action, DMF has a therapeutic potential much broader than expected. In this comprehensive review, we discuss the state-of-the-art and future perspectives regarding the potential repurposing of DMF in the context of chronic inflammatory diseases of the intestine, such as inflammatory bowel disorders (i.e., Crohn's disease and ulcerative colitis) and celiac disease. DMF's mechanisms of action, as well as an exhaustive analysis of the in vitro/in vivo evidence of its beneficial effects on the intestine and the gut microbiota, together with observational studies on multiple sclerosis patients, are here reported. Based on the collected evidence, we highlight the new potential applications of this molecule in the context of inflammatory and immune-mediated intestinal diseases.
Collapse
Affiliation(s)
- Federico Manai
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Lisa Zanoletti
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy
- Department of Chronic Diseases and Metabolism (CHROMETA), Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Davide Arfini
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Simone Giorgio De Micco
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Arolda Gjyzeli
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Sergio Comincini
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, 27100 Pavia, Italy
| | - Marialaura Amadio
- Department of Drug Sciences, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
9
|
Chen J, Cao Y, Jia O, Wang X, Luo Y, Cheuk YC, Zhu T, Zhu D, Zhang Y, Wang J. Monomethyl fumarate prevents alloimmune rejection in mouse heart transplantation by inducing tolerogenic dendritic cells. Acta Biochim Biophys Sin (Shanghai) 2023. [PMID: 37184280 DOI: 10.3724/abbs.2023088] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023] Open
Abstract
Dendritic cells (DCs) are important targets for eliciting allograft rejection after transplantation. Previous studies have demonstrated that metabolic reprogramming of DCs can transform their immune functions and induce their differentiation into tolerogenic DCs. In this study, we aim to investigate the protective effects and mechanisms of monomethyl fumarate (MMF), a bioactive metabolite of fumaric acid esters, in a mouse model of allogeneic heart transplantation. Bone marrow-derived DCs are harvested and treated with MMF to determine the impact of MMF on the phenotype and immunosuppressive function of DCs by flow cytometry and T-cell proliferation assays. RNA sequencing and Seahorse analyses are performed for mature DCs and MMF-treated DCs (MMF-DCs) to investigate the underlying mechanism. Our results show that MMF prolongs the survival time of heart grafts and inhibits the activation of DCs in vivo. MMF-DCs exhibit a tolerogenic phenotype and function in vitro. RNA sequencing and Seahorse analyses reveal that MMF activates the Nrf2 pathway and mediates metabolic reprogramming. Additionally, MMF-DC infusion prolongs cardiac allograft survival, induces regulatory T cells, and inhibits T-cell activation. MMF prevents allograft rejection in mouse heart transplantation by inducing tolerogenic DCs.
Collapse
Affiliation(s)
- Juntao Chen
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai 200032, China
| | - Yirui Cao
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai 200032, China
| | - Ouyang Jia
- Nursing Department of Huashan Hospital Affiliated to Fudan University, Shanghai 200031, China
| | - Xuanchuan Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai 200032, China
| | - Yongsheng Luo
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai 200032, China
| | - Yin Celeste Cheuk
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai 200032, China
| | - Tongyu Zhu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai 200032, China
| | - Dong Zhu
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai 200032, China
- Department of Urology, Zhongshan Hospital, Fudan University (Xiamen branch), Xiamen 361015, China
| | - Yi Zhang
- Shanghai Key Laboratory of Organ Transplantation, Shanghai 200032, China
- Biomedical Research Center, Institute for Clinical Sciences, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jina Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Organ Transplantation, Shanghai 200032, China
| |
Collapse
|
10
|
Kohle F, Dalakas MC, Lehmann HC. Repurposing MS immunotherapies for CIDP and other autoimmune neuropathies: unfulfilled promise or efficient strategy? Ther Adv Neurol Disord 2023; 16:17562864221137129. [PMID: 36620728 PMCID: PMC9810996 DOI: 10.1177/17562864221137129] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 10/19/2022] [Indexed: 01/03/2023] Open
Abstract
Despite advances in the treatment of chronic inflammatory demyelinating polyradiculoneuropathy (CIDP) and other common autoimmune neuropathies (AN), still-many patients with these diseases do not respond satisfactorily to the available treatments. Repurposing of disease-modifying therapies (DMTs) from other autoimmune conditions, particularly multiple sclerosis (MS) and neuromyelitis optica spectrum disorders (NMOSD), is a promising strategy that may accelerate the establishment of novel treatment choices for AN. This approach appears attractive due to homologies in the pathogenesis of these diseases and the extensive post-marketing experience that has been gathered from treating MS and NMOSD patients. The idea is also strengthened by a number of studies that explored the efficacy of DMTs in animal models of AN but also in some CIDP patients. We here review the available preclinical and clinical data of approved MS therapeutics in terms of their applicability to AN, especially CIDP. Promising therapeutic approaches appear to be B cell-directed and complement-targeting strategies, such as anti-CD20/anti-CD19 agents, Bruton's tyrosine kinase inhibitors and anti-C5 agents, as they exert their effects in the periphery. This is a major advantage because, in contrast to MS, their action in the periphery is sufficient to exert significant immunomodulation.
Collapse
Affiliation(s)
- Felix Kohle
- Department of Neurology, Faculty of Medicine,
University of Cologne and University Hospital Cologne, Cologne,
Germany
| | - Marinos C. Dalakas
- Department of Neurology, Thomas Jefferson
University, Philadelphia, PA, USA
- Neuroimmunology Unit, National and Kapodistrian
University of Athens Medical School, Athens, Greece
| | - Helmar C. Lehmann
- Department of Neurology, Faculty of Medicine,
University of Cologne and University Hospital Cologne, Kerpener Strasse, 62,
50937 Cologne, Germany
| |
Collapse
|
11
|
Wu Z, Bian Y, Chu T, Wang Y, Man S, Song Y, Wang Z. The role of angiogenesis in melanoma: Clinical treatments and future expectations. Front Pharmacol 2022; 13:1028647. [PMID: 36588679 PMCID: PMC9797529 DOI: 10.3389/fphar.2022.1028647] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/30/2022] [Indexed: 12/23/2022] Open
Abstract
The incidence of melanoma has increased rapidly over the past few decades, with mortality accounting for more than 75% of all skin cancers. The high metastatic potential of Melanoma is an essential factor in its high mortality. Vascular angiogenic system has been proved to be crucial for the metastasis of melanoma. An in-depth understanding of angiogenesis will be of great benefit to melanoma treatment and may promote the development of melanoma therapies. This review summarizes the recent advances and challenges of anti-angiogenic agents, including monoclonal antibodies, tyrosine kinase inhibitors, human recombinant Endostatin, and traditional Chinese herbal medicine. We hope to provide a better understanding of the mechanisms, clinical research progress, and future research directions of melanoma.
Collapse
Affiliation(s)
- Zhuzhu Wu
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China,Institute for Literature and Culture of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yifei Bian
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Tianjiao Chu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuman Wang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shuai Man
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China,Key Laboratory of Traditional Chinese Medicine for Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China,Shandong Provincial Key Laboratory of Traditional Chinese Medicine for Basic Research, Shandong University of Traditional Chinese Medicine, Jinan, China,*Correspondence: Shuai Man, ; Yongmei Song, ; Zhenguo Wang,
| | - Yongmei Song
- Institute for Literature and Culture of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China,*Correspondence: Shuai Man, ; Yongmei Song, ; Zhenguo Wang,
| | - Zhenguo Wang
- Institute for Literature and Culture of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China,Key Laboratory of Traditional Chinese Medicine for Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China,*Correspondence: Shuai Man, ; Yongmei Song, ; Zhenguo Wang,
| |
Collapse
|
12
|
Bizoń A, Chojdak-Łukasiewicz J, Kołtuniuk A, Budrewicz S, Pokryszko-Dragan A, Piwowar A. Evaluation of Selected Oxidant/Antioxidant Parameters in Patients with Relapsing-Remitting Multiple Sclerosis Undergoing Disease-Modifying Therapies. Antioxidants (Basel) 2022; 11:antiox11122416. [PMID: 36552624 PMCID: PMC9774652 DOI: 10.3390/antiox11122416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/02/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022] Open
Abstract
The aim of this study was to evaluate oxidative stress parameters, specifically the concentration of advanced oxidation protein products (AOPP) and ferric-reducing antioxidant power (FRAP), in the serum of patients with relapsing-remitting multiple sclerosis (RRMS). We also analyzed the relationships between each parameter and selected clinical/laboratory multiple-sclerosis-related parameters. The study group comprised 204 patients with RRMS and 29 healthy, age-matched controls. The concentration of AOPP was significantly higher in the RRMS patients than in controls. ROC analysis showed the ability of AOPP to distinguish between the patients with RRMS and controls (the value of AUC was 94.8%, with a sensitivity of 89.69% and specificity of 89.3%). AOPP and FRAP were significantly higher in male than in female RRMS patients. Correlations were found between AOPP and the laboratory markers of inflammation. AOPP differed in the subgroups of patients treated with particular medications. Our findings indicate an increase in the markers of oxidative stress in the serum of RRMS patients, possibly linked with chronic inflammation. Gender and type of treatment affected the markers of oxidative stress.
Collapse
Affiliation(s)
- Anna Bizoń
- Department of Toxicology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland
- Correspondence: ; Tel.: +48-71-784-0455
| | | | - Aleksandra Kołtuniuk
- Department of Nervous System Diseases, Faculty of Health Sciences, Wroclaw Medical University, Bartla 5, 51-618 Wroclaw, Poland
| | - Sławomir Budrewicz
- Department of Neurology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland
| | - Anna Pokryszko-Dragan
- Department of Neurology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland
| | - Agnieszka Piwowar
- Department of Toxicology, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland
| |
Collapse
|
13
|
Liu R, Du S, Zhao L, Jain S, Sahay K, Rizvanov A, Lezhnyova V, Khaibullin T, Martynova E, Khaiboullina S, Baranwal M. Autoreactive lymphocytes in multiple sclerosis: Pathogenesis and treatment target. Front Immunol 2022; 13:996469. [PMID: 36211343 PMCID: PMC9539795 DOI: 10.3389/fimmu.2022.996469] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 09/08/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) characterized by destruction of the myelin sheath structure. The loss of myelin leads to damage of a neuron’s axon and cell body, which is identified as brain lesions on magnetic resonance image (MRI). The pathogenesis of MS remains largely unknown. However, immune mechanisms, especially those linked to the aberrant lymphocyte activity, are mainly responsible for neuronal damage. Th1 and Th17 populations of lymphocytes were primarily associated with MS pathogenesis. These lymphocytes are essential for differentiation of encephalitogenic CD8+ T cell and Th17 lymphocyte crossing the blood brain barrier and targeting myelin sheath in the CNS. B-lymphocytes could also contribute to MS pathogenesis by producing anti-myelin basic protein antibodies. In later studies, aberrant function of Treg and Th9 cells was identified as contributing to MS. This review summarizes the aberrant function and count of lymphocyte, and the contributions of these cell to the mechanisms of MS. Additionally, we have outlined the novel MS therapeutics aimed to amend the aberrant function or counts of these lymphocytes.
Collapse
Affiliation(s)
- Rongzeng Liu
- Department of Immunology, School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang, China
| | - Shushu Du
- Department of Immunology, School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang, China
| | - Lili Zhao
- Department of Immunology, School of Basic Medical Sciences, Henan University of Science and Technology, Luoyang, China
| | - Sahil Jain
- Department of Biochemistry and Molecular Biology, Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Kritika Sahay
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India
| | - Albert Rizvanov
- Gene and cell Department, Kazan Federal University, Kazan, Russia
| | - Vera Lezhnyova
- Gene and cell Department, Kazan Federal University, Kazan, Russia
| | - Timur Khaibullin
- Neurological Department, Republican Clinical Neurological Center, Kazan, Russia
| | | | - Svetlana Khaiboullina
- Gene and cell Department, Kazan Federal University, Kazan, Russia
- *Correspondence: Svetlana Khaiboullina, ; Manoj Baranwal, ;
| | - Manoj Baranwal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India
- *Correspondence: Svetlana Khaiboullina, ; Manoj Baranwal, ;
| |
Collapse
|
14
|
Eguchi N, Damyar K, Alexander M, Dafoe D, Lakey JRT, Ichii H. Anti-Oxidative Therapy in Islet Cell Transplantation. Antioxidants (Basel) 2022; 11:1038. [PMID: 35739935 PMCID: PMC9219662 DOI: 10.3390/antiox11061038] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/16/2022] [Accepted: 05/18/2022] [Indexed: 01/27/2023] Open
Abstract
Islet cell transplantation has become a favorable therapeutic approach in the treatment of Type 1 Diabetes due to the lower surgical risks and potential complications compared to conventional pancreas transplantation. Despite significant improvements in islet cell transplantation outcomes, several limitations hamper long-term graft survival due to tremendous damage and loss of islet cells during the islet cell transplantation process. Oxidative stress has been identified as an omnipresent stressor that negatively affects both the viability and function of isolated islets. Furthermore, it has been established that at baseline, pancreatic β cells exhibit reduced antioxidative capacity, rendering them even more susceptible to oxidative stress during metabolic stress. Thus, identifying antioxidants capable of conferring protection against oxidative stressors present throughout the islet transplantation process is a valuable approach to improving the overall outcomes of islet cell transplantation. In this review we discuss the potential application of antioxidative therapy during each step of islet cell transplantation.
Collapse
Affiliation(s)
- Natsuki Eguchi
- Department of Surgery, University of California, Irvine, CA 92697, USA; (N.E.); (K.D.); (M.A.); (D.D.); (J.R.T.L.)
| | - Kimia Damyar
- Department of Surgery, University of California, Irvine, CA 92697, USA; (N.E.); (K.D.); (M.A.); (D.D.); (J.R.T.L.)
| | - Michael Alexander
- Department of Surgery, University of California, Irvine, CA 92697, USA; (N.E.); (K.D.); (M.A.); (D.D.); (J.R.T.L.)
| | - Donald Dafoe
- Department of Surgery, University of California, Irvine, CA 92697, USA; (N.E.); (K.D.); (M.A.); (D.D.); (J.R.T.L.)
| | - Jonathan R. T. Lakey
- Department of Surgery, University of California, Irvine, CA 92697, USA; (N.E.); (K.D.); (M.A.); (D.D.); (J.R.T.L.)
- Department of Biomedical Engineering, University of California, Irvine, CA 92686, USA
| | - Hirohito Ichii
- Department of Surgery, University of California, Irvine, CA 92697, USA; (N.E.); (K.D.); (M.A.); (D.D.); (J.R.T.L.)
| |
Collapse
|
15
|
Eguchi N, Toribio AJ, Alexander M, Xu I, Whaley DL, Hernandez LF, Dafoe D, Ichii H. Dysregulation of β-Cell Proliferation in Diabetes: Possibilities of Combination Therapy in the Development of a Comprehensive Treatment. Biomedicines 2022; 10:biomedicines10020472. [PMID: 35203680 PMCID: PMC8962301 DOI: 10.3390/biomedicines10020472] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/12/2022] [Accepted: 02/15/2022] [Indexed: 02/01/2023] Open
Abstract
Diabetes mellitus (DM) is a metabolic disorder characterized by chronic hyperglycemia as a result of insufficient insulin levels and/or impaired function as a result of autoimmune destruction or insulin resistance. While Type 1 DM (T1DM) and Type 2 DM (T2DM) occur through different pathological processes, both result in β-cell destruction and/or dysfunction, which ultimately lead to insufficient β-cell mass to maintain normoglycemia. Therefore, therapeutic agents capable of inducing β-cell proliferation is crucial in treating and reversing diabetes; unfortunately, adult human β-cell proliferation has been shown to be very limited (~0.2% of β-cells/24 h) and poorly responsive to many mitogens. Furthermore, diabetogenic insults result in damage to β cells, making it ever more difficult to induce proliferation. In this review, we discuss β-cell mass/proliferation pathways dysregulated in diabetes and current therapeutic agents studied to induce β-cell proliferation. Furthermore, we discuss possible combination therapies of proliferation agents with immunosuppressants and antioxidative therapy to improve overall long-term outcomes of diabetes.
Collapse
|
16
|
Ulasov AV, Rosenkranz AA, Georgiev GP, Sobolev AS. Nrf2/Keap1/ARE signaling: Towards specific regulation. Life Sci 2022; 291:120111. [PMID: 34732330 PMCID: PMC8557391 DOI: 10.1016/j.lfs.2021.120111] [Citation(s) in RCA: 258] [Impact Index Per Article: 86.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/22/2021] [Accepted: 10/27/2021] [Indexed: 02/06/2023]
Abstract
The Nrf2 transcription factor governs the expression of hundreds genes involved in cell defense against oxidative stress, the hallmark of numerous diseases such as neurodegenerative, cardiovascular, some viral pathologies, diabetes and others. The main route for Nrf2 activity regulation is via interactions with the Keap1 protein. Under the normoxia the Keap1 binds the Nrf2 and targets it to the proteasomal degradation, while the Keap1 is regenerated. Upon oxidative stress the interactions between Nrf2 and Keap1 are interrupted and the Nrf2 activates the transcription of the protective genes. Currently, the Nrf2 system activation is considered as a powerful cytoprotective strategy for treatment of different pathologies, which pathogenesis relies on oxidative stress including viral diseases of pivotal importance such as COVID-19. The implementation of this strategy is accomplished mainly through the inactivation of the Keap1 "guardian" function. Two approaches are now developing: the Keap1 modification via electrophilic agents, which leads to the Nrf2 release, and direct interruption of the Nrf2:Keap1 protein-protein interactions (PPI). Because of theirs chemical structure, the Nrf2 electrophilic inducers could non-specifically interact with others cellular proteins leading to undesired effects. Whereas the non-electrophilic inhibitors of the Nrf2:Keap1 PPI could be more specific, thereby widening the therapeutic window.
Collapse
Affiliation(s)
- Alexey V Ulasov
- Department of Molecular Genetics of Intracellular Transport, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov St., 119334 Moscow, Russia.
| | - Andrey A Rosenkranz
- Department of Molecular Genetics of Intracellular Transport, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov St., 119334 Moscow, Russia; Faculty of Biology, Moscow State University, 1-12 Leninskiye Gory St., 119234 Moscow, Russia
| | - Georgii P Georgiev
- Department of Molecular Genetics of Intracellular Transport, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov St., 119334 Moscow, Russia
| | - Alexander S Sobolev
- Department of Molecular Genetics of Intracellular Transport, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov St., 119334 Moscow, Russia; Faculty of Biology, Moscow State University, 1-12 Leninskiye Gory St., 119234 Moscow, Russia
| |
Collapse
|
17
|
Baryła M, Semeniuk-Wojtaś A, Róg L, Kraj L, Małyszko M, Stec R. Oncometabolites-A Link between Cancer Cells and Tumor Microenvironment. BIOLOGY 2022; 11:biology11020270. [PMID: 35205136 PMCID: PMC8869548 DOI: 10.3390/biology11020270] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/31/2022] [Accepted: 02/02/2022] [Indexed: 12/12/2022]
Abstract
The tumor microenvironment is the space between healthy tissues and cancer cells, created by the extracellular matrix, blood vessels, infiltrating cells such as immune cells, and cancer-associated fibroblasts. These components constantly interact and influence each other, enabling cancer cells to survive and develop in the host organism. Accumulated intermediate metabolites favoring dysregulation and compensatory responses in the cell, called oncometabolites, provide a method of communication between cells and might also play a role in cancer growth. Here, we describe the changes in metabolic pathways that lead to accumulation of intermediate metabolites: lactate, glutamate, fumarate, and succinate in the tumor and their impact on the tumor microenvironment. These oncometabolites are not only waste products, but also link all types of cells involved in tumor survival and progression. Oncometabolites play a particularly important role in neoangiogenesis and in the infiltration of immune cells in cancer. Oncometabolites are also associated with a disrupted DNA damage response and make the tumor microenvironment more favorable for cell migration. The knowledge summarized in this article will allow for a better understanding of associations between therapeutic targets and oncometabolites, as well as the direct effects of these particles on the formation of the tumor microenvironment. In the future, targeting oncometabolites could improve treatment standards or represent a novel method for fighting cancer.
Collapse
Affiliation(s)
- Maksymilian Baryła
- Department of Oncology, Medical University of Warsaw, 02-097 Warsaw, Poland; (M.B.); (L.R.); (L.K.); (M.M.); (R.S.)
| | - Aleksandra Semeniuk-Wojtaś
- Department of Oncology, Medical University of Warsaw, 02-097 Warsaw, Poland; (M.B.); (L.R.); (L.K.); (M.M.); (R.S.)
- Correspondence:
| | - Letycja Róg
- Department of Oncology, Medical University of Warsaw, 02-097 Warsaw, Poland; (M.B.); (L.R.); (L.K.); (M.M.); (R.S.)
| | - Leszek Kraj
- Department of Oncology, Medical University of Warsaw, 02-097 Warsaw, Poland; (M.B.); (L.R.); (L.K.); (M.M.); (R.S.)
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, 05-552 Jastrzębiec, Poland
| | - Maciej Małyszko
- Department of Oncology, Medical University of Warsaw, 02-097 Warsaw, Poland; (M.B.); (L.R.); (L.K.); (M.M.); (R.S.)
| | - Rafał Stec
- Department of Oncology, Medical University of Warsaw, 02-097 Warsaw, Poland; (M.B.); (L.R.); (L.K.); (M.M.); (R.S.)
| |
Collapse
|
18
|
Oncel I, Yousefi M, Inci A, Arslan Gulten Z, Teke Kisa P, Karaca M, Ünal Ö, Gündüz M, Kor D, Onenli Mungan N, Arslan N, Tumer L, Gucuyener K, Vural A, Anlar B. Investigating myelin oligodendrocyte glycoprotein antibodies in hereditary citrullinemia. Med Hypotheses 2022. [DOI: 10.1016/j.mehy.2022.110781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
19
|
Ikeguchi R, Shimizu Y, Kondo A, Kanda N, So H, Kojima H, Kitagawa K. Melanoma Cell Adhesion Molecule Expressing Helper T Cells in CNS Inflammatory Demyelinating Diseases. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2021; 8:8/6/e1069. [PMID: 34429366 PMCID: PMC8387012 DOI: 10.1212/nxi.0000000000001069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 07/21/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND OBJECTIVE To elucidate the relationship between melanoma cell adhesion molecule (MCAM)-expressing lymphocytes and pathogenesis of CNS inflammatory demyelinating diseases (IDDs). METHODS Patients with multiple sclerosis (MS) (n = 72) and neuromyelitis optica spectrum disorder (NMOSD, n = 29) were included. We analyzed the frequency and absolute numbers of MCAM+ lymphocytes (memory helper T [mTh] cells, naive helper T cells, CD8+ T cells, and B cells) in the peripheral blood (PB) and the CSF of patients with MS and NMOSD, treated with/without disease-modifying drugs (DMDs) or steroids, using flow cytometry. RESULTS The frequency of MCAM+ cells was higher in the mTh cell subset than that in other lymphocyte subsets. A significant increase in the frequency and the absolute number of MCAM+ mTh cells was observed in the PB of patients with NMOSD, whereas no increase was observed in the PB of patients with MS. The frequency of CSF MCAM+ mTh cells was higher in relapsing patients with MS and NMOSD than that in the control group. Although there was no difference in the frequencies of MCAM+ lymphocytes among the DMD-treated groups, fingolimod decreased the absolute number of MCAM+ lymphocytes. DISCUSSION MCAM+ mTh cells were elevated in the CSF of relapsing patients with MS and in both the PB and CSF of patients with NMOSD. These results indicate that MCAM contributes to the pathogenesis of MS and NMOSD through different mechanisms. MCAM could be a therapeutic target of CNS IDDs, and further study is needed to elucidate the underlying mechanism of MCAM in CNS IDD pathogenesis.
Collapse
Affiliation(s)
- Ryotaro Ikeguchi
- From the Department of Neurology, Tokyo Women's Medical University, Japan
| | - Yuko Shimizu
- From the Department of Neurology, Tokyo Women's Medical University, Japan.
| | - Akihiro Kondo
- From the Department of Neurology, Tokyo Women's Medical University, Japan
| | - Natsuki Kanda
- From the Department of Neurology, Tokyo Women's Medical University, Japan
| | - Hayato So
- From the Department of Neurology, Tokyo Women's Medical University, Japan
| | - Haruka Kojima
- From the Department of Neurology, Tokyo Women's Medical University, Japan
| | - Kazuo Kitagawa
- From the Department of Neurology, Tokyo Women's Medical University, Japan
| |
Collapse
|
20
|
Sivinski J, Zhang DD, Chapman E. Targeting NRF2 to treat cancer. Semin Cancer Biol 2021; 76:61-73. [PMID: 34102289 DOI: 10.1016/j.semcancer.2021.06.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 12/17/2022]
Abstract
NRF2 is a basic leucine zipper (bZip) transcription factor that is the master regulator of redox homeostasis. Under basal conditions, the cellular level of NRF2 is low due to a posttranslational regulation by the ubiquitin proteasome system (UPS). But, when an organism is challenged with oxidative or xenobiotic stress, the NRF2 pathway is activated by inhibition of the E3 ubiquitin ligase complex that normally marks NRF2 for destruction. For several decades, researchers have searched for molecules that can intentionally activate NRF2, as this was shown to be a means to prevent certain diseases, at least in animal models. In the present era, there are many compounds known to activate the NRF2 pathway including natural products and synthetic compounds, covalent and non-covalent compounds, and others. However, it was also revealed that like many protective pathways, the NRF2 pathway has a dark side. Just as NRF2 can protect normal cells from damage, it can protect malignant cells from damage. As cells transform, they are exposed to many stressors and aberrant upregulation of NRF2 can facilitate transformation and it can help cancer cells to grow, to spread, and to resist treatment. For this reason, researchers are also interested in the discovery and development of NRF2 inhibitors. In the present review, we will begin with a general discussion of NRF2 structure and function, we will discuss the latest in NRF2 non-covalent activators, and we will discuss the current state of NRF2 inhibitors.
Collapse
Affiliation(s)
- Jared Sivinski
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721, USA
| | - Donna D Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721, USA
| | - Eli Chapman
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ, 85721, USA.
| |
Collapse
|
21
|
Alborghetti M, Bellucci G, Gentile A, Calderoni C, Nicoletti F, Capra R, Salvetti M, Centonze D. Drugs used in the treatment of multiple sclerosis during COVID-19 pandemic: a critical viewpoint. Curr Neuropharmacol 2021; 20:107-125. [PMID: 33784961 PMCID: PMC9199540 DOI: 10.2174/1570159x19666210330094017] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 03/15/2021] [Accepted: 03/24/2021] [Indexed: 11/22/2022] Open
Abstract
Since COVID-19 has emerged as a word public health problem, attention has been focused on how immune-suppressive drugs used for the treatment of autoimmune disorders influence the risk for SARS-CoV-2 infection and the development of acute respiratory distress syndrome (ARDS). Here, we discuss the disease-modifying agents approved for the treatment of multiple sclerosis (MS) within this context. Interferon (IFN)-β1a and -1b, which display antiviral activity, could be protective in the early stage of COVID-19 infection, although SARS-CoV-2 may have developed resistance to IFNs. However, in the hyperinflammation stage, IFNs may become detrimental by facilitating macrophage invasion in the lung and other organs. Glatiramer acetate and its analogues should not interfere with the development of COVID-19 and may be considered safe. Teriflunomide, a first-line oral drug used in the treatment of relapsing-remitting MS (RRMS), may display antiviral activity by depleting cellular nucleotides necessary for viral replication. The other first-line drug, dimethyl fumarate, may afford protection against SARS-CoV-2 by activating the Nrf-2 pathway and reinforcing the cellular defenses against oxidative stress. Concern has been raised regarding the use of second-line treatments for MS during the COVID-19 pandemic. However, this concern is not always justified. For example, fingolimod might be highly beneficial during the hyperinflammatory stage of COVID-19 for a number of mechanisms, including the reinforcement of the endothelial barrier. Caution is suggested for the use of natalizumab, cladribine, alemtuzumab, and ocrelizumab, although MS disease recurrence after discontinuation of these drugs may overcome a potential risk for COVID-19 infection.
Collapse
Affiliation(s)
- Marika Alborghetti
- Departments of Neuroscience Mental Health and Sensory Organs (NESMOS), University Sapienza of Rome. Italy
| | - Gianmarco Bellucci
- Departments of Neuroscience Mental Health and Sensory Organs (NESMOS), University Sapienza of Rome. Italy
| | - Antonietta Gentile
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, 00166 Rome. Italy
| | - Chiara Calderoni
- Departments of Physiology and Pharmacology, University Sapienza of Rome. Italy
| | | | - Ruggero Capra
- Multiple Sclerosis Center, ASST Ospedali Civili, Brescia. Italy
| | - Marco Salvetti
- Departments of Neuroscience Mental Health and Sensory Organs (NESMOS),University Sapienza of Rome. Italy
| | - Diego Centonze
- Department of Systems Medicine, Tor Vergata University, 00133 Rome. Italy
| |
Collapse
|
22
|
Hosseini A, Gharibi T, Mohammadzadeh A, Ebrahimi-Kalan A, Jadidi-Niaragh F, Babaloo Z, Shanehbandi D, Baghbani E, Baradaran B. Ruxolitinib attenuates experimental autoimmune encephalomyelitis (EAE) development as animal models of multiple sclerosis (MS). Life Sci 2021; 276:119395. [PMID: 33781828 DOI: 10.1016/j.lfs.2021.119395] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 03/19/2021] [Accepted: 03/22/2021] [Indexed: 01/07/2023]
Abstract
AIMS STAT3 signaling is critical for Th17 development that plays an important role in multiple sclerosis pathogenesis. To evaluate the anti-inflammatory and regulatory T cells effects of JAK1/2 and STAT3 inhibition, we assessed the JAK 1/2 inhibitor ruxolitinib effects on Th17 cell/Tregs balance. MAIN METHODS Ruxolitinib was administered to experimental autoimmune encephalomyelitis (EAE) mice via oral gavage, and its effects were assessed. The expression of pro-inflammatory and anti-inflammatory cytokines, including IL-17A and IL-10, were analyzed by real-time PCR. The frequency of Th17 cells and Tregs were evaluated by flow cytometry. KEY FINDING Ruxolitinib ameliorated the EAE severity and decreased the proportion of Th17 cells and inflammatory markers levels. In contrast, the balance of Tregs and the level of anti-inflammatory cytokine were increased in ruxolitinib-treated mice. Furthermore, ruxolitinib markedly decreased the expression of Th17 related transcription factor, RORɣt, whereas FOXP3 expression associated with Treg differentiation was increased. SIGNIFICANCE Our results show that ruxolitinib may be a promising therapeutic strategy for multiple sclerosis.
Collapse
Affiliation(s)
- Arezoo Hosseini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Gharibi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Adel Mohammadzadeh
- Department of Immunology and Genetics, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Abbas Ebrahimi-Kalan
- Neuroscience Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Neurosciences and Cognition, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Jadidi-Niaragh
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zohreh Babaloo
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Baghbani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
23
|
Chen K, Wu S, Ye S, Huang H, Zhou Y, Zhou H, Wu S, Mao Y, Shangguan F, Lan L, Chen B. Dimethyl Fumarate Induces Metabolic Crisie to Suppress Pancreatic Carcinoma. Front Pharmacol 2021; 12:617714. [PMID: 33692690 PMCID: PMC7937954 DOI: 10.3389/fphar.2021.617714] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 01/15/2021] [Indexed: 01/22/2023] Open
Abstract
Dimethyl fumarate (DMF) is an approved drug used in the treatment of multiple sclerosis (MS) and psoriasis therapy. Multiple studies have demonstrated other pharmacological activities of DMF such as an anti-cancer agent. In particular, studies have shown that DMF can modulate the NRF2/HO1/NQO1 antioxidant signal pathway and inactivate NF-κB to suppress the growth of colon and breast cancer cells, and induce cell death. In this study, we aimed to evaluate the anti-tumor activities of DMF in pancreatic cancer (PC) focusing on cell death as the predominant mechanism of response. We showed that both mitochondrial respiration and aerobic glycolysis were severely depressed following treatment with DMF and the effects could be abrogated by treatment with L-cysteine and N-acetyl-L-cysteine (NAC). Importantly, we verified that DMF induced metabolic crisis and that cell death was not related to alterations in ROS. Our data implied that MTHFD1 could be a potential downstream target of DMF identified by molecular docking analysis. Finally, we confirmed that MTHFD1 is up-regulated in PC and overexpression of MTHFD1 was negatively related to outcomes of PC patients. Our data indicate that DMF induces metabolic crisie to suppress cell growth and could be a potential novel therapy in the treatment of PC.
Collapse
Affiliation(s)
- Kaiyuan Chen
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shanshan Wu
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Laboratory of Precision Medical Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Sisi Ye
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Laboratory of Precision Medical Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Huimin Huang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yi Zhou
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hongfei Zhou
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shijia Wu
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yefan Mao
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Fugen Shangguan
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Linhua Lan
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Bicheng Chen
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
24
|
Choi I, Son H, Baek JH. Tricarboxylic Acid (TCA) Cycle Intermediates: Regulators of Immune Responses. Life (Basel) 2021; 11:69. [PMID: 33477822 PMCID: PMC7832849 DOI: 10.3390/life11010069] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/15/2021] [Accepted: 01/18/2021] [Indexed: 02/06/2023] Open
Abstract
The tricarboxylic acid cycle (TCA) is a series of chemical reactions used in aerobic organisms to generate energy via the oxidation of acetylcoenzyme A (CoA) derived from carbohydrates, fatty acids and proteins. In the eukaryotic system, the TCA cycle occurs completely in mitochondria, while the intermediates of the TCA cycle are retained inside mitochondria due to their polarity and hydrophilicity. Under cell stress conditions, mitochondria can become disrupted and release their contents, which act as danger signals in the cytosol. Of note, the TCA cycle intermediates may also leak from dysfunctioning mitochondria and regulate cellular processes. Increasing evidence shows that the metabolites of the TCA cycle are substantially involved in the regulation of immune responses. In this review, we aimed to provide a comprehensive systematic overview of the molecular mechanisms of each TCA cycle intermediate that may play key roles in regulating cellular immunity in cell stress and discuss its implication for immune activation and suppression.
Collapse
Affiliation(s)
| | | | - Jea-Hyun Baek
- School of Life Science, Handong Global University, Pohang, Gyeongbuk 37554, Korea; (I.C.); (H.S.)
| |
Collapse
|
25
|
Pilo de la Fuente B, Sabín J, Galán V, Thuissard I, Sainz de la Maza S, Costa-Frossard L, Gómez-Moreno M, Díaz-Díaz J, Oreja-Guevara C, Lozano-Ros A, García-Domínguez JM, Borrego L, Ayuso L, Castro A, Sánchez P, Meca-Lallana V, Muñoz C, Casanova I, López de Silanes C, Martín H, Rodríguez-García E, Andreu-Vázquez C, Blasco R, García-Merino JA, Aladro Y. Three-Year Effectiveness of Dimethyl Fumarate in Multiple Sclerosis: A Prospective Multicenter Real-World Study. CNS Drugs 2020; 34:1275-1286. [PMID: 33226562 DOI: 10.1007/s40263-020-00775-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/05/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Dimethyl fumarate (DMF) has demonstrated efficacy in phase III studies. However, real-world data are still limited. OBJECTIVE The objective of this study was to describe the profile of patients who receive DMF and to assess the effectiveness of DMF regarding relapses, disability progression, magnetic resonance imaging activity, and NEDA (No Evidence Disease Activity)-3 status in a Spanish population in a real-world setting. METHODS We conducted a multicenter prospective study of patients who started DMF between 2014 and 2019 in Spain. Three subgroups were considered: naïve, switch to DMF because of inefficacy, and switch to DMF because of adverse effects. The effects of DMF on clinical and radiological measures were evaluated. RESULTS Among 886 patients, 25.3% were naïve, 28.8% switched because of adverse effects, and 45.9% because of inefficacy. Median follow-up was 38.9 (interquartile range 22.6-41.8) months. Annualized relapse rates were 0.15, 0.10, and 0.10 at 12, 24, and 36 months respectively, and 77.7% of patients were relapse free at month 42. At 12, 24, and 42 months, 96.1%, 87.4%, and 79.7% of patients were progression free, respectively. The number of T1 gadolinium-enhancement (T1Gd+) lesions was 0.19, 0.14, and 0.18 at 12, 24, and 36 months. NEDA-3 status at month 42 was maintained by 49.8% of patients. Relapsing was associated with higher annualized relapse rates the year before (hazard ratio 1.34, p < 0.001) and to the inefficacy switch vs naïve group (hazard ratio 1.76, p = 0.003). A higher baseline Expanded Disability Status Scale score was associated with disability progression (hazard ratio 1.15, p = 0.003) and more T1Gd+ lesions (hazard ratio 1.07, p < 0.001) with radiological progression. A higher baseline Expanded Disability Status Scale score, a larger number of T1Gd+ lesions, and a switch because of inefficacy (vs adverse events) were all risk factors for losing NEDA-3 status. DMF was discontinued in 29.9% of patients, in 13.5% because of inefficacy. CONCLUSIONS Our findings confirm the sustained effectiveness of DMF on the clinical and radiological activity of multiple sclerosis in a real-world setting, both in naïve patients and in those switching from other multiple sclerosis therapies.
Collapse
Affiliation(s)
- Belen Pilo de la Fuente
- Multiple Sclerosis Unit, Department of Neurology, S. de Neurología, Hospital Universitario de Getafe, Carretera Toledo Km 12.5, Getafe, 28905, Madrid, Spain
| | - Julia Sabín
- Multiple Sclerosis Unit, Department of Neurology, Hospital Universitario de Puerta de Hierro Majadahonda, Madrid, Spain
| | - Victoria Galán
- Multiple Sclerosis Unit, Department of Neurology, Hospital Universitario, 12 de Octubre, Madrid, Spain
| | - Israel Thuissard
- Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, Madrid, Spain
| | - Susana Sainz de la Maza
- Multiple Sclerosis Unit, Department of Neurology, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Lucienne Costa-Frossard
- Multiple Sclerosis Unit, Department of Neurology, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Mayra Gómez-Moreno
- Department of Neurology, Hospital Universitario "Infanta Leonor", Madrid, Spain
| | - Judit Díaz-Díaz
- Multiple Sclerosis Unit, Department of Neurology, Hospital Universitario Clínico San Carlos, Madrid, Spain
| | - Celia Oreja-Guevara
- Multiple Sclerosis Unit, Department of Neurology, Hospital Universitario Clínico San Carlos, Madrid, Spain
| | - Alberto Lozano-Ros
- Multiple Sclerosis Unit, Department of Neurology, Hospital Universitario "Gregorio Marañón", Madrid, Spain
| | - José M García-Domínguez
- Multiple Sclerosis Unit, Department of Neurology, Hospital Universitario "Gregorio Marañón", Madrid, Spain
| | - Laura Borrego
- Department of Neurology, Hospital Universitario "Fundación de Alcorcón", Madrid, Spain
| | - Lucía Ayuso
- Department of Neurology, Hospital Universitario "Príncipe de Asturias", Alcalá de Henares, Madrid, Spain
| | - Andy Castro
- Department of Neurology, Hospital Universitario "Príncipe de Asturias", Alcalá de Henares, Madrid, Spain
| | - Pedro Sánchez
- Multiple Sclerosis Unit, Department of Neurology, Hospital Universitario "La Princesa", Madrid, Spain
| | - Virginia Meca-Lallana
- Multiple Sclerosis Unit, Department of Neurology, Hospital Universitario "La Princesa", Madrid, Spain
| | - Carmen Muñoz
- Department of Neurology, Hospital Complejo Torrecárdenas, Almería, Spain
| | - Ignacio Casanova
- Department of Neurology, Hospital Universitario de Torrejon, Madrid, Spain
| | | | - Hugo Martín
- Department of Neurology, Hospital Universitario "Infanta Cristina", Madrid, Spain
| | | | | | - Rosario Blasco
- Multiple Sclerosis Unit, Department of Neurology, Hospital Universitario de Puerta de Hierro Majadahonda, Madrid, Spain
| | - Juan A García-Merino
- Multiple Sclerosis Unit, Department of Neurology, Hospital Universitario de Puerta de Hierro Majadahonda, Madrid, Spain
| | - Yolanda Aladro
- Multiple Sclerosis Unit, Department of Neurology, S. de Neurología, Hospital Universitario de Getafe, Carretera Toledo Km 12.5, Getafe, 28905, Madrid, Spain.
| | | |
Collapse
|
26
|
Zouali M. DNA methylation signatures of autoimmune diseases in human B lymphocytes. Clin Immunol 2020; 222:108622. [PMID: 33188932 DOI: 10.1016/j.clim.2020.108622] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/28/2020] [Accepted: 11/07/2020] [Indexed: 12/15/2022]
Abstract
B lymphocytes play key roles in adaptive and innate immunity. In autoimmune diseases, their participation in disease instigation and/or progression has been demonstrated in both experimental models and clinical trials. Recent epigenetic investigations of human B lymphocyte subsets revealed the importance of DNA methylation in exquisitely regulating the cellular activation and differentiation programs. This review discusses recent advances on the potential of DNA methylation to shape events that impart generation of plasma cells and memory B cells, providing novel insight into homeostatic regulation of the immune system. In parallel, epigenetic profiling of B cells from patients with systemic or organo-specific autoimmune diseases disclosed distinctive differential methylation regions that, in some cases, could stratify patients from controls. Development of tools for editing DNA methylation in the mammalian genome could be useful for future functional studies of epigenetic regulation by offering the possibility to edit locus-specific methylation, with potential translational applications.
Collapse
Affiliation(s)
- Moncef Zouali
- Graduate Institute of Biomedical Sciences, China Medical University, No. 91, Xueshi Road, North District, Taichung City, Taïwan 404, Taichung, Taiwan.
| |
Collapse
|
27
|
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
|
28
|
Abstract
Covering: up to 2020The transcription factor NRF2 is one of the body's major defense mechanisms, driving transcription of >300 antioxidant response element (ARE)-regulated genes that are involved in many critical cellular processes including redox regulation, proteostasis, xenobiotic detoxification, and primary metabolism. The transcription factor NRF2 and natural products have an intimately entwined history, as the discovery of NRF2 and much of its rich biology were revealed using natural products both intentionally and unintentionally. In addition, in the last decade a more sinister aspect of NRF2 biology has been revealed. NRF2 is normally present at very low cellular levels and only activated when needed, however, it has been recently revealed that chronic, high levels of NRF2 can lead to diseases such as diabetes and cancer, and may play a role in other diseases. Again, this "dark side" of NRF2 was revealed and studied largely using a natural product, the quassinoid, brusatol. In the present review, we provide an overview of NRF2 structure and function to orient the general reader, we will discuss the history of NRF2 and NRF2-activating compounds and the biology these have revealed, and we will delve into the dark side of NRF2 and contemporary issues related to the dark side biology and the role of natural products in dissecting this biology.
Collapse
Affiliation(s)
- Donna D Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ 85721, USA.
| | | |
Collapse
|
29
|
Bombeiro AL, Pereira BTN, Bonfanti AP, Oliveira ALRD. Immunomodulation by dimethyl fumarate treatment improves mouse sciatic nerve regeneration. Brain Res Bull 2020; 160:24-32. [PMID: 32305403 DOI: 10.1016/j.brainresbull.2020.04.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/13/2020] [Accepted: 04/07/2020] [Indexed: 02/07/2023]
Abstract
Traumatic injury to the peripheral nervous system (PNS) often generates sensorimotor deficits that impair the quality of life of the patient. The success of nerve regeneration is related to tissue clearance and the formation of a microenvironment that sustains and stimulates axon growth up to the target. In this sense, macrophages are important for axon and myelin debris removal, neovascularization and the production of neurotrophic factors. Macrophage activation is improved by T helper (Th) lymphocytes, whose role remains few explored upon traumatic nerve injuries. Dimethyl fumarate (DMF) is the first-line drug for the treatment of multiple sclerosis due to its neuroprotective, anti-inflammatory and immunomodulatory properties. DMF improves nerve regeneration via antioxidant and cytoprotective cell signaling pathways. However, the direct activity on the cell immune response following nerve axotomy requires further investigation. In the present study, we evaluated DMF activity on Th cells and macrophage polarization, axonal regeneration and motor recovery following sciatic nerve crush in mice. For this aim, operated animals received DMF or vehicle once a day, starting at 3 days postinjury (dpi). Using an in vivo cell migration assay, we observed reduced lymphocyte infiltration in the nerves of DMF-treated mice at 7 dpi. Flow cytometry revealed DMF-responsive lymphocyte polarization from the pro- (Th1) to anti-inflammatory (Th2) phenotype at 7 dpi but not at 14 dpi. No effect was observed on macrophage polarization (from M1 to M2), although DMF reduced the frequency of the proinflammatory M1 subset from 7 to 14 dpi. Quantification of neurofilament (axon marker) and growth-associated protein 43 (GAP-43) immunolabeling showed improved axonal regeneration under DMF treatment at 14 dpi. Better motor recovery was observed in the DMF-treated group, as verified by an automated walking track test. Overall, our data reinforce the pro-regenerative capacity of DMF after traumatic nerve injury based on downmodulation of the proinflammatory immune response.
Collapse
Affiliation(s)
- André Luis Bombeiro
- Department of Structural and Functional Biology, Institute of Biology. P.O. Box: 6109, University of Campinas - UNICAMP. 13083-970, Campinas, SP, Brazil.
| | - Bruna Toledo Nunes Pereira
- Department of Structural and Functional Biology, Institute of Biology. P.O. Box: 6109, University of Campinas - UNICAMP. 13083-970, Campinas, SP, Brazil.
| | - Amanda Pires Bonfanti
- Department of Structural and Functional Biology, Institute of Biology. P.O. Box: 6109, University of Campinas - UNICAMP. 13083-970, Campinas, SP, Brazil.
| | - Alexandre Leite Rodrigues de Oliveira
- Department of Structural and Functional Biology, Institute of Biology. P.O. Box: 6109, University of Campinas - UNICAMP. 13083-970, Campinas, SP, Brazil.
| |
Collapse
|
30
|
Najjar E, Staun-Ram E, Volkowich A, Miller A. Dimethyl fumarate promotes B cell-mediated anti-inflammatory cytokine profile in B and T cells, and inhibits immune cell migration in patients with MS. J Neuroimmunol 2020; 343:577230. [PMID: 32247228 DOI: 10.1016/j.jneuroim.2020.577230] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 02/23/2020] [Accepted: 03/25/2020] [Indexed: 01/08/2023]
Abstract
Dimethyl Fumarate (DMF), known for its mechanism of action targeting Nrf2 and related redox homeostasis, is an approved immunotherapy for patients with Multiple Sclerosis (PwMS) in the relapsing form. We assessed how DMF modulates immune cell functions, namely the cytokine profile of co-cultured B and T cells, and the chemokine-mediated migration of immune cells. Following DMF therapy, LTα+, TNFα+ and IFNγ+ B cells were reduced while TGFβ and IL10 expression elevated. B cells from DMF-treated patients increased TGFβ and LTα expression on T cells, while DMF directly reduced TNFα+ and IFNγ+ T cells. CXCL12/CXCL13-mediated migration of B cells, Monocytes, CD4 and CD8 T cells was reduced, with altered CXCR5 and CXCR4 expression. Induction of regulatory B and T cells and reduced migration of immune cells may be part of the beneficial mechanism of DMF in PwMS.
Collapse
Affiliation(s)
- Eiman Najjar
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Elsebeth Staun-Ram
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel; Neuroimmunology Unit & Multiple Sclerosis Center, Department of Neurology, Carmel Medical Center, Haifa, Israel
| | - Anat Volkowich
- Neuroimmunology Unit & Multiple Sclerosis Center, Department of Neurology, Carmel Medical Center, Haifa, Israel
| | - Ariel Miller
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel; Neuroimmunology Unit & Multiple Sclerosis Center, Department of Neurology, Carmel Medical Center, Haifa, Israel.
| |
Collapse
|
31
|
Nani BD, Sardi JDCO, Lazarini JG, Silva DR, Massariolli AP, Cunha TM, de Alencar SM, Franchin M, Rosalen PL. Anti-inflammatory and anti- Candida Effects of Brazilian Organic Propolis, a Promising Source of Bioactive Molecules and Functional Food. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:2861-2871. [PMID: 31369255 DOI: 10.1021/acs.jafc.8b07304] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Brazilian organic propolis (BOP) is an unexplored Brazilian propolis that is produced organically and certified according to international legislation. Our results showed that BOP has strong anti-inflammatory effects and acts by reducing nuclear factor κB activation, tumor necrosis factor α release, and neutrophil migration. In addition, BOP6 exhibited antifungal activity on planktonic and biofilm cultures of Candida albicans, Candida glabrata, Candida tropicalis, Candida krusei, and Candida parapsisolis and reduced in vitro yeast cell adhesion to human keratinocytes at sub-inhibitory concentrations. BOP demonstrated significantly low toxicity in Galleria melonella larvae at antifungal doses. Lastly, a chemical analysis revealed the presence of caffeoyltartaric acid, 3,4-dicaffeoylquinic acid, quercetin, and gibberellins A7, A9, and A20, which may be responsible for the biological properties observed. Thus, our data indicate that BOP is a promising source of anti-inflammatory and antifungal molecules that may be used as a functional food.
Collapse
Affiliation(s)
- Bruno Dias Nani
- Department of Physiological Sciences, Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo 13414-018, Brazil
| | - Janaína de Cássia Orlandi Sardi
- Department of Physiological Sciences, Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo 13414-018, Brazil
| | - Josy Goldoni Lazarini
- Department of Physiological Sciences, Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo 13414-018, Brazil
| | - Diego Romário Silva
- Department of Physiological Sciences, Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo 13414-018, Brazil
| | - Adna Prado Massariolli
- Department of Agri-Food Industry, Food and Nutrition, "Luiz de Queiroz" College of Agriculture, University of São Paulo, Piracicaba, São Paulo 13418-900, Brazil
| | - Thiago Mattar Cunha
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo 14049-900, Brazil
| | - Severino Matias de Alencar
- Department of Agri-Food Industry, Food and Nutrition, "Luiz de Queiroz" College of Agriculture, University of São Paulo, Piracicaba, São Paulo 13418-900, Brazil
| | - Marcelo Franchin
- Department of Physiological Sciences, Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo 13414-018, Brazil
| | - Pedro Luiz Rosalen
- Department of Physiological Sciences, Piracicaba Dental School, University of Campinas, Piracicaba, São Paulo 13414-018, Brazil
| |
Collapse
|
32
|
Mimpen M, Smolders J, Hupperts R, Damoiseaux J. Natural killer cells in multiple sclerosis: A review. Immunol Lett 2020; 222:1-11. [PMID: 32113900 DOI: 10.1016/j.imlet.2020.02.012] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/17/2020] [Accepted: 02/26/2020] [Indexed: 12/13/2022]
Abstract
As the most common non-traumatic disabling disease among adolescents, multiple sclerosis (MS) is a devastating neurological inflammatory disease of the central nervous system. Research has not yet fully elucidated its pathogenesis, but it has shown MS to be a complex, multifactorial disease with many interplaying factors. One of these factors, natural killer (NK) cells, lymphocytes of the innate immune system, have recently gained attention due to the effects of daclizumab therapy, causing an expansion of the immunoregulatory subset of NK cells. Since then, NK cells and their relation to MS have been the focus of research, with many new findings being published in the last decade. In this review, NK cells are pictured as potent cytotoxic killers, as well as unique immune-regulators. Additionally, an overview of our current knowledge regarding NK cells in MS is given. The role of NK cells in MS is reviewed in the context of well-established environmental factors and current disease modifying therapies to gain further understanding of the pathogenesis and treatment options in MS.
Collapse
Affiliation(s)
- Max Mimpen
- School for Mental Health and Neuroscience, University of Maastricht, Maastricht The Netherlands
| | - Joost Smolders
- Department of Neurology, Erasmus University Medical Center, Rotterdam The Netherlands; Department of Neuroimmunology, Netherlands Institute for Neuroscience, Amsterdam The Netherlands
| | - Raymond Hupperts
- School for Mental Health and Neuroscience, University of Maastricht, Maastricht The Netherlands; Department of Neurology, Zuyderland Medical Center, Sittard The Netherlands
| | - Jan Damoiseaux
- Central Diagnostic Laboratory, Maastricht University Medical Center, Maastricht The Netherlands.
| |
Collapse
|
33
|
Palte MJ, Wehr A, Tawa M, Perkin K, Leigh-Pemberton R, Hanna J, Miller C, Penner N. Improving the Gastrointestinal Tolerability of Fumaric Acid Esters: Early Findings on Gastrointestinal Events with Diroximel Fumarate in Patients with Relapsing-Remitting Multiple Sclerosis from the Phase 3, Open-Label EVOLVE-MS-1 Study. Adv Ther 2019; 36:3154-3165. [PMID: 31538304 PMCID: PMC6822793 DOI: 10.1007/s12325-019-01085-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Diroximel fumarate (DRF) is a novel oral fumarate in development for patients with relapsing forms of multiple sclerosis (MS). Clinical findings from the DRF development program suggest that rates of gastrointestinal (GI) treatment-emergent adverse events (TEAEs) and discontinuation due to GI TEAEs are low, based on clinical and real-world observations of other fumaric acid esters, including dimethyl fumarate (DMF). The incidence of GI TEAEs varies from 40 to 88% in clinical and real-world studies of DMF. The objective of this study is to present GI tolerability findings from the EVOLVE-MS-1 study and present biologic hypotheses for the improved GI properties of DRF. METHODS GI TEAEs and treatment discontinuation because of GI TEAEs were assessed in DRF-treated patients with relapsing-remitting MS who were participating in the ongoing, 96-week, open-label, phase 3 EVOLVE-MS-1 study. RESULTS As of March 30, 2018, a total of 696 patients were enrolled in EVOLVE-MS-1. GI TEAEs were reported in 30.9% (215/696) of patients; the vast majority (96%; 207/215) experienced events that were mild or moderate in severity. When GI AEs did occur, they occurred early in treatment, resolved (88.8%; 191/215), and were of short duration [median 7.5 (range 1-87) days] in most patients. GI TEAEs led to < 1% of patients discontinuing treatment. CONCLUSIONS We suggest that the distinct chemical structure of DRF contributes to the observed low rates of GI TEAEs and GI-associated treatment discontinuations, possibly due to a combination of several factors. We hypothesize that these factors may include less reactivity with off-target proteins and/or lower production of a methanol leaving group that may contribute to GI irritation. A direct comparison of GI tolerability with DRF versus DMF is being evaluated in the EVOLVE-MS-2 study. TRIAL REGISTRATION ClinicalTrials.gov number NCT02634307. FUNDING Alkermes Inc. (Waltham, MA, USA) and Biogen (Cambridge, MA, USA).
Collapse
|