1
|
Raghib MF, Bernitsas E. From Animal Models to Clinical Trials: The Potential of Antimicrobials in Multiple Sclerosis Treatment. Biomedicines 2023; 11:3069. [PMID: 38002068 PMCID: PMC10668955 DOI: 10.3390/biomedicines11113069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 11/05/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic, autoimmune, demyelinating disease of the central nervous system (CNS). Microbes, including bacteria and certain viruses, particularly Epstein-Barr virus (EBV), have been linked to the pathogenesis of MS. While there is currently no cure for MS, antibiotics and antivirals have been studied as potential treatment options due to their immunomodulatory ability that results in the regulation of the immune process. The current issue addressed in this systematic review is the effect of antimicrobials, including antibiotics, antivirals, and antiparasitic agents in animals and humans. We performed a comprehensive search of PubMed, Google Scholar, and Scopus for articles on antimicrobials in experimental autoimmune encephalomyelitis animal models of MS, as well as in people with MS (pwMS). In animal models, antibiotics tested included beta-lactams, minocycline, rapamycin, macrolides, and doxycycline. Antivirals included acyclovir, valacyclovir, and ganciclovir. Hydroxychloroquine was the only antiparasitic that was tested. In pwMS, we identified a total of 24 studies, 17 of them relevant to antibiotics, 6 to antivirals, and 1 relevant to antiparasitic hydroxychloroquine. While the effect of antimicrobials in animal models was promising, only minocycline and hydroxychloroquine improved outcome measures in pwMS. No favorable effect of the antivirals in humans has been observed yet. The number and size of clinical trials testing antimicrobials have been limited. Large, multicenter, well-designed studies are needed to further evaluate the effect of antimicrobials in MS.
Collapse
Affiliation(s)
- Muhammad Faraz Raghib
- Department of Neurology, Wayne State University School of Medicine, Detroit, MI 48201, USA;
| | - Evanthia Bernitsas
- Department of Neurology, Wayne State University School of Medicine, Detroit, MI 48201, USA;
- Sastry Neuroimaging Laboratory, Department of Neurology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
2
|
Li X, Hu B, Guan X, Wang Z, Zhou Y, Sun H, Zhang X, Li Y, Huang X, Zhao Y, Wang X, Xu H, Zhang YW, Wang Z, Zheng H. Minocycline protects against microgliopathy in a Csf1r haplo-insufficient mouse model of adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP). J Neuroinflammation 2023; 20:134. [PMID: 37259140 DOI: 10.1186/s12974-023-02774-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 04/05/2023] [Indexed: 06/02/2023] Open
Abstract
BACKGROUND Mutations in colony-stimulating factor 1 receptor (CSF1R) are known to cause adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP), which has been recently demonstrated as a primary microgliopathy characterized by cognitive impairment. Although the molecular mechanism underlying CSF1R-mediated microgliopathy remains unclear, therapeutic strategies have generally targeted modulation of microglial function. In particular, the microglial inhibitor, minocycline, has been shown to attenuate learning and memory deficits in several neurodegenerative diseases. The objectives of this study were to investigate the pathogenic mechanisms underlying ALSP and to explore the therapeutic effects of minocycline in an in vivo model of ALSP. We hypothesized that inhibiting microglial activation via minocycline could reverse the behavior and pathological defects in ALSP model mice. METHODS We generated a Csf1r haploinsufficiency mouse model of ALSP using CRISPR/Cas9 genome editing and conducted electrophysiological recordings of long-term potentiation (LTP) and behavioral tests to validate the recapitulation of clinical ALSP characteristics in 8- to 11-month-old mice. RNA-sequencing was used to explore enriched gene expression in the molecular pathogenesis of ALSP. Microglial activation was assessed by immunofluorescent detection of Iba1 and CD68 in brain sections of male ALSP mice and pro-inflammatory activation and phagocytosis were assessed in Csf1r+/- microglia. Therapeutic effects were assessed by behavioral tests, histological analysis, and morphological examination after four weeks of intraperitoneal injection with minocycline or vehicle control in Csf1r+/- mice and wild-type control littermates. RESULTS We found that synaptic function was reduced in LTP recordings of neurons in the hippocampal CA1 region, while behavioral tests showed impaired spatial and cognitive memory specifically in male Csf1r+/- mice. Increased activation, pro-inflammatory cytokine production, and enhanced phagocytic capacity were also observed in Csf1r+/- microglia. Treatment with minocycline could suppress the activation of Csf1r+/- microglia both in vitro and in vivo. Notably, the behavioral and pathological deficits in Csf1r+/- mice were partially rescued by minocycline administration, potentially due to inhibition of microglial inflammation and phagocytosis in Csf1r+/- mice. CONCLUSIONS Our study shows that CSF1R deficiency results in aberrant microglial activation, characterized by a pro-inflammatory phenotype and enhanced phagocytosis of myelin. Our results also indicate that microglial inhibition by minocycline can ameliorate behavioral impairment and ALSP pathogenesis in CSF1R-deficient male mice, suggesting a potential therapeutic target for CSF1R-related leukoencephalopathy. Collectively, these data support that minocycline confers protective effects against CSF1R-related microgliopathy in male ALSP model mice.
Collapse
Affiliation(s)
- Xin Li
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Banglian Hu
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Xiaoyan Guan
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Ziwei Wang
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Yuhang Zhou
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Hao Sun
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Xian Zhang
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Yanfang Li
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Xiaohua Huang
- Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Yingjun Zhao
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Xin Wang
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
- State Key Laboratory of Cellular Stress Biology, Xiamen University, Xiamen, 361102, China
| | - Huaxi Xu
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Yun-Wu Zhang
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China
| | - Zhanxiang Wang
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China.
- Department of Neurosurgery, Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, Xiamen, 361102, China.
| | - Honghua Zheng
- Xiamen Key Laboratory of Brain Center, The First Affiliated Hospital of Xiamen University, Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China.
- Basic Medical Sciences, School of Medicine, Xiamen University, Xiamen, 361102, Fujian, China.
| |
Collapse
|
3
|
The potential use of tetracyclines in neurodegenerative diseases and the role of nano-based drug delivery systems. Eur J Pharm Sci 2022; 175:106237. [PMID: 35710076 DOI: 10.1016/j.ejps.2022.106237] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 05/07/2022] [Accepted: 06/12/2022] [Indexed: 11/21/2022]
Abstract
Neurodegenerative diseases are still a challenge for effective treatments. The high cost of approved drugs, severity of side effects, injection site pain, and restrictions on drug delivery to the Central Nervous System (CNS) can overshadow the management of these diseases. Due to the chronic and progressive evolution of neurodegenerative disorders and since there is still no cure for them, new therapeutic strategies such as the combination of several drugs or the use of existing drugs with new therapeutic applications are valuable strategies. Tetracyclines are traditionally classified as antibiotics. However, in this class of drugs, doxycycline and minocycline exhibit also anti-inflammatory effects by inhibiting microglia/macrophages. Hence, they have been studied as potential agents for the treatment of neurodegenerative diseases. The results of in vitro and in vivo studies confirm the effective role of these two drugs as anti-inflammatory agents in experimentally induced models of neurodegenerative diseases. In clinical studies, satisfactory results have been obtained in Multiple sclerosis (MS) but not yet in other disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), or Amyotrophic lateral sclerosis (ALS). In recent years, researchers have developed and evaluated nanoparticulate drug delivery systems to improve the clinical efficacy of these two tetracyclines for their potential application in neurodegenerative diseases. This study reviews the neuroprotective roles of minocycline and doxycycline in four of the main neurodegenerative disorders: AD, PD, ALS and MS. Moreover, the potential applications of nanoparticulate delivery systems developed for both tetracyclines are also reviewed.
Collapse
|
4
|
Therapeutic Potential of Combined Therapy of Vitamin A and Vitamin C in the Experimental Autoimmune Encephalomyelitis (EAE) in Lewis Rats. Mol Neurobiol 2022; 59:2328-2347. [PMID: 35072933 DOI: 10.1007/s12035-022-02755-0] [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: 10/06/2021] [Accepted: 01/17/2022] [Indexed: 10/19/2022]
Abstract
Demyelination, inflammation, oxidative injury, and glial activation are the main pathological hallmarks of multiple sclerosis (MS). Vitamins, as essential micronutrients, seem to be crucial in the pathogenesis of MS, and particularly vitamins A and C were found to have a protective role in MS development or progression. In this study, the therapeutic potential of combined therapy of vitamins A and C on progression of experimental autoimmune encephalomyelitis (EAE) and myelin repair mechanisms was examined. EAE, an animal model of MS, was induced in female Lewis rats. The rats were treated with daily intraperitoneal injections of vitamins A and C and their combination. We found that co-supplementation of vitamins A and C mitigated neurological severity and EAE disease progression. Histological study confirmed a significant reduction in demyelination size, inflammation and immune cell infiltration as well as microglia and astrocyte activation following co-administration of vitamins A and C. Co-administration of vitamins A and C also decreased the levels of pro-inflammatory cytokines (TNF-α, IL1β) and iNOS and increased gene expressions of IL-10, Nrf-2, HO-1, and MBP. Combination therapy of vitamins A and C also increased the total antioxidant capacity and decreased levels of oxidative stress markers. Finally, we proved that co-administration of vitamins A and C has anti-apoptotic and neuroprotective impacts in EAE via decreasing caspase-3 and increasing BDNF and NeuN expressing cells. The present study suggests that combined therapy of vitamins A and C may be an effective strategy for development of alternative medicine in boosting myelin repair in demyelinating diseases.
Collapse
|
5
|
Florou DT, Mavropoulos A, Dardiotis E, Tsimourtou V, Siokas V, Aloizou AM, Liaskos C, Tsigalou C, Katsiari C, Sakkas LI, Hadjigeorgiou G, Bogdanos DP. Tetracyclines Diminish In Vitro IFN-γ and IL-17-Producing Adaptive and Innate Immune Cells in Multiple Sclerosis. Front Immunol 2021; 12:739186. [PMID: 34899697 PMCID: PMC8662812 DOI: 10.3389/fimmu.2021.739186] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 09/16/2021] [Indexed: 11/28/2022] Open
Abstract
Introduction Limited data from clinical trials in multiple sclerosis (MS) reported that minocycline, a widely used antibiotic belonging to the family of tetracyclines (TCs), exerts a beneficial short-lived clinical effect A similar anti-inflammatory effect of minocycline attributed to a deviation from Th1 to Th2 immune response has been reported in experimental models of MS. Whether such an immunomodulatory mechanism is operated in the human disease remains largely unknown. Aim To assess the in vitro immunomodulatory effect of tetracyclines, and in particular minocycline and doxycycline, in naïve and treated patients with MS. Material and Methods Peripheral blood mononuclear cells from 45 individuals (35 MS patients, amongst which 15 naïve patients and 10 healthy controls, HCs) were cultured with minocycline or doxycycline and conventional stimulants (PMA/Ionomycin or IL-12/IL-18). IFN-γ and IL-17 producing T-, NK- and NKT cells were assessed by flow cytometry. The effect of TCs on cell viability and apoptosis was further assessed by flow cytometry with Annexin V staining. Results Both tetracyclines significantly decreased, in a dose dependent manner, IFN-γ production in NKT and CD4+ T lymphocytes from MS patients (naïve or treated) stimulated with IL-12/IL-18 but did not decrease IFN-γ producing CD8+ T cells from naive MS or treated RRMS patients. They also decreased IL-17+ T and NKT cells following PMA and Ionomycin-stimulation. Tetracyclines did not affect the viability of cell subsets. Conclusion Tetracyclines can in vitro suppress IFN-γ and IL-17- producing cells from MS patients, and this may explain their potential therapeutic effect in vivo.
Collapse
Affiliation(s)
- Despoina T Florou
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece.,Department of Neurology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Athanasios Mavropoulos
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Efthymios Dardiotis
- Department of Neurology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Vana Tsimourtou
- Department of Neurology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Vasileios Siokas
- Department of Neurology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Athina-Maria Aloizou
- Department of Neurology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Christos Liaskos
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Christina Tsigalou
- Laboratory of Microbiology, Faculty of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Christina Katsiari
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Lazaros I Sakkas
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | - Georgios Hadjigeorgiou
- Department of Neurology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece.,Medical School, University of Cyprus, Nicosia, Cyprus
| | - Dimitrios P Bogdanos
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| |
Collapse
|
6
|
Nasrnezhad R, Halalkhor S, Sadeghi F, Pourabdolhossein F. Piperine Improves Experimental Autoimmune Encephalomyelitis (EAE) in Lewis Rats Through its Neuroprotective, Anti-inflammatory, and Antioxidant Effects. Mol Neurobiol 2021; 58:5473-5493. [PMID: 34338970 DOI: 10.1007/s12035-021-02497-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/14/2021] [Indexed: 12/12/2022]
Abstract
Inflammation, demyelination, glial activation, and oxidative damage are the most pathological hallmarks of multiple sclerosis (MS). Piperine, a main bioactive alkaloid of black pepper, possesses antioxidant, anti-inflammatory, and neuroprotective properties whose therapeutic potential has been less studied in the experimental autoimmune encephalomyelitis (EAE) models. In this study, the efficiency of piperine on progression of EAE model and myelin repair mechanisms was investigated. EAE was induced in female Lewis rats and piperine and its vehicle were daily administrated intraperitoneally from day 8 to 29 post immunization. We found that piperine alleviated neurological deficits and EAE disease progression. Luxol fast blue and H&E staining and immunostaining of lumbar spinal cord cross sections confirmed that piperine significantly reduced the extent of demyelination, inflammation, immune cell infiltration, microglia, and astrocyte activation. Gene expression analysis in lumbar spinal cord showed that piperine treatment decreased the level of pro-inflammatory cytokines (TNF-α, IL-1β) and iNOS and enhanced IL-10, Nrf2, HO-1, and MBP expressions. Piperine supplementation also enhanced the total antioxidant capacity (FRAP) and reduced the level of oxidative stress marker (MDA) in the CNS of EAE rats. Finally, we found that piperine has anti-apoptotic and neuroprotective effect in EAE through reducing caspase-3 (apoptosis marker) and enhancing BDNF and NeuN expressing cells. This study strongly indicates that piperine has a beneficial effect on the EAE progression and could be considered as a potential therapeutic target for MS treatment. Upcoming clinical trials will provide a deeper understanding of piperine's role for the treatment of the MS.
Collapse
Affiliation(s)
- Reza Nasrnezhad
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran.,Department of Biochemistry, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Sohrab Halalkhor
- Department of Biochemistry, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Farzin Sadeghi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Fereshteh Pourabdolhossein
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran. .,Department of Physiology, School of Medicine, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
7
|
Molecular and Biochemical Basis of Minocycline-Induced Hyperpigmentation-The Study on Normal Human Melanocytes Exposed to UVA and UVB Radiation. Int J Mol Sci 2021; 22:ijms22073755. [PMID: 33916535 PMCID: PMC8038496 DOI: 10.3390/ijms22073755] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/01/2021] [Accepted: 04/01/2021] [Indexed: 01/04/2023] Open
Abstract
Minocycline is a drug which induces skin hyperpigmentation. Its frequency reaches up to 50% of treated patients. The adverse effect diminishes the great therapeutic potential of minocycline, including antibacterial, neuroprotective, anti-inflammatory and anti-cancer actions. It is supposed that an elevated melanin level and drug accumulation in melanin-containing cells are related to skin hyperpigmentation. This study aimed to evaluate molecular and biochemical mechanism of minocycline-induced hyperpigmentation in human normal melanocytes, as well as the contribution of UV radiation to this side effect. The experiments involved the evaluation of cyto- and phototoxic potential of the drug using cell imaging with light and confocal microscopes as well as biochemical and molecular analysis of melanogenesis. We showed that minocycline induced melanin synthesis in epidermal melanocytes. The action was intensified by UV irradiation, especially with the UVB spectrum. Minocycline stimulated the expression of microphthalmia-associated transcription factor (MITF) and tyrosinase (TYR) gene. Higher levels of melanin and increased activity of tyrosinase were also observed in treated cells. Moreover, minocycline triggered the supranuclear accumulation of tyrosinase, similar to UV radiation. The decreased level of premelanosome protein PMEL17 observed in all minocycline-treated cultures suggests disorder of the formation, maturation or distribution of melanosomes. The study revealed that minocycline itself was able to enhance melanin synthesis. The action was intensified by irradiation, especially with the UVB spectrum. Demonstrated results confirmed the potential role of melanin and UV radiation minocycline-induced skin hyperpigmentation.
Collapse
|
8
|
Microglial Pruning: Relevance for Synaptic Dysfunction in Multiple Sclerosis and Related Experimental Models. Cells 2021; 10:cells10030686. [PMID: 33804596 PMCID: PMC8003660 DOI: 10.3390/cells10030686] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/12/2021] [Accepted: 03/17/2021] [Indexed: 12/11/2022] Open
Abstract
Microglia, besides being able to react rapidly to a wide range of environmental changes, are also involved in shaping neuronal wiring. Indeed, they actively participate in the modulation of neuronal function by regulating the elimination (or “pruning”) of weaker synapses in both physiologic and pathologic processes. Mounting evidence supports their crucial role in early synaptic loss, which is emerging as a hallmark of several neurodegenerative diseases, including multiple sclerosis (MS) and its preclinical models. MS is an inflammatory, immune-mediated pathology of the white matter in which demyelinating lesions may cause secondary neuronal death. Nevertheless, primitive grey matter (GM) damage is emerging as an important contributor to patients’ long-term disability, since it has been associated with early and progressive cognitive decline (CD), which seriously worsens the quality of life of MS patients. Widespread synapse loss even in the absence of demyelination, axon degeneration and neuronal death has been demonstrated in different GM structures, thus raising the possibility that synaptic dysfunction could be an early and possibly independent event in the neurodegenerative process associated with MS. This review provides an overview of microglial-dependent synapse elimination in the neuroinflammatory process that underlies MS and its experimental models.
Collapse
|
9
|
Al Mamun A, Monalisa I, Tul Kubra K, Akter A, Akter J, Sarker T, Munir F, Wu Y, Jia C, Afrin Taniya M, Xiao J. Advances in immunotherapy for the treatment of spinal cord injury. Immunobiology 2020; 226:152033. [PMID: 33321368 DOI: 10.1016/j.imbio.2020.152033] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 10/19/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022]
Abstract
Spinal cord injury (SCI) is a leading cause of morbidity and disability in the world. Over the past few decades, the exact molecular mechanisms describing secondary, persistent injuries, as well as primary and transient injuries, have attracted massive attention to the clinicians and researchers. Recent investigations have distinctly shown the critical roles of innate and adaptive immune responses in regulating sterile neuroinflammation and functional outcomes after SCI. In past years, some promising advances in immunotherapeutic options have efficaciously been identified for the treatment of SCI. In our narrative review, we have mainly focused on the new therapeutic strategies such as the maturation and apoptosis of immune cells by several agents, mesenchymal stem cells (MSCs) as well as multi-factor combination therapy, which have recently provided novel ideas and prospects for the future treatment of SCI. This article also illustrates the latest progress in clarifying the potential roles of innate and adaptive immune responses in SCI, the progression and specification of prospective immunotherapy and outstanding issues in the area.
Collapse
Affiliation(s)
- Abdullah Al Mamun
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035 Zhejiang Province, China
| | - Ilma Monalisa
- Department of Pharmacy, Southeast University, Banani, Dhaka 1213, Bangladesh
| | - Khadija Tul Kubra
- Department of Pharmacy, University of Development Alternative, Dhaka 1209, Bangladesh
| | - Afroza Akter
- Department of Microbiology, Noakhali Science and Technology University, Noakhali 3814, Bangladesh
| | - Jaheda Akter
- Department of Pharmacy, International Islamic University Chittagong, Kumira, Chattogram-4318, Chittagong, Bangladesh
| | - Tamanna Sarker
- Department of Pharmacy, University of Asia Pacific, Dhaka 1205, Bangladesh
| | - Fahad Munir
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000 Zhejiang Province, China
| | - Yanqing Wu
- Institute of Life Sciences, Wenzhou University, Wenzhou, 325035 Zhejiang Province, China
| | - Chang Jia
- Pediatric Research Institute, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027 Zhejiang Province, China
| | - Masuma Afrin Taniya
- Department of Life Sciences, School of Environment and Life Sciences, Independent University, Bangladesh, Dhaka 1229, Bangladesh
| | - Jian Xiao
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035 Zhejiang Province, China.
| |
Collapse
|
10
|
Chauhan P, Kakkar AK, Singh H, Gautam CS. Minocycline for the management of multiple sclerosis: repositioning potential, opportunities, and challenges. Expert Rev Neurother 2020; 21:35-43. [PMID: 33059513 DOI: 10.1080/14737175.2020.1838276] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Multiple sclerosis (MS) is a chronic demyelinating inflammatory disorder with variable clinical and pathologic characteristics reflecting multiple underlying pathophysiologic mechanisms. Repositioning of existing drugs for the new indications offers several advantages including significant reduction in the cost and time of drug development and exemption from early phase clinical trials. Minocycline has been reported to exhibit immunomodulation in several pre-clinical and clinical studies through suppression of migratory inflammatory cells, modulation of peripheral immune response, and inhibition of microglial activation within the CNS. AREAS COVERED Here, the authors review the repositioning potential of minocycline for the treatment of MS along with appraisal of the evidence obtained from preclinical and clinical research. The authors also discuss the advantages and potential safety concerns related to the use of minocycline for the management of MS. EXPERT OPINION Minocycline offers several distinct advantages in terms of well-known safety profile, lower cost of therapy, widespread availability, and being available as an oral formulation. The authors call upon the public and private funders to facilitate well designed and adequately powered randomized clinical trials that can provide conclusive evidence regarding the safety and efficacy of minocycline in patients with MS.
Collapse
Affiliation(s)
- Prerna Chauhan
- Department of Pharmacology, All India Institute of Medical Sciences , New Delhi, India
| | - Ashish Kumar Kakkar
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research , Chandigarh, India
| | - Harmanjit Singh
- Department of Pharmacology, Government Medical College and Hospital , Chandigarh, India
| | - C S Gautam
- Department of Pharmacology, Government Medical College and Hospital , Chandigarh, India
| |
Collapse
|
11
|
Ochoa-Repáraz J, Ramelow CC, Kasper LH. A Gut Feeling: The Importance of the Intestinal Microbiota in Psychiatric Disorders. Front Immunol 2020; 11:510113. [PMID: 33193297 PMCID: PMC7604426 DOI: 10.3389/fimmu.2020.510113] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 09/30/2020] [Indexed: 12/11/2022] Open
Abstract
The intestinal microbiota constitutes a complex ecosystem in constant reciprocal interactions with the immune, neuroendocrine, and neural systems of the host. Recent molecular technological advances allow for the exploration of this living organ and better facilitates our understanding of the biological importance of intestinal microbes in health and disease. Clinical and experimental studies demonstrate that intestinal microbes may be intimately involved in the progression of diseases of the central nervous system (CNS), including those of affective and psychiatric nature. Gut microbes regulate neuroinflammatory processes, play a role in balancing the concentrations of neurotransmitters and could provide beneficial effects against neurodegeneration. In this review, we explore some of these reciprocal interactions between gut microbes and the CNS during experimental disease and suggest that therapeutic approaches impacting the gut-brain axis may represent the next avenue for the treatment of psychiatric disorders.
Collapse
Affiliation(s)
| | | | - Lloyd H. Kasper
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth College, Hanover, NH, United States
| |
Collapse
|
12
|
Gautam SS, Gautam CS, Garg VK, Singh H. Combining hydroxychloroquine and minocycline: potential role in moderate to severe COVID-19 infection. Expert Rev Clin Pharmacol 2020; 13:1183-1190. [PMID: 33008280 DOI: 10.1080/17512433.2020.1832889] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Patients with moderate to severe COVID-19 infection require specific drugs to prevent the morbidity and mortality. Hydroxychloroquine (HCQ) has shown some promise in the management of COVID 19. Minocycline, because of its anticytokine and other useful properties can be an ideal candidate for combining with HCQ. AREAS COVERED Here we review the need and mechanisms and reasons for combining HCQ and minocycline moderate to severe COVID-19 infection. We also reviewed the advantages, potential safety concerns and precautions to be taken, while combining HCQ and minocycline. EXPERT OPINION Combining HCQ and minocycline offers many advantages in the management of moderate to severe COVID-19 infection. Both drugs are cheaper, widely available and long-term safety data and contraindications are well known. We do not recommend this combination for prophylaxis or use in asymptomatic or mild disease patients as this can lead to unnecessary safety concerns. Additive antimicrobial and anticytokine effects of both drugs may reduce the morbidity and mortality among patients with COVID-19 and may act as a cheaper alternative to the costlier drugs, however, thorough clinical research is warranted. We call upon public and private healthcare bodies to come up with large well-designed clinical studies for generating evidence-based recommendations.
Collapse
Affiliation(s)
| | - C S Gautam
- Department of Pharmacology, Government Medical College and Hospital , Chandigarh, India
| | - Vivek Kumar Garg
- Department of Biochemistry, Government Medical College and Hospital , Chandigarh, India
| | - Harmanjit Singh
- Department of Pharmacology, Government Medical College and Hospital , Chandigarh, India
| |
Collapse
|
13
|
Fresegna D, Bullitta S, Musella A, Rizzo FR, De Vito F, Guadalupi L, Caioli S, Balletta S, Sanna K, Dolcetti E, Vanni V, Bruno A, Buttari F, Stampanoni Bassi M, Mandolesi G, Centonze D, Gentile A. Re-Examining the Role of TNF in MS Pathogenesis and Therapy. Cells 2020; 9:cells9102290. [PMID: 33066433 PMCID: PMC7602209 DOI: 10.3390/cells9102290] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/10/2020] [Accepted: 10/12/2020] [Indexed: 12/11/2022] Open
Abstract
Multiple sclerosis (MS) is a common neurological disorder of putative autoimmune origin. Clinical and experimental studies delineate abnormal expression of specific cytokines over the course of the disease. One major cytokine that has been shown to play a pivotal role in MS is tumor necrosis factor (TNF). TNF is a pleiotropic cytokine regulating many physiological and pathological functions of both the immune system and the central nervous system (CNS). Convincing evidence from studies in human and experimental MS have demonstrated the involvement of TNF in various pathological hallmarks of MS, including immune dysregulation, demyelination, synaptopathy and neuroinflammation. However, due to the complexity of TNF signaling, which includes two-ligands (soluble and transmembrane TNF) and two receptors, namely TNF receptor type-1 (TNFR1) and type-2 (TNFR2), and due to its cell- and context-differential expression, targeting the TNF system in MS is an ongoing challenge. This review summarizes the evidence on the pathophysiological role of TNF in MS and in different MS animal models, with a special focus on pharmacological treatment aimed at controlling the dysregulated TNF signaling in this neurological disorder.
Collapse
Affiliation(s)
- Diego Fresegna
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, 00166 Rome, Italy; (D.F.); (S.B.); (A.M.); (L.G.); (V.V.); (G.M.); (A.G.)
| | - Silvia Bullitta
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, 00166 Rome, Italy; (D.F.); (S.B.); (A.M.); (L.G.); (V.V.); (G.M.); (A.G.)
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (F.R.R.); (S.B.); (K.S.); (E.D.); (A.B.)
| | - Alessandra Musella
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, 00166 Rome, Italy; (D.F.); (S.B.); (A.M.); (L.G.); (V.V.); (G.M.); (A.G.)
- Department of Human Sciences and Quality of Life Promotion, University of Rome San Raffaele, 00166 Roma, Italy
| | - Francesca Romana Rizzo
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (F.R.R.); (S.B.); (K.S.); (E.D.); (A.B.)
| | - Francesca De Vito
- Unit of Neurology, IRCCS Neuromed, Pozzilli (Is), 86077 Pozzilli, Italy; (F.D.V.); (S.C.); (F.B.); (M.S.B.)
| | - Livia Guadalupi
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, 00166 Rome, Italy; (D.F.); (S.B.); (A.M.); (L.G.); (V.V.); (G.M.); (A.G.)
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (F.R.R.); (S.B.); (K.S.); (E.D.); (A.B.)
| | - Silvia Caioli
- Unit of Neurology, IRCCS Neuromed, Pozzilli (Is), 86077 Pozzilli, Italy; (F.D.V.); (S.C.); (F.B.); (M.S.B.)
| | - Sara Balletta
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (F.R.R.); (S.B.); (K.S.); (E.D.); (A.B.)
| | - Krizia Sanna
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (F.R.R.); (S.B.); (K.S.); (E.D.); (A.B.)
| | - Ettore Dolcetti
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (F.R.R.); (S.B.); (K.S.); (E.D.); (A.B.)
| | - Valentina Vanni
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, 00166 Rome, Italy; (D.F.); (S.B.); (A.M.); (L.G.); (V.V.); (G.M.); (A.G.)
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (F.R.R.); (S.B.); (K.S.); (E.D.); (A.B.)
| | - Antonio Bruno
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (F.R.R.); (S.B.); (K.S.); (E.D.); (A.B.)
| | - Fabio Buttari
- Unit of Neurology, IRCCS Neuromed, Pozzilli (Is), 86077 Pozzilli, Italy; (F.D.V.); (S.C.); (F.B.); (M.S.B.)
| | - Mario Stampanoni Bassi
- Unit of Neurology, IRCCS Neuromed, Pozzilli (Is), 86077 Pozzilli, Italy; (F.D.V.); (S.C.); (F.B.); (M.S.B.)
| | - Georgia Mandolesi
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, 00166 Rome, Italy; (D.F.); (S.B.); (A.M.); (L.G.); (V.V.); (G.M.); (A.G.)
- Department of Human Sciences and Quality of Life Promotion, University of Rome San Raffaele, 00166 Roma, Italy
| | - Diego Centonze
- Synaptic Immunopathology Lab, Department of Systems Medicine, Tor Vergata University, 00133 Rome, Italy; (F.R.R.); (S.B.); (K.S.); (E.D.); (A.B.)
- Unit of Neurology, IRCCS Neuromed, Pozzilli (Is), 86077 Pozzilli, Italy; (F.D.V.); (S.C.); (F.B.); (M.S.B.)
- Correspondence: ; Tel.: +39-06-7259-6010; Fax: +39-06-7259-6006
| | - Antonietta Gentile
- Synaptic Immunopathology Lab, IRCCS San Raffaele Pisana, 00166 Rome, Italy; (D.F.); (S.B.); (A.M.); (L.G.); (V.V.); (G.M.); (A.G.)
| |
Collapse
|
14
|
Rok J, Rzepka Z, Beberok A, Pawlik J, Wrześniok D. Cellular and Molecular Aspects of Anti-Melanoma Effect of Minocycline-A Study of Cytotoxicity and Apoptosis on Human Melanotic Melanoma Cells. Int J Mol Sci 2020; 21:E6917. [PMID: 32967177 PMCID: PMC7555712 DOI: 10.3390/ijms21186917] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 09/09/2020] [Accepted: 09/18/2020] [Indexed: 01/01/2023] Open
Abstract
Minocycline is a tetracycline compound with pleiotropic pharmacological properties. In addition to its antibacterial action, it shows many non-antimicrobial effects, including an anti-cancer activity. The anti-cancer action was confirmed in studies on ovarian carcinoma cells, hepatocellular carcinoma cells, glioma cells, or acute myeloid leukemia cells. Malignant melanoma remains a serious medical problem despite the extensive knowledge of the disease. The low effectiveness of the standard treatment, as well as the resistance to therapy, result in high mortality rates. This work aimed to investigate the potential and mechanisms of anti-melanoma action of minocycline. Human skin melanotic melanoma cell line COLO 829 was used in the study. The obtained results showed that minocycline decreased cell viability and inhibited the growth of melanoma cells, proportional to the drug concentration as well as to the time of incubation. The EC50 values were calculated to be 78.6 µM, 31.7 µM, and 13.9 µM for 24 h, 48 h, and 72 h, respectively. It was observed that treated cells had a disturbed cell cycle and significantly changed morphology. Moreover, minocycline caused a decrease in mitochondrial membrane potential and an increase in cells with a low level of reduced thiols. Finally, it was found that the anti-melanoma effect of minocycline was related to the induction of apoptosis. The drug activated caspases 8, 9, and 3/7 as well as increased the number of annexin V-positive cells. The presented results show that minocycline possesses anti-melanoma potential.
Collapse
Affiliation(s)
- Jakub Rok
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Katowice, Jagiellońska 4, 41-200 Sosnowiec, Poland; (Z.R.); (A.B.); (J.P.); (D.W.)
| | | | | | | | | |
Collapse
|
15
|
Mallah K, Couch C, Borucki DM, Toutonji A, Alshareef M, Tomlinson S. Anti-inflammatory and Neuroprotective Agents in Clinical Trials for CNS Disease and Injury: Where Do We Go From Here? Front Immunol 2020; 11:2021. [PMID: 33013859 PMCID: PMC7513624 DOI: 10.3389/fimmu.2020.02021] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/27/2020] [Indexed: 02/06/2023] Open
Abstract
Neurological disorders are major contributors to death and disability worldwide. The pathology of injuries and disease processes includes a cascade of events that often involve molecular and cellular components of the immune system and their interaction with cells and structures within the central nervous system. Because of this, there has been great interest in developing neuroprotective therapeutic approaches that target neuroinflammatory pathways. Several neuroprotective anti-inflammatory agents have been investigated in clinical trials for a variety of neurological diseases and injuries, but to date the results from the great majority of these trials has been disappointing. There nevertheless remains great interest in the development of neuroprotective strategies in this arena. With this in mind, the complement system is being increasingly discussed as an attractive therapeutic target for treating brain injury and neurodegenerative conditions, due to emerging data supporting a pivotal role for complement in promoting multiple downstream activities that promote neuroinflammation and degeneration. As we move forward in testing additional neuroprotective and immune-modulating agents, we believe it will be useful to review past trials and discuss potential factors that may have contributed to failure, which will assist with future agent selection and trial design, including for complement inhibitors. In this context, we also discuss inhibition of the complement system as a potential neuroprotective strategy for neuropathologies of the central nervous system.
Collapse
Affiliation(s)
- Khalil Mallah
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Christine Couch
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Department of Health Sciences and Research, College of Health Professions, Medical University of South Carolina, Charleston, SC, United States
| | - Davis M. Borucki
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, United States
- Medical Scientist Training Program, Medical University of South Carolina, Charleston, SC, United States
| | - Amer Toutonji
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, United States
- Medical Scientist Training Program, Medical University of South Carolina, Charleston, SC, United States
| | - Mohammed Alshareef
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Department of Neurological Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Ralph Johnson VA Medical Center, Charleston, SC, United States
| |
Collapse
|
16
|
Boguszewska-Czubara A, Budzynska B, Skalicka-Wozniak K, Kurzepa J. Perspectives and New Aspects of Metalloproteinases' Inhibitors in the Therapy of CNS Disorders: From Chemistry to Medicine. Curr Med Chem 2019; 26:3208-3224. [PMID: 29756562 DOI: 10.2174/0929867325666180514111500] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 03/31/2017] [Accepted: 04/05/2018] [Indexed: 11/22/2022]
Abstract
Matrix metalloproteinases (MMPs) play a key role in remodeling of the extracellular matrix (ECM) and, at the same time, influence cell differentiation, migration, proliferation, and survival. Their importance in a variety of human diseases including cancer, rheumatoid arthritis, pulmonary emphysema and fibrotic disorders has been known for many years but special attention should be paid on the role of MMPs in the central nervous system (CNS) disorders. Till now, there are not many well documented physiological MMP target proteins in the brain but only some pathological ones. Numerous neurodegenerative diseases are a consequence of or result in disturbed remodeling of brain ECM, therefore proper action of MMPs as well as control of their activity may play crucial roles in the development of these diseases. In the present review, we discuss the role of metalloproteinase inhibitors, from the wellknown natural endogenous tissue inhibitors of metalloproteinases (TIMPs) to the exogenous synthetic ones like (4-phenoxyphenylsulfonyl)methylthiirane (SB-3CT), tetracyclines, batimastat (BB-94) and FN-439. As the MMP-TIMP system has been well described in physiological development as well as in pathological conditions mainly in neoplastic diseases, the knowledge about the enzymatic system in mammalian brain tissue still remains poorly understood in this context. Therefore, we focus on MMPs inhibition in the context of the physiological function of the adult brain as well as pathological conditions including neurodegenerative diseases, brain injuries, and others.
Collapse
Affiliation(s)
| | - Barbara Budzynska
- Department of Pharmacology and Pharmacodynamics, Medical University of Lublin, Lublin, Poland
| | - Krystyna Skalicka-Wozniak
- Department of Pharmacognosy with Medicinal Plants Unit, Medical University of Lublin, Lublin, Poland
| | - Jacek Kurzepa
- Department of Medical Chemistry, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
17
|
Lee J, Hamanaka G, Lo EH, Arai K. Heterogeneity of microglia and their differential roles in white matter pathology. CNS Neurosci Ther 2019; 25:1290-1298. [PMID: 31733036 PMCID: PMC6887901 DOI: 10.1111/cns.13266] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 10/29/2019] [Accepted: 10/30/2019] [Indexed: 12/12/2022] Open
Abstract
Microglia are resident immune cells that play multiple roles in central nervous system (CNS) development and disease. Although the classical concept of microglia/macrophage activation is based on a biphasic beneficial‐versus‐deleterious polarization, growing evidence now suggests a much more heterogenous profile of microglial activation that underlie their complex roles in the CNS. To date, the majority of data are focused on microglia in gray matter. However, demyelination is a prominent pathologic finding in a wide range of diseases including multiple sclerosis, Alzheimer's disease, and vascular cognitive impairment and dementia. In this mini‐review, we discuss newly discovered functional subsets of microglia that contribute to white matter response in CNS disease onset and progression. Microglia show different molecular patterns and morphologies depending on disease type and brain region, especially in white matter. Moreover, in later stages of disease, microglia demonstrate unconventional immuno‐regulatory activities such as increased phagocytosis of myelin debris and secretion of trophic factors that stimulate oligodendrocyte lineage cells to facilitate remyelination and disease resolution. Further investigations of these multiple microglia subsets may lead to novel therapeutic approaches to treat white matter pathology in CNS injury and disease.
Collapse
Affiliation(s)
- Janice Lee
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Gen Hamanaka
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Eng H Lo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
18
|
Albrecht J, Baine PA, Ladizinski B, Jemec GB, Bigby M. Long-term clinical safety of clindamycin and rifampicin combination for the treatment of hidradenitis suppurativa. A Critically Appraised Topic. Br J Dermatol 2019; 180:749-755. [PMID: 30281779 DOI: 10.1111/bjd.17265] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/26/2018] [Indexed: 12/15/2022]
Abstract
CLINICAL QUESTION/SCENARIO Can therapy with clindamycin and rifampicin be safely continued long term beyond the recommended 10-week course? BACKGROUND Clindamycin and rifampicin are used in combination to treat hidradenitis suppurativa (HS). There is no data on the efficacy and safety of clindamycin/rifampicin combination therapy for HS beyond 10 weeks. METHODS We identified the following major concerns that still lack a proper evidenced-based analysis: for rifampicin, drug-induced liver injury, interstitial nephritis, drug interaction and hepatic p450 3A4 enzyme induction; for clindamycin, the concern was community-acquired Clostridium difficile infection (CA-CDI); and experience with long-term treatment. Data sources were used as appropriate to answer the question. Systematic searches were used to assess the risk of CA-CDI and experience with long-term treatment with clindamycin. RESULTS/IDENTIFIED EVIDENCE The risk for rifampicin-induced liver injury is highest in the first 6 weeks of treatment, whereas interstitial nephritis is primarily observed during intermittent treatment. Enzyme induction due to rifampicin is usually complete after about 2 weeks of treatment and reduces clindamycin blood levels by about 90%. Three meta-analyses identified antibiotic use as a risk factor for CA-CDI. Two of them assigned the highest risk to clindamycin. None of them stratified by length of treatment. There is extensive experience with rifampicin, primarily for the treatment of tuberculosis. Long-term experience with clindamycin is limited. DISCUSSION AND RECOMMENDATION FOR CLINICAL CARE The analysed risks associated with a combination of clindamycin and rifampicin for hidradenitis suppurative cluster within the first 10 weeks. Treatment can be continued beyond 10 weeks, if clinically necessary.
Collapse
Affiliation(s)
- J Albrecht
- Division of Dermatology, Department of Medicine, J.H. Stroger Hospital of Cook County, 1900 West Polk Street, Chicago, IL, 60612, U.S.A.,Department of Dermatology, Rush Medical College, Chicago, IL, U.S.A
| | - P A Baine
- Countway Library of Medicine, Harvard Medical School, Boston, MA, U.S.A
| | - B Ladizinski
- Division of Dermatology, Department of Medicine, J.H. Stroger Hospital of Cook County, 1900 West Polk Street, Chicago, IL, 60612, U.S.A
| | - G B Jemec
- Department of Dermatology, Zealand University Hospital, Roskilde, Denmark.,Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - M Bigby
- Department of Dermatology, Harvard Medical School and Beth Israel Deaconess Medical Center, Boston, MA, U.S.A
| |
Collapse
|
19
|
Ramírez-Cheyne JA, Duque GA, Ayala-Zapata S, Saldarriaga-Gil W, Hagerman P, Hagerman R, Payán-Gómez C. Fragile X syndrome and connective tissue dysregulation. Clin Genet 2018; 95:262-267. [DOI: 10.1111/cge.13469] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 11/03/2018] [Indexed: 12/15/2022]
Affiliation(s)
| | | | | | | | - Paul Hagerman
- UC Davis MIND Institute, University of California; Davis California
| | - Randi Hagerman
- UC Davis MIND Institute, University of California; Davis California
| | - César Payán-Gómez
- Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario; Bogotá Colombia
| |
Collapse
|
20
|
De Angelis F, Plantone D, Chataway J. Pharmacotherapy in Secondary Progressive Multiple Sclerosis: An Overview. CNS Drugs 2018; 32:499-526. [PMID: 29968175 DOI: 10.1007/s40263-018-0538-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Multiple sclerosis is an immune-mediated inflammatory disease of the central nervous system characterised by demyelination, neuroaxonal loss and a heterogeneous clinical course. Multiple sclerosis presents with different phenotypes, most commonly a relapsing-remitting course and, less frequently, a progressive accumulation of disability from disease onset (primary progressive multiple sclerosis). The majority of people with relapsing-remitting multiple sclerosis, after a variable time, switch to a stage characterised by gradual neurological worsening known as secondary progressive multiple sclerosis. We have a limited understanding of the mechanisms underlying multiple sclerosis, and it is believed that multiple genetic, environmental and endogenous factors are elements driving inflammation and ultimately neurodegeneration. Axonal loss and grey matter damage have been regarded as amongst the leading causes of irreversible neurological disability in the progressive stages. There are over a dozen disease-modifying therapies currently licenced for relapsing-remitting multiple sclerosis, but none of these has provided evidence of effectiveness in secondary progressive multiple sclerosis. Recently, there has been some early modest success with siponimod in secondary progressive multiple sclerosis and ocrelizumab in primary progressive multiple sclerosis. Finding treatments to delay or prevent the courses of secondary progressive multiple sclerosis is an unmet and essential goal of the research in multiple sclerosis. In this review, we discuss new findings regarding drugs with immunomodulatory, neuroprotective or regenerative properties and possible treatment strategies for secondary progressive multiple sclerosis. We examine the field broadly to include trials where participants have progressive or relapsing phenotypes. We summarise the most relevant results from newer investigations from phase II and III randomised controlled trials over the past decade, with particular attention to the last 5 years.
Collapse
Affiliation(s)
- Floriana De Angelis
- Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, UCL Institute of Neurology, Faculty of Brain Sciences, UCL, London, UK.
| | - Domenico Plantone
- Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, UCL Institute of Neurology, Faculty of Brain Sciences, UCL, London, UK
| | - Jeremy Chataway
- Queen Square Multiple Sclerosis Centre, Department of Neuroinflammation, UCL Institute of Neurology, Faculty of Brain Sciences, UCL, London, UK
| |
Collapse
|
21
|
Abstract
Accumulating research substantiates the statement that inflammation plays an important role in the development of stroke. Both proinflammatory and anti-inflammatory mediators are involved in the pathogenesis of stroke, an imbalance of which leads to inflammation. Anti-inflammation is a kind of hopeful strategy for the prevention and treatment of stroke. Substantial studies have demonstrated that minocycline, a second-generation semisynthetic antibiotic belonging to the tetracycline family, can inhibit neuroinflammation, inflammatory mediators and microglia activation, and improve neurological outcome. Experimental and clinical data have found the preclinical and clinical potential of minocycline in the treatment of stroke due to its anti-inflammation properties and anti-inflammation-induced pathogeneses, including antioxidative stress, antiapoptosis, inhibiting leukocyte migration and microglial activation, and decreasing matrix metalloproteinases activity. Hence, it suggests a great future for minocycline in the therapeutics of stroke that diminish the inflammatory progress of stroke.
Collapse
|
22
|
Fu Y, Yan Y. Emerging Role of Immunity in Cerebral Small Vessel Disease. Front Immunol 2018; 9:67. [PMID: 29422904 PMCID: PMC5788893 DOI: 10.3389/fimmu.2018.00067] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 01/10/2018] [Indexed: 01/06/2023] Open
Abstract
Cerebral small vessel disease (CSVD) is one of the main causes of vascular dementia in older individuals. Apart from risk containment, efforts to prevent or treat CSVD are ineffective due to the unknown pathogenesis of the disease. CSVD, a subtype of stroke, is characterized by recurrent strokes and neurodegeneration. Blood-brain barrier (BBB) impairment, chronic inflammatory responses, and leukocyte infiltration are classical pathological features of CSVD. Understanding how BBB disruption instigates inflammatory and degenerative processes may be informative for CSVD therapy. Antigens derived from the brain are found in the peripheral blood of lacunar stroke patients, and antibodies and sensitized T cells against brain antigens are also detected in patients with leukoaraiosis. These findings suggest that antigen-specific immune responses could occur in CSVD. This review describes the neurovascular unit features of CSVD, the immune responses to specific neuronal and glial processes that may be involved in a distinct mechanism of CSVD, and the current evidence of the association between mechanisms of inflammation and interventions in CSVD. We suggest that autoimmune activity should be assessed in future studies; this knowledge would benefit the development of effective therapeutic interventions in CSVD.
Collapse
Affiliation(s)
- Ying Fu
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| | - Yaping Yan
- Key Laboratory of the Ministry of Education for Medicinal Resources and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi'an, China
| |
Collapse
|
23
|
Mancini A, Tantucci M, Mazzocchetti P, de Iure A, Durante V, Macchioni L, Giampà C, Alvino A, Gaetani L, Costa C, Tozzi A, Calabresi P, Di Filippo M. Microglial activation and the nitric oxide/cGMP/PKG pathway underlie enhanced neuronal vulnerability to mitochondrial dysfunction in experimental multiple sclerosis. Neurobiol Dis 2018; 113:97-108. [PMID: 29325869 DOI: 10.1016/j.nbd.2018.01.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 12/07/2017] [Accepted: 01/04/2018] [Indexed: 12/24/2022] Open
Abstract
During multiple sclerosis (MS), a close link has been demonstrated to occur between inflammation and neuro-axonal degeneration, leading to the hypothesis that immune mechanisms may promote neurodegeneration, leading to irreversible disease progression. Energy deficits and inflammation-driven mitochondrial dysfunction seem to be involved in this process. In this work we investigated, by the use of striatal electrophysiological field-potential recordings, if the inflammatory process associated with experimental autoimmune encephalomyelitis (EAE) is able to influence neuronal vulnerability to the blockade of mitochondrial complex IV, a crucial component for mitochondrial activity responsible of about 90% of total cellular oxygen consumption. We showed that during the acute relapsing phase of EAE, neuronal susceptibility to mitochondrial complex IV inhibition is markedly enhanced. This detrimental effect was counteracted by the pharmacological inhibition of microglia, of nitric oxide (NO) synthesis and its intracellular pathway (involving soluble guanylyl cyclase, sGC, and protein kinase G, PKG). The obtained results suggest that mitochondrial complex IV exerts an important role in maintaining neuronal energetic homeostasis during EAE. The pathological processes associated with experimental MS, and in particular the activation of microglia and of the NO pathway, lead to an increased neuronal vulnerability to mitochondrial complex IV inhibition, representing promising pharmacological targets.
Collapse
Affiliation(s)
- Andrea Mancini
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Michela Tantucci
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Petra Mazzocchetti
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Antonio de Iure
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Valentina Durante
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Lara Macchioni
- Sezione di Fisiologia e Biochimica, Dipartimento di Medicina Sperimentale, Università degli Studi di Perugia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Carmela Giampà
- Università Cattolica del Sacro Cuore, Istituto di Anatomia Umana e Biologia Cellulare, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Alessandra Alvino
- Università Cattolica del Sacro Cuore, Istituto di Anatomia Umana e Biologia Cellulare, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Lorenzo Gaetani
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Cinzia Costa
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Alessandro Tozzi
- Sezione di Fisiologia e Biochimica, Dipartimento di Medicina Sperimentale, Università degli Studi di Perugia, S. Andrea delle Fratte, 06132 Perugia, Italy; IRCCS, Fondazione Santa Lucia, via del Fosso di Fiorano 64, 00143 Rome, Italy
| | - Paolo Calabresi
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy; IRCCS, Fondazione Santa Lucia, via del Fosso di Fiorano 64, 00143 Rome, Italy
| | - Massimiliano Di Filippo
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy.
| |
Collapse
|
24
|
Bahrami Z, Firouzi M, Hashemi-Monfared A, Zahednasab H, Harirchian MH. The effect of minocycline on indolamine 2, 3 dioxygenase expression and the levels of kynurenic acid and quinolinic acid in LPS-activated primary rat microglia. Cytokine 2017; 107:125-129. [PMID: 29246652 DOI: 10.1016/j.cyto.2017.12.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 12/11/2017] [Indexed: 12/11/2022]
Abstract
Microglia are one of the most important neural cells in the central nervous system (CNS) which account for 10-15% of all cells found in the brain. A vast majority of studies indicate that microglia play a pivotal role in protection and damage of the CNS. It has been shown that microglia are mainly scavenger cells but also produce a barrage of factors that are involved in tissue repair and neural regeneration. Several lines of evidence indicate that unregulated activation of microglia in response to either endogenous or exogenous insults results in the production of toxic factors that propagate neuronal injury. Studies demonstrated that the activated microglia secret the excessive amounts of quinolinic acid (QA) and kynurenic acid (KYNA) which are highly toxic for the neuronal cells. In line with this, indolamine 2, 3 dioxygenase (IDO), an enzyme producing KYNA and QA has been shown to be elevated during the inflammation in microglia. In this study, we established primary microglial cell cultures obtained from cerebral cortices of 1-day neonatal Wistar rats. Minocycline (20-60 µM) or its vehicle was added to the culture media 60 min prior to 48 h incubation with lipopolysaccharide (LPS; 10 ng/mL). Using a specific process of adhesion and shaking of the cultured glial cells, a purified culture of approximately 94% enriched microglia was obtained and then, corroborated by immunocytochemistry (ICC). The cell viability after minocycline treatments was assessed using the MTT colorimetric assay. The expression of IDO was evaluated using qPCR. The levels of KYNA and QA were determined using enzyme-linked immunosorbent assay (ELISA). The results showed that minocycline significantly decreased the levels of both KYNA and QA in glia cells exposed to LPS. Moreover, minocycline decreased the expression of IDO in treated LPS-induced microglia. It seems that minocycline has a potent ability to oppress the inflammatory process via the decrease in production of IDO expression and the concentrations of KYNA and QA.
Collapse
Affiliation(s)
- Zahra Bahrami
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Masoumeh Firouzi
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| | | | - Hamid Zahednasab
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Mohammad Hossein Harirchian
- Iranian Centre of Neurological Research, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
25
|
Wekerle H. Brain Autoimmunity and Intestinal Microbiota: 100 Trillion Game Changers. Trends Immunol 2017; 38:483-497. [DOI: 10.1016/j.it.2017.03.008] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 02/17/2017] [Accepted: 03/31/2017] [Indexed: 02/07/2023]
|
26
|
Wekerle H. The gut-brain connection: triggering of brain autoimmune disease by commensal gut bacteria. Rheumatology (Oxford) 2017; 55:ii68-ii75. [PMID: 27856664 DOI: 10.1093/rheumatology/kew353] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Revised: 08/23/2016] [Indexed: 01/07/2023] Open
Abstract
In a transgenic model of spontaneous experimental autoimmune encephalomyelitis, autoimmune attack against the CNS requires the presence of an intact commensal gut flora. Extending this observation to human autoimmune disease, such as multiple sclerosis, we postulate that the pathogenic reaction requires the coincidence of at least three factors: a permissive genetic disposition, a pro-inflammatory intestinal microbial profile, and the accumulation of autoreactive T cells in the gut-associated lymphatic tissue. This concept may offer new approaches to diagnostic markers and non-invasive therapies.
Collapse
Affiliation(s)
- Hartmut Wekerle
- Hertie Emeritus Group, Max-Planck-Institut für Neurobiologie, Martinsried, Germany
| |
Collapse
|
27
|
Metz LM, Li DKB, Traboulsee AL, Duquette P, Eliasziw M, Cerchiaro G, Greenfield J, Riddehough A, Yeung M, Kremenchutzky M, Vorobeychik G, Freedman MS, Bhan V, Blevins G, Marriott JJ, Grand'Maison F, Lee L, Thibault M, Hill MD, Yong VW. Trial of Minocycline in a Clinically Isolated Syndrome of Multiple Sclerosis. N Engl J Med 2017; 376:2122-2133. [PMID: 28564557 DOI: 10.1056/nejmoa1608889] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND On the basis of encouraging preliminary results, we conducted a randomized, controlled trial to determine whether minocycline reduces the risk of conversion from a first demyelinating event (also known as a clinically isolated syndrome) to multiple sclerosis. METHODS During the period from January 2009 through July 2013, we randomly assigned participants who had had their first demyelinating symptoms within the previous 180 days to receive either 100 mg of minocycline, administered orally twice daily, or placebo. Administration of minocycline or placebo was continued until a diagnosis of multiple sclerosis was established or until 24 months after randomization, whichever came first. The primary outcome was conversion to multiple sclerosis (diagnosed on the basis of the 2005 McDonald criteria) within 6 months after randomization. Secondary outcomes included conversion to multiple sclerosis within 24 months after randomization and changes on magnetic resonance imaging (MRI) at 6 months and 24 months (change in lesion volume on T2-weighted MRI, cumulative number of new lesions enhanced on T1-weighted MRI ["enhancing lesions"], and cumulative combined number of unique lesions [new enhancing lesions on T1-weighted MRI plus new and newly enlarged lesions on T2-weighted MRI]). RESULTS A total of 142 eligible participants underwent randomization at 12 Canadian multiple sclerosis clinics; 72 participants were assigned to the minocycline group and 70 to the placebo group. The mean age of the participants was 35.8 years, and 68.3% were women. The unadjusted risk of conversion to multiple sclerosis within 6 months after randomization was 61.0% in the placebo group and 33.4% in the minocycline group, a difference of 27.6 percentage points (95% confidence interval [CI], 11.4 to 43.9; P=0.001). After adjustment for the number of enhancing lesions at baseline, the difference in the risk of conversion to multiple sclerosis within 6 months after randomization was 18.5 percentage points (95% CI, 3.7 to 33.3; P=0.01); the unadjusted risk difference was not significant at the 24-month secondary outcome time point (P=0.06). All secondary MRI outcomes favored minocycline over placebo at 6 months but not at 24 months. Trial withdrawals and adverse events of rash, dizziness, and dental discoloration were more frequent among participants who received minocycline than among those who received placebo. CONCLUSIONS The risk of conversion from a clinically isolated syndrome to multiple sclerosis was significantly lower with minocycline than with placebo over 6 months but not over 24 months. (Funded by the Multiple Sclerosis Society of Canada; ClinicalTrials.gov number, NCT00666887 .).
Collapse
Affiliation(s)
- Luanne M Metz
- From the Cumming School of Medicine and the Hotchkiss Brain Institute, Calgary, AB (L.M.M., G.C., J.G., M.Y., M.D.H., V.W.Y.), the University of British Columbia, Vancouver (D.K.B.L., A.L.T., A.R.), the University of Montreal, Montreal (P.D.), Western University, London, ON (M.K.), Fraser Health MS Clinic, Burnaby, BC (G.V.), the University of Ottawa and the Ottawa Hospital Research Institute, Ottawa (M.S.F.), Dalhousie University, Halifax, NS (V.B.), the University of Alberta, Edmonton (G.B.), the University of Manitoba, Winnipeg (J.J.M.), Clinique Neuro Rive-Sud, Greenfield Park, QC (F.G.), the University of Toronto, Toronto (L.L.), and CHA-Hôpital Enfant-Jésus, Quebec, QC (M.T.) - all in Canada; and Tufts University, Boston (M.E.)
| | - David K B Li
- From the Cumming School of Medicine and the Hotchkiss Brain Institute, Calgary, AB (L.M.M., G.C., J.G., M.Y., M.D.H., V.W.Y.), the University of British Columbia, Vancouver (D.K.B.L., A.L.T., A.R.), the University of Montreal, Montreal (P.D.), Western University, London, ON (M.K.), Fraser Health MS Clinic, Burnaby, BC (G.V.), the University of Ottawa and the Ottawa Hospital Research Institute, Ottawa (M.S.F.), Dalhousie University, Halifax, NS (V.B.), the University of Alberta, Edmonton (G.B.), the University of Manitoba, Winnipeg (J.J.M.), Clinique Neuro Rive-Sud, Greenfield Park, QC (F.G.), the University of Toronto, Toronto (L.L.), and CHA-Hôpital Enfant-Jésus, Quebec, QC (M.T.) - all in Canada; and Tufts University, Boston (M.E.)
| | - Anthony L Traboulsee
- From the Cumming School of Medicine and the Hotchkiss Brain Institute, Calgary, AB (L.M.M., G.C., J.G., M.Y., M.D.H., V.W.Y.), the University of British Columbia, Vancouver (D.K.B.L., A.L.T., A.R.), the University of Montreal, Montreal (P.D.), Western University, London, ON (M.K.), Fraser Health MS Clinic, Burnaby, BC (G.V.), the University of Ottawa and the Ottawa Hospital Research Institute, Ottawa (M.S.F.), Dalhousie University, Halifax, NS (V.B.), the University of Alberta, Edmonton (G.B.), the University of Manitoba, Winnipeg (J.J.M.), Clinique Neuro Rive-Sud, Greenfield Park, QC (F.G.), the University of Toronto, Toronto (L.L.), and CHA-Hôpital Enfant-Jésus, Quebec, QC (M.T.) - all in Canada; and Tufts University, Boston (M.E.)
| | - Pierre Duquette
- From the Cumming School of Medicine and the Hotchkiss Brain Institute, Calgary, AB (L.M.M., G.C., J.G., M.Y., M.D.H., V.W.Y.), the University of British Columbia, Vancouver (D.K.B.L., A.L.T., A.R.), the University of Montreal, Montreal (P.D.), Western University, London, ON (M.K.), Fraser Health MS Clinic, Burnaby, BC (G.V.), the University of Ottawa and the Ottawa Hospital Research Institute, Ottawa (M.S.F.), Dalhousie University, Halifax, NS (V.B.), the University of Alberta, Edmonton (G.B.), the University of Manitoba, Winnipeg (J.J.M.), Clinique Neuro Rive-Sud, Greenfield Park, QC (F.G.), the University of Toronto, Toronto (L.L.), and CHA-Hôpital Enfant-Jésus, Quebec, QC (M.T.) - all in Canada; and Tufts University, Boston (M.E.)
| | - Misha Eliasziw
- From the Cumming School of Medicine and the Hotchkiss Brain Institute, Calgary, AB (L.M.M., G.C., J.G., M.Y., M.D.H., V.W.Y.), the University of British Columbia, Vancouver (D.K.B.L., A.L.T., A.R.), the University of Montreal, Montreal (P.D.), Western University, London, ON (M.K.), Fraser Health MS Clinic, Burnaby, BC (G.V.), the University of Ottawa and the Ottawa Hospital Research Institute, Ottawa (M.S.F.), Dalhousie University, Halifax, NS (V.B.), the University of Alberta, Edmonton (G.B.), the University of Manitoba, Winnipeg (J.J.M.), Clinique Neuro Rive-Sud, Greenfield Park, QC (F.G.), the University of Toronto, Toronto (L.L.), and CHA-Hôpital Enfant-Jésus, Quebec, QC (M.T.) - all in Canada; and Tufts University, Boston (M.E.)
| | - Graziela Cerchiaro
- From the Cumming School of Medicine and the Hotchkiss Brain Institute, Calgary, AB (L.M.M., G.C., J.G., M.Y., M.D.H., V.W.Y.), the University of British Columbia, Vancouver (D.K.B.L., A.L.T., A.R.), the University of Montreal, Montreal (P.D.), Western University, London, ON (M.K.), Fraser Health MS Clinic, Burnaby, BC (G.V.), the University of Ottawa and the Ottawa Hospital Research Institute, Ottawa (M.S.F.), Dalhousie University, Halifax, NS (V.B.), the University of Alberta, Edmonton (G.B.), the University of Manitoba, Winnipeg (J.J.M.), Clinique Neuro Rive-Sud, Greenfield Park, QC (F.G.), the University of Toronto, Toronto (L.L.), and CHA-Hôpital Enfant-Jésus, Quebec, QC (M.T.) - all in Canada; and Tufts University, Boston (M.E.)
| | - Jamie Greenfield
- From the Cumming School of Medicine and the Hotchkiss Brain Institute, Calgary, AB (L.M.M., G.C., J.G., M.Y., M.D.H., V.W.Y.), the University of British Columbia, Vancouver (D.K.B.L., A.L.T., A.R.), the University of Montreal, Montreal (P.D.), Western University, London, ON (M.K.), Fraser Health MS Clinic, Burnaby, BC (G.V.), the University of Ottawa and the Ottawa Hospital Research Institute, Ottawa (M.S.F.), Dalhousie University, Halifax, NS (V.B.), the University of Alberta, Edmonton (G.B.), the University of Manitoba, Winnipeg (J.J.M.), Clinique Neuro Rive-Sud, Greenfield Park, QC (F.G.), the University of Toronto, Toronto (L.L.), and CHA-Hôpital Enfant-Jésus, Quebec, QC (M.T.) - all in Canada; and Tufts University, Boston (M.E.)
| | - Andrew Riddehough
- From the Cumming School of Medicine and the Hotchkiss Brain Institute, Calgary, AB (L.M.M., G.C., J.G., M.Y., M.D.H., V.W.Y.), the University of British Columbia, Vancouver (D.K.B.L., A.L.T., A.R.), the University of Montreal, Montreal (P.D.), Western University, London, ON (M.K.), Fraser Health MS Clinic, Burnaby, BC (G.V.), the University of Ottawa and the Ottawa Hospital Research Institute, Ottawa (M.S.F.), Dalhousie University, Halifax, NS (V.B.), the University of Alberta, Edmonton (G.B.), the University of Manitoba, Winnipeg (J.J.M.), Clinique Neuro Rive-Sud, Greenfield Park, QC (F.G.), the University of Toronto, Toronto (L.L.), and CHA-Hôpital Enfant-Jésus, Quebec, QC (M.T.) - all in Canada; and Tufts University, Boston (M.E.)
| | - Michael Yeung
- From the Cumming School of Medicine and the Hotchkiss Brain Institute, Calgary, AB (L.M.M., G.C., J.G., M.Y., M.D.H., V.W.Y.), the University of British Columbia, Vancouver (D.K.B.L., A.L.T., A.R.), the University of Montreal, Montreal (P.D.), Western University, London, ON (M.K.), Fraser Health MS Clinic, Burnaby, BC (G.V.), the University of Ottawa and the Ottawa Hospital Research Institute, Ottawa (M.S.F.), Dalhousie University, Halifax, NS (V.B.), the University of Alberta, Edmonton (G.B.), the University of Manitoba, Winnipeg (J.J.M.), Clinique Neuro Rive-Sud, Greenfield Park, QC (F.G.), the University of Toronto, Toronto (L.L.), and CHA-Hôpital Enfant-Jésus, Quebec, QC (M.T.) - all in Canada; and Tufts University, Boston (M.E.)
| | - Marcelo Kremenchutzky
- From the Cumming School of Medicine and the Hotchkiss Brain Institute, Calgary, AB (L.M.M., G.C., J.G., M.Y., M.D.H., V.W.Y.), the University of British Columbia, Vancouver (D.K.B.L., A.L.T., A.R.), the University of Montreal, Montreal (P.D.), Western University, London, ON (M.K.), Fraser Health MS Clinic, Burnaby, BC (G.V.), the University of Ottawa and the Ottawa Hospital Research Institute, Ottawa (M.S.F.), Dalhousie University, Halifax, NS (V.B.), the University of Alberta, Edmonton (G.B.), the University of Manitoba, Winnipeg (J.J.M.), Clinique Neuro Rive-Sud, Greenfield Park, QC (F.G.), the University of Toronto, Toronto (L.L.), and CHA-Hôpital Enfant-Jésus, Quebec, QC (M.T.) - all in Canada; and Tufts University, Boston (M.E.)
| | - Galina Vorobeychik
- From the Cumming School of Medicine and the Hotchkiss Brain Institute, Calgary, AB (L.M.M., G.C., J.G., M.Y., M.D.H., V.W.Y.), the University of British Columbia, Vancouver (D.K.B.L., A.L.T., A.R.), the University of Montreal, Montreal (P.D.), Western University, London, ON (M.K.), Fraser Health MS Clinic, Burnaby, BC (G.V.), the University of Ottawa and the Ottawa Hospital Research Institute, Ottawa (M.S.F.), Dalhousie University, Halifax, NS (V.B.), the University of Alberta, Edmonton (G.B.), the University of Manitoba, Winnipeg (J.J.M.), Clinique Neuro Rive-Sud, Greenfield Park, QC (F.G.), the University of Toronto, Toronto (L.L.), and CHA-Hôpital Enfant-Jésus, Quebec, QC (M.T.) - all in Canada; and Tufts University, Boston (M.E.)
| | - Mark S Freedman
- From the Cumming School of Medicine and the Hotchkiss Brain Institute, Calgary, AB (L.M.M., G.C., J.G., M.Y., M.D.H., V.W.Y.), the University of British Columbia, Vancouver (D.K.B.L., A.L.T., A.R.), the University of Montreal, Montreal (P.D.), Western University, London, ON (M.K.), Fraser Health MS Clinic, Burnaby, BC (G.V.), the University of Ottawa and the Ottawa Hospital Research Institute, Ottawa (M.S.F.), Dalhousie University, Halifax, NS (V.B.), the University of Alberta, Edmonton (G.B.), the University of Manitoba, Winnipeg (J.J.M.), Clinique Neuro Rive-Sud, Greenfield Park, QC (F.G.), the University of Toronto, Toronto (L.L.), and CHA-Hôpital Enfant-Jésus, Quebec, QC (M.T.) - all in Canada; and Tufts University, Boston (M.E.)
| | - Virender Bhan
- From the Cumming School of Medicine and the Hotchkiss Brain Institute, Calgary, AB (L.M.M., G.C., J.G., M.Y., M.D.H., V.W.Y.), the University of British Columbia, Vancouver (D.K.B.L., A.L.T., A.R.), the University of Montreal, Montreal (P.D.), Western University, London, ON (M.K.), Fraser Health MS Clinic, Burnaby, BC (G.V.), the University of Ottawa and the Ottawa Hospital Research Institute, Ottawa (M.S.F.), Dalhousie University, Halifax, NS (V.B.), the University of Alberta, Edmonton (G.B.), the University of Manitoba, Winnipeg (J.J.M.), Clinique Neuro Rive-Sud, Greenfield Park, QC (F.G.), the University of Toronto, Toronto (L.L.), and CHA-Hôpital Enfant-Jésus, Quebec, QC (M.T.) - all in Canada; and Tufts University, Boston (M.E.)
| | - Gregg Blevins
- From the Cumming School of Medicine and the Hotchkiss Brain Institute, Calgary, AB (L.M.M., G.C., J.G., M.Y., M.D.H., V.W.Y.), the University of British Columbia, Vancouver (D.K.B.L., A.L.T., A.R.), the University of Montreal, Montreal (P.D.), Western University, London, ON (M.K.), Fraser Health MS Clinic, Burnaby, BC (G.V.), the University of Ottawa and the Ottawa Hospital Research Institute, Ottawa (M.S.F.), Dalhousie University, Halifax, NS (V.B.), the University of Alberta, Edmonton (G.B.), the University of Manitoba, Winnipeg (J.J.M.), Clinique Neuro Rive-Sud, Greenfield Park, QC (F.G.), the University of Toronto, Toronto (L.L.), and CHA-Hôpital Enfant-Jésus, Quebec, QC (M.T.) - all in Canada; and Tufts University, Boston (M.E.)
| | - James J Marriott
- From the Cumming School of Medicine and the Hotchkiss Brain Institute, Calgary, AB (L.M.M., G.C., J.G., M.Y., M.D.H., V.W.Y.), the University of British Columbia, Vancouver (D.K.B.L., A.L.T., A.R.), the University of Montreal, Montreal (P.D.), Western University, London, ON (M.K.), Fraser Health MS Clinic, Burnaby, BC (G.V.), the University of Ottawa and the Ottawa Hospital Research Institute, Ottawa (M.S.F.), Dalhousie University, Halifax, NS (V.B.), the University of Alberta, Edmonton (G.B.), the University of Manitoba, Winnipeg (J.J.M.), Clinique Neuro Rive-Sud, Greenfield Park, QC (F.G.), the University of Toronto, Toronto (L.L.), and CHA-Hôpital Enfant-Jésus, Quebec, QC (M.T.) - all in Canada; and Tufts University, Boston (M.E.)
| | - Francois Grand'Maison
- From the Cumming School of Medicine and the Hotchkiss Brain Institute, Calgary, AB (L.M.M., G.C., J.G., M.Y., M.D.H., V.W.Y.), the University of British Columbia, Vancouver (D.K.B.L., A.L.T., A.R.), the University of Montreal, Montreal (P.D.), Western University, London, ON (M.K.), Fraser Health MS Clinic, Burnaby, BC (G.V.), the University of Ottawa and the Ottawa Hospital Research Institute, Ottawa (M.S.F.), Dalhousie University, Halifax, NS (V.B.), the University of Alberta, Edmonton (G.B.), the University of Manitoba, Winnipeg (J.J.M.), Clinique Neuro Rive-Sud, Greenfield Park, QC (F.G.), the University of Toronto, Toronto (L.L.), and CHA-Hôpital Enfant-Jésus, Quebec, QC (M.T.) - all in Canada; and Tufts University, Boston (M.E.)
| | - Liesly Lee
- From the Cumming School of Medicine and the Hotchkiss Brain Institute, Calgary, AB (L.M.M., G.C., J.G., M.Y., M.D.H., V.W.Y.), the University of British Columbia, Vancouver (D.K.B.L., A.L.T., A.R.), the University of Montreal, Montreal (P.D.), Western University, London, ON (M.K.), Fraser Health MS Clinic, Burnaby, BC (G.V.), the University of Ottawa and the Ottawa Hospital Research Institute, Ottawa (M.S.F.), Dalhousie University, Halifax, NS (V.B.), the University of Alberta, Edmonton (G.B.), the University of Manitoba, Winnipeg (J.J.M.), Clinique Neuro Rive-Sud, Greenfield Park, QC (F.G.), the University of Toronto, Toronto (L.L.), and CHA-Hôpital Enfant-Jésus, Quebec, QC (M.T.) - all in Canada; and Tufts University, Boston (M.E.)
| | - Manon Thibault
- From the Cumming School of Medicine and the Hotchkiss Brain Institute, Calgary, AB (L.M.M., G.C., J.G., M.Y., M.D.H., V.W.Y.), the University of British Columbia, Vancouver (D.K.B.L., A.L.T., A.R.), the University of Montreal, Montreal (P.D.), Western University, London, ON (M.K.), Fraser Health MS Clinic, Burnaby, BC (G.V.), the University of Ottawa and the Ottawa Hospital Research Institute, Ottawa (M.S.F.), Dalhousie University, Halifax, NS (V.B.), the University of Alberta, Edmonton (G.B.), the University of Manitoba, Winnipeg (J.J.M.), Clinique Neuro Rive-Sud, Greenfield Park, QC (F.G.), the University of Toronto, Toronto (L.L.), and CHA-Hôpital Enfant-Jésus, Quebec, QC (M.T.) - all in Canada; and Tufts University, Boston (M.E.)
| | - Michael D Hill
- From the Cumming School of Medicine and the Hotchkiss Brain Institute, Calgary, AB (L.M.M., G.C., J.G., M.Y., M.D.H., V.W.Y.), the University of British Columbia, Vancouver (D.K.B.L., A.L.T., A.R.), the University of Montreal, Montreal (P.D.), Western University, London, ON (M.K.), Fraser Health MS Clinic, Burnaby, BC (G.V.), the University of Ottawa and the Ottawa Hospital Research Institute, Ottawa (M.S.F.), Dalhousie University, Halifax, NS (V.B.), the University of Alberta, Edmonton (G.B.), the University of Manitoba, Winnipeg (J.J.M.), Clinique Neuro Rive-Sud, Greenfield Park, QC (F.G.), the University of Toronto, Toronto (L.L.), and CHA-Hôpital Enfant-Jésus, Quebec, QC (M.T.) - all in Canada; and Tufts University, Boston (M.E.)
| | - V Wee Yong
- From the Cumming School of Medicine and the Hotchkiss Brain Institute, Calgary, AB (L.M.M., G.C., J.G., M.Y., M.D.H., V.W.Y.), the University of British Columbia, Vancouver (D.K.B.L., A.L.T., A.R.), the University of Montreal, Montreal (P.D.), Western University, London, ON (M.K.), Fraser Health MS Clinic, Burnaby, BC (G.V.), the University of Ottawa and the Ottawa Hospital Research Institute, Ottawa (M.S.F.), Dalhousie University, Halifax, NS (V.B.), the University of Alberta, Edmonton (G.B.), the University of Manitoba, Winnipeg (J.J.M.), Clinique Neuro Rive-Sud, Greenfield Park, QC (F.G.), the University of Toronto, Toronto (L.L.), and CHA-Hôpital Enfant-Jésus, Quebec, QC (M.T.) - all in Canada; and Tufts University, Boston (M.E.)
| |
Collapse
|
28
|
Korean Red Ginseng mitigates spinal demyelination in a model of acute multiple sclerosis by downregulating p38 mitogen-activated protein kinase and nuclear factor-κB signaling pathways. J Ginseng Res 2017; 42:436-446. [PMID: 30337803 PMCID: PMC6187097 DOI: 10.1016/j.jgr.2017.04.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 04/12/2017] [Accepted: 04/26/2017] [Indexed: 01/08/2023] Open
Abstract
Background The potential therapeutic values of Korean Red Ginseng extract (KRGE) in autoimmune disorders of nervous system have not been fully investigated. Methods We used an acute experimental autoimmune encephalomyelitis animal model of multiple sclerosis and determined the effects and mechanism of KRGE on spinal myelination. Results Pretreatment with KRGE (100 mg/kg, orally) for 10 days before immunization with myelin basic protein (MBP)68-82 peptide exerted a protective effect against demyelination in the spinal cord, with inhibited recruitment and activation of immune cells including microglia, decreased mRNA expression of detrimental inflammatory mediators (interleukin-6, interferon-γ, and cyclooxygenase-2), but increased mRNA expression of protective inflammatory mediators (insulin-like growth factor β1, transforming growth factor β, and vascular endothelial growth factor-1). These results were associated with significant downregulation of p38 mitogen-activated protein kinase and nuclear factor-κB signaling pathways in microglia/macrophages, T cells, and astrocytes. Conclusion Our findings suggest that KRGE alleviates spinal demyelination in acute experimental autoimmune encephalomyelitis through inhibiting the activation of the p38 mitogen-activated protein kinase/nuclear factor-κB signaling pathway. Therefore, KRGE might be used as a new therapeutic for autoimmune disorders such as multiple sclerosis, although further investigation is needed.
Collapse
|
29
|
|
30
|
Selective dentate gyrus disruption causes memory impairment at the early stage of experimental multiple sclerosis. Brain Behav Immun 2017; 60:240-254. [PMID: 27847283 DOI: 10.1016/j.bbi.2016.11.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 11/07/2016] [Accepted: 11/12/2016] [Indexed: 11/21/2022] Open
Abstract
Memory impairment is an early and disabling manifestation of multiple sclerosis whose anatomical and biological substrates are still poorly understood. We thus investigated whether memory impairment encountered at the early stage of the disease could be explained by a differential vulnerability of particular hippocampal subfields. By using experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis, we identified that early memory impairment was associated with selective alteration of the dentate gyrus as pinpointed in vivo with diffusion-tensor-imaging (DTI). Neuromorphometric analyses and electrophysiological recordings confirmed dendritic degeneration, alteration in glutamatergic synaptic transmission and impaired long-term synaptic potentiation selectively in the dentate gyrus, but not in CA1, together with a more severe pattern of microglial activation in this subfield. Systemic injections of the microglial inhibitor minocycline prevented DTI, morphological, electrophysiological and behavioral impairments in EAE-mice. Furthermore, daily infusions of minocycline specifically within the dentate gyrus were sufficient to prevent memory impairment in EAE-mice while infusions of minocycline within CA1 were inefficient. We conclude that early memory impairment in EAE is due to a selective disruption of the dentate gyrus associated with microglia activation. These results open new pathophysiological, imaging, and therapeutic perspectives for memory impairment in multiple sclerosis.
Collapse
|
31
|
Madeira MH, Boia R, Ambrósio AF, Santiago AR. Having a Coffee Break: The Impact of Caffeine Consumption on Microglia-Mediated Inflammation in Neurodegenerative Diseases. Mediators Inflamm 2017; 2017:4761081. [PMID: 28250576 PMCID: PMC5307009 DOI: 10.1155/2017/4761081] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 01/12/2017] [Indexed: 12/20/2022] Open
Abstract
Caffeine is the major component of coffee and the most consumed psychostimulant in the world and at nontoxic doses acts as a nonselective adenosine receptor antagonist. Epidemiological evidence suggests that caffeine consumption reduces the risk of several neurological and neurodegenerative diseases. However, despite the beneficial effects of caffeine consumption in human health and behaviour, the mechanisms by which it impacts the pathophysiology of neurodegenerative diseases still remain to be clarified. A promising hypothesis is that caffeine controls microglia-mediated neuroinflammatory response associated with the majority of neurodegenerative conditions. Accordingly, it has been already described that the modulation of adenosine receptors, namely, the A2A receptor, affords neuroprotection through the control of microglia reactivity and neuroinflammation. In this review, we will summarize the main effects of caffeine in the modulation of neuroinflammation in neurodegenerative diseases.
Collapse
Affiliation(s)
- Maria H. Madeira
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- CNC.IBILI Consortium, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Raquel Boia
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- CNC.IBILI Consortium, University of Coimbra, 3004-504 Coimbra, Portugal
| | - António F. Ambrósio
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- CNC.IBILI Consortium, University of Coimbra, 3004-504 Coimbra, Portugal
- Association for Innovation and Biomedical Research on Light and Image (AIBILI), 3000-548 Coimbra, Portugal
| | - Ana R. Santiago
- Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- CNC.IBILI Consortium, University of Coimbra, 3004-504 Coimbra, Portugal
- Association for Innovation and Biomedical Research on Light and Image (AIBILI), 3000-548 Coimbra, Portugal
| |
Collapse
|
32
|
Abstract
As the immune-competent cells of the brain, microglia play an increasingly important role in maintaining normal brain function. They invade the brain early in development, transform into a highly ramified phenotype, and constantly screen their environment. Microglia are activated by any type of pathologic event or change in brain homeostasis. This activation process is highly diverse and depends on the context and type of the stressor or pathology. Microglia can strongly influence the pathologic outcome or response to a stressor due to the release of a plethora of substances, including cytokines, chemokines, and growth factors. They are the professional phagocytes of the brain and help orchestrate the immunological response by interacting with infiltrating immune cells. We describe here the diversity of microglia phenotypes and their responses in health, aging, and disease. We also review the current literature about the impact of lifestyle on microglia responses and discuss treatment options that modulate microglial phenotypes.
Collapse
Affiliation(s)
- Susanne A Wolf
- Cellular Neurosciences, Max Delbrück Centre for Molecular Medicine in the Helmholtz Association, Berlin 13092, Germany;
| | - H W G M Boddeke
- Department of Neuroscience, University of Groningen, University Medical Center Groningen, Groningen 9713, The Netherlands
| | - Helmut Kettenmann
- Cellular Neurosciences, Max Delbrück Centre for Molecular Medicine in the Helmholtz Association, Berlin 13092, Germany;
| |
Collapse
|
33
|
Hahn JN, Kaushik DK, Mishra MK, Wang J, Silva C, Yong VW. Impact of Minocycline on Extracellular Matrix Metalloproteinase Inducer, a Factor Implicated in Multiple Sclerosis Immunopathogenesis. THE JOURNAL OF IMMUNOLOGY 2016; 197:3850-3860. [PMID: 27733550 DOI: 10.4049/jimmunol.1600436] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Accepted: 09/12/2016] [Indexed: 12/28/2022]
Abstract
Extracellular matrix metalloproteinase inducer (EMMPRIN, CD147) is a transmembrane glycoprotein that is upregulated on leukocytes in active lesions in multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE). Administration of anti-EMMPRIN Abs reduces the severity of EAE. Minocycline is a tetracycline antibiotic with immune-modulatory properties that decreases the severity of EAE; it was recently found to attenuate the conversion from a first demyelinating event to clinically definite MS in a phase III trial. We investigated whether and how minocycline affects the expression of EMMPRIN on T cells in culture and in mice afflicted with EAE. EMMPRIN expression in cultures of mouse splenocytes or human PBMCs was elevated upon polyclonal T cell activation, and this was reduced by minocycline correspondent with decreased P-Akt levels. An established MS medication, IFN-β, also diminished EMMPRIN levels on human cells whereas this was not readily observed for fingolimod or monomethylfumarate. In EAE-afflicted mice, minocycline treatment significantly reduced EMMPRIN levels on splenic lymphocytes at the presymptomatic (day 7) phase, and prevented the development of disease. Day 7 spleen transcripts from minocycline-treated EAE mice had a significantly lower MMP-9/TIMP-1 ratio, and significantly lower MCT-1 and CD98 levels, factors associated with EMMPRIN function. Day 16 (peak clinical severity) CNS samples from EAE mice had prominent representation of inflammatory perivascular cuffs, inflammatory molecules and EMMPRIN, and these were abrogated by minocycline. Overall, minocycline attenuated the activation-induced elevation of EMMPRIN on T cells in culture and in EAE mice, correspondent with reduced immune function and EAE CNS pathology.
Collapse
Affiliation(s)
- Jennifer N Hahn
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Deepak K Kaushik
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Manoj K Mishra
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Jianxiong Wang
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Claudia Silva
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - V Wee Yong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| |
Collapse
|
34
|
Minocycline ameliorates D-galactose-induced memory deficits and loss of Arc/Arg3.1 expression. Mol Biol Rep 2016; 43:1157-63. [PMID: 27497819 DOI: 10.1007/s11033-016-4051-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 08/01/2016] [Indexed: 12/13/2022]
Abstract
Dysfunction of learning and memory is widely found in many neurological diseases. Understanding how to preserve the normal function of learning and memory will be extremely beneficial for the treatment of these diseases. However, the possible protective effect of minocycline in memory impairment is unknown. We used the well-established D-galactose rat amnesia model and two behavioral tasks, the Morris water maze and the step-down task, for memory evaluation. Western blot and PCR were used to examine the protein and mRNA levels of Arc/Arg3.1. We report that minocycline supplementation ameliorates both the spatial and fear memory deficits caused by D-galactose. We also found that Arc/Arg3.1, c-fos, and brain-derived neurotrophic factor levels are decreased in the D-galactose animal model, and that minocycline reverses the protein and mRNA levels of Arc in the hippocampus, suggesting the potential role of Arc/Arg3.1 in minocycline's neuroprotective mechanism. Our study strongly suggests that minocycline can be used as a novel treatment for memory impairment in neurological diseases.
Collapse
|
35
|
Davies CL, Miron VE. Distinct origins, gene expression and function of microglia and monocyte-derived macrophages in CNS myelin injury and regeneration. Clin Immunol 2016; 189:57-62. [PMID: 27377535 DOI: 10.1016/j.clim.2016.06.016] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 06/27/2016] [Accepted: 06/30/2016] [Indexed: 01/07/2023]
Abstract
Central nervous system (CNS) injury incurs a rapid innate immune response, including that from macrophages derived from endogenous microglia and circulating monocytes infiltrating the lesion site. One example of such injury is the demyelination observed in the autoimmune disease multiple sclerosis (MS), where macrophages are implicated in both myelin injury and regeneration. Although initially microglia and monocyte-derived macrophages were considered to have identical origins, gene expression, and function, recent advances have revealed important distinctions in all three categories and have caused a paradigm shift in view of their unique identity and roles. This has important consequences for understanding their individual contribution to neurological function and therapeutic targeting of these populations in diseases like MS. Here, we address the differences between CNS endogenous and exogenously-derived macrophages with a particular focus on myelin damage and regeneration.
Collapse
Affiliation(s)
- Claire L Davies
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, United Kingdom
| | - Veronique E Miron
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, United Kingdom.
| |
Collapse
|
36
|
Möller T, Bard F, Bhattacharya A, Biber K, Campbell B, Dale E, Eder C, Gan L, Garden GA, Hughes ZA, Pearse DD, Staal RGW, Sayed FA, Wes PD, Boddeke HWGM. Critical data-based re-evaluation of minocycline as a putative specific microglia inhibitor. Glia 2016; 64:1788-94. [PMID: 27246804 DOI: 10.1002/glia.23007] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Accepted: 05/04/2016] [Indexed: 12/11/2022]
Abstract
Minocycline, a second generation broad-spectrum antibiotic, has been frequently postulated to be a "microglia inhibitor." A considerable number of publications have used minocycline as a tool and concluded, after achieving a pharmacological effect, that the effect must be due to "inhibition" of microglia. It is, however, unclear how this "inhibition" is achieved at the molecular and cellular levels. Here, we weigh the evidence whether minocycline is indeed a bona fide microglia inhibitor and discuss how data generated with minocycline should be interpreted. GLIA 2016;64:1788-1794.
Collapse
Affiliation(s)
- Thomas Möller
- Neuroinflammation Disease Biology Unit, Lundbeck Research USA, Paramus, New Jersey.,Department of Neurology, University of Washington, Seattle, Washington
| | | | - Anindya Bhattacharya
- Janssen Research & Development, LLC., Neuroscience Drug Discovery, San Diego, California
| | - Knut Biber
- Department of Psychiatry and Psychotherapy, University Hospital Freiburg, Freiburg, Germany.,Department of Neuroscience, University Medical Center Groningen, Groningen, The Netherlands
| | - Brian Campbell
- Neuroinflammation Disease Biology Unit, Lundbeck Research USA, Paramus, New Jersey
| | - Elena Dale
- Neuroinflammation Disease Biology Unit, Lundbeck Research USA, Paramus, New Jersey
| | - Claudia Eder
- Institute for Infection and Immunity, St. George's - University of London, London, United Kingdom
| | - Li Gan
- Gladstone Institute for Neurodegeneration, San Francisco, California
| | - Gwenn A Garden
- Department of Neurology, University of Washington, Seattle, Washington
| | - Zoë A Hughes
- Neuroscience & Pain Research Unit, Pfizer Global Research, Cambridge, Massachusetts
| | - Damien D Pearse
- Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Roland G W Staal
- Neuroinflammation Disease Biology Unit, Lundbeck Research USA, Paramus, New Jersey
| | - Faten A Sayed
- Gladstone Institute for Neurodegeneration, San Francisco, California.,Neuroscience Graduate Program, University of California, San Francisco, San Francisco, California
| | - Paul D Wes
- Neuroinflammation Disease Biology Unit, Lundbeck Research USA, Paramus, New Jersey
| | | |
Collapse
|
37
|
Di Filippo M, de Iure A, Giampà C, Chiasserini D, Tozzi A, Orvietani PL, Ghiglieri V, Tantucci M, Durante V, Quiroga-Varela A, Mancini A, Costa C, Sarchielli P, Fusco FR, Calabresi P. Persistent activation of microglia and NADPH oxidase [corrected] drive hippocampal dysfunction in experimental multiple sclerosis. Sci Rep 2016; 6:20926. [PMID: 26887636 PMCID: PMC4757867 DOI: 10.1038/srep20926] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 01/13/2016] [Indexed: 01/08/2023] Open
Abstract
Cognitive impairment is common in multiple sclerosis (MS). Unfortunately, the synaptic and molecular mechanisms underlying MS-associated cognitive dysfunction are largely unknown. We explored the presence and the underlying mechanism of cognitive and synaptic hippocampal dysfunction during the remission phase of experimental MS. Experiments were performed in a chronic-relapsing experimental autoimmune encephalomyelitis (EAE) model of MS, after the resolution of motor deficits. Immunohistochemistry and patch-clamp recordings were performed in the CA1 hippocampal area. The hole-board was utilized as cognitive/behavioural test. In the remission phase of experimental MS, hippocampal microglial cells showed signs of activation, CA1 hippocampal synapses presented an impaired long-term potentiation (LTP) and an alteration of spatial tests became evident. The activation of hippocampal microglia mediated synaptic and cognitive/behavioural alterations during EAE. Specifically, LTP blockade was found to be caused by the reactive oxygen species (ROS)-producing enzyme nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. We suggest that in the remission phase of experimental MS microglia remains activated, causing synaptic dysfunctions mediated by NADPH oxidase. Inhibition of microglial activation and NADPH oxidase may represent a promising strategy to prevent neuroplasticity impairment associated with active neuro-inflammation, with the aim to improve cognition and counteract MS disease progression.
Collapse
Affiliation(s)
- Massimiliano Di Filippo
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Antonio de Iure
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Carmela Giampà
- IRCCS, Fondazione Santa Lucia, via del Fosso di Fiorano 64, 00143, Rome, Italy
| | - Davide Chiasserini
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Alessandro Tozzi
- IRCCS, Fondazione Santa Lucia, via del Fosso di Fiorano 64, 00143, Rome, Italy.,Sezione di Fisiologia e Biochimica, Dipartimento di Medicina Sperimentale, Università degli Studi di Perugia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Pier Luigi Orvietani
- Sezione di Fisiologia e Biochimica, Dipartimento di Medicina Sperimentale, Università degli Studi di Perugia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Veronica Ghiglieri
- IRCCS, Fondazione Santa Lucia, via del Fosso di Fiorano 64, 00143, Rome, Italy
| | - Michela Tantucci
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Valentina Durante
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Ana Quiroga-Varela
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Andrea Mancini
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Cinzia Costa
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | - Paola Sarchielli
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy
| | | | - Paolo Calabresi
- Clinica Neurologica, Dipartimento di Medicina, Università degli Studi di Perugia, Ospedale Santa Maria della Misericordia, S. Andrea delle Fratte, 06132 Perugia, Italy.,IRCCS, Fondazione Santa Lucia, via del Fosso di Fiorano 64, 00143, Rome, Italy
| |
Collapse
|
38
|
Drenger B, Fellig Y, Ben-David D, Mintz B, Idrees S, Or O, Kaplan L, Ginosar Y, Barzilay Y. Minocycline Effectively Protects the Rabbit's Spinal Cord From Aortic Occlusion-Related Ischemia. J Cardiothorac Vasc Anesth 2015; 30:282-90. [PMID: 26853309 DOI: 10.1053/j.jvca.2015.11.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Indexed: 01/08/2023]
Abstract
OBJECTIVES To identify the minocycline anti-inflammatory and antiapoptotic mechanisms through which it is believed to exert spinal cord protection during aortic occlusion in the rabbit model. DESIGN An animal model of aortic occlusion-related spinal cord ischemia. Randomized study with a control group and pre-ischemia and post-ischemia escalating doses of minocycline to high-dose minocycline in the presence of either hyperglycemia, a pro-apoptotic maneuver, or wortmannin, a specific phosphatidylinositol 3-kinase antagonist. SETTING Tertiary medical center and school of medicine laboratory. PARTICIPANTS Laboratory animals-rabbits. INTERVENTIONS Balloon obstruction of infrarenal aorta introduced via femoral artery incision. RESULTS Severe hindlimb paralysis (mean Tarlov score 0.36±0.81 out of 3) was observed in all the control group animals (9 of 11 with paraplegia and 2 of 11 with paraparesis) compared with 11 of 12 neurologically intact animals (mean Tarlov score 2.58±0.90 [p = 0.001 compared with control]) in the high-dose minocycline group. This protective effect was observed partially during a state of hyperglycemia and was completely abrogated by wortmannin. Minocycline administration resulted in higher neurologic scores (p = 0.003) and a shift to viable neurons and more apoptotic-stained nuclei resulting from reduced necrosis (p = 0.001). CONCLUSIONS In a rabbit model of infrarenal aortic occlusion, minocycline effectively reduced paraplegia by increasing the number of viable neurons in a dose-dependent manner. Its action was completely abrogated by inhibiting the phosphatidylinositol 3-kinase pathway and was inhibited partially by the pro-apoptotic hyperglycemia maneuver, indicating that the activation of cell salvage pathways and mitochondrial sites are possible targets of minocycline action in an ischemic spinal cord.
Collapse
Affiliation(s)
| | - Yakov Fellig
- Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Dror Ben-David
- Department of Orthopedic Surgery, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Bella Mintz
- Department of Anesthesiology and Critical Care Medicine
| | - Suhel Idrees
- Department of Anesthesiology and Critical Care Medicine
| | - Omer Or
- Department of Orthopedic Surgery, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Leon Kaplan
- Department of Orthopedic Surgery, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | - Yair Barzilay
- Department of Orthopedic Surgery, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
39
|
μ-Opioid and N-methyl-D-aspartate receptors in the amygdala contribute to minocycline-induced potentiation of morphine analgesia in rats. Behav Pharmacol 2015; 26:383-92. [DOI: 10.1097/fbp.0000000000000126] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
40
|
Lee MJ, Jang M, Choi J, Chang BS, Kim DY, Kim SH, Kwak YS, Oh S, Lee JH, Chang BJ, Nah SY, Cho IH. Korean Red Ginseng and Ginsenoside-Rb1/-Rg1 Alleviate Experimental Autoimmune Encephalomyelitis by Suppressing Th1 and Th17 Cells and Upregulating Regulatory T Cells. Mol Neurobiol 2015; 53:1977-2002. [PMID: 25846819 DOI: 10.1007/s12035-015-9131-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 02/22/2015] [Indexed: 11/26/2022]
Abstract
The effects of Korean red ginseng extract (KRGE) on autoimmune disorders of the nervous system are not clear. We investigated whether KRGE has a beneficial effect on acute and chronic experimental autoimmune encephalomyelitis (EAE). Pretreatment (daily from 10 days before immunization with myelin basic protein peptide) with KRGE significantly attenuated clinical signs and loss of body weight and was associated with the suppression of spinal demyelination and glial activation in acute EAE rats, while onset treatment (daily after the appearance of clinical symptoms) did not. The suppressive effect of KRGE corresponded to the messenger RNA (mRNA) expression of proinflammatory cytokines (tumor necrosis factor-α [TNF-α] and interleukin [IL]-1β), chemokines (RANTES, monocyte chemotactic protein-1 [MCP-1], and macrophage inflammatory protein-1α [MIP-1α]), adhesion molecules (intercellular adhesion molecule-1 [ICAM-1], vascular cell adhesion molecule-1 [VCAM-1], and platelet endothelial cell adhesion molecule [PECAM-1]), and inducible nitric oxide synthase in the spinal cord after immunization. Interestingly, in acute EAE rats, pretreatment with KRGE significantly reduced the population of CD4(+), CD4(+)/IFN-γ(+), and CD4(+)/IL-17(+) T cells in the spinal cord and lymph nodes, corresponding to the downregulation of mRNA expression of IFN-γ, IL-17, and IL-23 in the spinal cord. On the other hand, KRGE pretreatment increased the population of CD4(+)/Foxp3(+) T cells in the spinal cord and lymph nodes of these rats, corresponding to the upregulation of mRNA expression of Foxp3 in the spinal cord. Interestingly, intrathecal pretreatment of rats with ginsenosides (Rg1 and Rb1) significantly decreased behavioral impairment. These results strongly indicate that KRGE has a beneficial effect on the development and progression of EAE by suppressing T helper 1 (Th1) and Th17 T cells and upregulating regulatory T cells. Additionally, pre- and onset treatment with KRGE alleviated neurological impairment of myelin oligodendrocyte glycoprotein(35-55)-induced mouse model of chronic EAE. These results warrant further investigation of KRGE as preventive or therapeutic strategies for autoimmune disorders, such as multiple sclerosis.
Collapse
MESH Headings
- Animals
- Blood-Brain Barrier/drug effects
- Blood-Brain Barrier/pathology
- Chemokines/metabolism
- Chronic Disease
- Demyelinating Diseases/complications
- Demyelinating Diseases/drug therapy
- Demyelinating Diseases/pathology
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Female
- Fibronectins/metabolism
- Ginsenosides/pharmacology
- Ginsenosides/therapeutic use
- Inflammation/complications
- Inflammation/drug therapy
- Inflammation/pathology
- Macrophages/drug effects
- Macrophages/metabolism
- Macrophages/pathology
- Mice, Inbred C57BL
- Neuroglia/drug effects
- Neuroglia/metabolism
- Neuroglia/pathology
- Panax/chemistry
- Plant Extracts/pharmacology
- Plant Extracts/therapeutic use
- Platelet Endothelial Cell Adhesion Molecule-1/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats, Inbred Lew
- Spinal Cord/drug effects
- Spinal Cord/pathology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- Th1 Cells/drug effects
- Th1 Cells/immunology
- Th17 Cells/drug effects
- Th17 Cells/immunology
- Up-Regulation/drug effects
Collapse
Affiliation(s)
- Min Jung Lee
- Department of Cancer Preventive Material Development, College of Korean Medicine, Kyung Hee University, Seoul, 130-701, Republic of Korea
- Department of Convergence Medical Sciences, College of Korean Medicine, Kyung Hee University, Seoul, 130-701, Republic of Korea
| | - Minhee Jang
- Department of Cancer Preventive Material Development, College of Korean Medicine, Kyung Hee University, Seoul, 130-701, Republic of Korea
- Department of Convergence Medical Sciences, College of Korean Medicine, Kyung Hee University, Seoul, 130-701, Republic of Korea
| | - Jonghee Choi
- Department of Convergence Medical Sciences, College of Korean Medicine, Kyung Hee University, Seoul, 130-701, Republic of Korea
- Brain Korea 21 Plus Program, Kyung Hee University, Seoul, 130-701, Republic of Korea
| | - Byung Soo Chang
- Department of Cosmetology, Hanseo University, Seosan, 356-706, Republic of Korea
| | - Do Young Kim
- Barrow Neurological Institute and St. Joseph's Medical Center, Phoenix, AZ, 85013, USA
| | - Sung-Hoon Kim
- Department of Cancer Preventive Material Development, College of Korean Medicine, Kyung Hee University, Seoul, 130-701, Republic of Korea
| | - Yi-Seong Kwak
- Central Research Institute, Korea Ginseng Corporation, Daejeon, 305-805, Republic of Korea
| | - Seikwan Oh
- Department of Neuroscience and Tissue Injury Defense Research Center, School of Medicine, Ewha Womans University, Seoul, 158-710, Republic of Korea
| | - Jong-Hwan Lee
- Department of Veterinary Anatomy, College of Veterinary Medicine, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Byung-Joon Chang
- Department of Veterinary Anatomy, College of Veterinary Medicine, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Ik-Hyun Cho
- Department of Convergence Medical Sciences, College of Korean Medicine, Kyung Hee University, Seoul, 130-701, Republic of Korea.
- Institute of Korean Medicine, Kyung Hee University, Seoul, 130-701, Republic of Korea.
- Brain Korea 21 Plus Program, Kyung Hee University, Seoul, 130-701, Republic of Korea.
| |
Collapse
|
41
|
Alfonso Romero-Sandoval E, Sweitzer S. Nonneuronal central mechanisms of pain: glia and immune response. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 131:325-58. [PMID: 25744678 DOI: 10.1016/bs.pmbts.2014.11.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The role of central glial cells in the mechanisms underlying pain has been intensively studied in the last two decades. Most studies on glia and pain focused on the potential detrimental role of glial cells following noxious stimulus/insults manifested as an "activation" or a "reactive" state (increase in glial marker expression and production of proinflammatory/nociceptive molecules). Therefore, "activated" or "reactive" glial cells became a target for the future generation of drugs to treat chronic pain. Several glial modulators that reduce the activation of glial cells have shown great efficacy in multiple animal (rodents mostly) models of pain (acute, subacute, chronic, inflammatory, neuropathic, surgical, etc.). These encouraging findings inspired clinical trials that have been completed in the last 5 years. Unfortunately, all clinical trials with these glial modulators have failed to demonstrate efficacy for the treatment of pain. New lines of investigation and elegant experimental designs are shedding light on alternative glial functions, which demonstrate that "glial reactivity" is not necessarily deleterious in some pathological conditions. New strategies to validate findings through our current animal models are necessary to enhance the translational value of our preclinical studies. Also, more studies using human subjects would enhance our understanding of glial cells in the context of pain. This chapter explores the available literature to objectively ponder the potential role of glial cells in human pain conditions.
Collapse
Affiliation(s)
- E Alfonso Romero-Sandoval
- Department of Pharmaceutical and Administrative Sciences, Presbyterian College School of Pharmacy, Clinton, South Carolina, USA.
| | - Sarah Sweitzer
- Department of Pharmaceutical and Administrative Sciences, Presbyterian College School of Pharmacy, Clinton, South Carolina, USA
| |
Collapse
|
42
|
Identifying strains that contribute to complex diseases through the study of microbial inheritance. Proc Natl Acad Sci U S A 2015; 112:633-40. [PMID: 25576328 DOI: 10.1073/pnas.1418781112] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
It has been 35 y since Carl Woese reported in PNAS how sequencing ribosomal RNA genes could be used to distinguish the three domains of life on Earth. During the past decade, 16S rDNA sequencing has enabled the now frequent enumeration of bacterial communities that populate the bodies of humans representing different ages, cultural traditions, and health states. A challenge going forward is to quantify the contributions of community members to wellness, disease risk, and disease pathogenesis. Here, we explore a theoretical framework for studies of the inheritance of bacterial strains and discuss the advantages and disadvantages of various study designs for assessing the contribution of strains to complex diseases.
Collapse
|
43
|
Minocycline But Not Tigecycline Is Neuroprotective and Reduces the Neuroinflammatory Response Induced by the Superimposition of Sepsis Upon Traumatic Brain Injury*. Crit Care Med 2014; 42:e570-82. [DOI: 10.1097/ccm.0000000000000414] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
44
|
Oliveira GB, Fontes EDA, de Carvalho S, da Silva JB, Fernandes LMP, Oliveira MCSP, Prediger RD, Gomes-Leal W, Lima RR, Maia CSF. Minocycline mitigates motor impairments and cortical neuronal loss induced by focal ischemia in rats chronically exposed to ethanol during adolescence. Brain Res 2014; 1561:23-34. [PMID: 24637259 DOI: 10.1016/j.brainres.2014.03.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 01/29/2014] [Accepted: 03/07/2014] [Indexed: 12/12/2022]
Abstract
Ethanol is an important risk factor for the occurrence of cerebral ischemia contributing to poor prognosis and inefficacy of drug treatments for stroke-related symptoms. Females have a higher lifetime risk for stroke than males. Moreover, female gender has been associated with increased ethanol consumption during adolescence. In the present study, we investigated whether chronic ethanol exposure during adolescence may potentiate the motor impairments and cortical damage induced by focal ischemia in female rats. We also addressed whether these effects can be mitigated by minocycline, which has been shown to be neuroprotective against different insults in the CNS. Female rats were treated with distilled water or ethanol (6.5 g/kg/day, 22.5% w/v) for 55 days by gavage. Focal ischemia was induced by microinjections of endothelin-1 (ET-1) into the motor cortex. Animals of both groups were treated daily with minocycline (25-50 mg/kg, i.p.) or sterile saline (i.p.) for 5 days, and motor function was assessed using open field, inclined plane and rotarod tests. Chronic ethanol exposure exacerbated locomotor activity and motor coordination impairments induced by focal ischemia in rats. Moreover, histological analysis revealed that microinjections of ET-1 induced pyramidal neuron loss and microglial activation in the motor cortex. Minocycline reversed the observed motor impairments, microglial activation and pyramidal neuron loss in the motor cortex of ischemic rats even in those exposed to ethanol. These results suggest that minocycline induces neuroprotection and functional recovery in ischemic female rats intoxicated with ethanol during adolescence. Furthermore, the mechanism underlying this protective effect may be related to the modulation of neuroinflammation.
Collapse
Affiliation(s)
- Gedeão Batista Oliveira
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Instituto de Ciências da Saúde, Universidade Federal do Pará, 66075-900 Belém, Pará, Brazil
| | - Enéas de Andrade Fontes
- Programa de Pós-graduação em Neurociências e Biologia Celular, Faculdade de Farmácia, Instituto de Ciências da Saúde, Universidade Federal do Pará, 66075-900 Belém, Pará, Brazil
| | - Sabrina de Carvalho
- Laboratório de Farmacologia da Inflamação e do Comportamento, Faculdade de Farmácia, Instituto de Ciências da Saúde, Universidade Federal do Pará, 66075-900 Belém, Pará, Brazil
| | - Josiane Batista da Silva
- Laboratório de Farmacologia da Inflamação e do Comportamento, Faculdade de Farmácia, Instituto de Ciências da Saúde, Universidade Federal do Pará, 66075-900 Belém, Pará, Brazil
| | - Luanna Melo Pereira Fernandes
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Instituto de Ciências da Saúde, Universidade Federal do Pará, 66075-900 Belém, Pará, Brazil; Laboratório de Farmacologia da Inflamação e do Comportamento, Faculdade de Farmácia, Instituto de Ciências da Saúde, Universidade Federal do Pará, 66075-900 Belém, Pará, Brazil
| | - Maria Cristina Souza Pereira Oliveira
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Instituto de Ciências da Saúde, Universidade Federal do Pará, 66075-900 Belém, Pará, Brazil
| | - Rui Daniel Prediger
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, 88049-900 Florianópolis, SC, Brazil
| | - Walace Gomes-Leal
- Laboratório de Neuroproteção e Neurorregeneração Experimental do Instituto de Ciências Biológicas, Universidade Federal do Pará, 66075-900 Belém, Pará, Brazil
| | - Rafael Rodrigues Lima
- Laboratório de Neuroproteção e Neurorregeneração Experimental do Instituto de Ciências Biológicas, Universidade Federal do Pará, 66075-900 Belém, Pará, Brazil
| | - Cristiane Socorro Ferraz Maia
- Programa de Pós-Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Instituto de Ciências da Saúde, Universidade Federal do Pará, 66075-900 Belém, Pará, Brazil; Programa de Pós-graduação em Neurociências e Biologia Celular, Faculdade de Farmácia, Instituto de Ciências da Saúde, Universidade Federal do Pará, 66075-900 Belém, Pará, Brazil; Laboratório de Farmacologia da Inflamação e do Comportamento, Faculdade de Farmácia, Instituto de Ciências da Saúde, Universidade Federal do Pará, 66075-900 Belém, Pará, Brazil.
| |
Collapse
|
45
|
Hebbring SJ. The challenges, advantages and future of phenome-wide association studies. Immunology 2014; 141:157-65. [PMID: 24147732 PMCID: PMC3904236 DOI: 10.1111/imm.12195] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 10/16/2013] [Accepted: 10/17/2013] [Indexed: 12/11/2022] Open
Abstract
Over the last decade, significant technological breakthroughs have revolutionized human genomic research in the form of genome-wide association studies (GWASs). GWASs have identified thousands of statistically significant genetic variants associated with hundreds of human conditions including many with immunological aetiologies (e.g. multiple sclerosis, ankylosing spondylitis and rheumatoid arthritis). Unfortunately, most GWASs fail to identify clinically significant associations. Identifying biologically significant variants by GWAS also presents a challenge. The GWAS is a phenotype-to-genotype approach. As a complementary/alternative approach to the GWAS, investigators have begun to exploit extensive electronic medical record systems to conduct a genotype-to-phenotype approach when studying human disease – specifically, the phenome-wide association study (PheWAS). Although the PheWAS approach is in its infancy, this method has already demonstrated its capacity to rediscover important genetic associations related to immunological diseases/conditions. Furthermore, PheWAS has the advantage of identifying genetic variants with pleiotropic properties. This is particularly relevant for HLA variants. For example, PheWAS results have demonstrated that the HLA-DRB1 variant associated with multiple sclerosis may also be associated with erythematous conditions including rosacea. Likewise, PheWAS has demonstrated that the HLA-B genotype is not only associated with spondylopathies, uveitis, and variability in platelet count, but may also play an important role in other conditions, such as mastoiditis. This review will discuss and compare general PheWAS methodologies, describe both the challenges and advantages of the PheWAS, and provide insight into the potential directions in which PheWAS may lead.
Collapse
Affiliation(s)
- Scott J Hebbring
- Center for Human Genetics, Marshfield Clinic Research Foundation, Marshfield, WI, USA
| |
Collapse
|
46
|
Viscomi MT, Molinari M. Remote neurodegeneration: multiple actors for one play. Mol Neurobiol 2014; 50:368-89. [PMID: 24442481 DOI: 10.1007/s12035-013-8629-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 12/24/2013] [Indexed: 12/19/2022]
Abstract
Remote neurodegeneration significantly influences the clinical outcome in many central nervous system (CNS) pathologies, such as stroke, multiple sclerosis, and traumatic brain and spinal cord injuries. Because these processes develop days or months after injury, they are accompanied by a therapeutic window of opportunity. The complexity and clinical significance of remote damage is prompting many groups to examine the factors of remote degeneration. This research is providing insights into key unanswered questions, opening new avenues for innovative neuroprotective therapies. In this review, we evaluate data from various remote degeneration models to describe the complexity of the systems that are involved and the importance of their interactions in reducing damage and promoting recovery after brain lesions. Specifically, we recapitulate the current data on remote neuronal degeneration, focusing on molecular and cellular events, as studied in stroke and brain and spinal cord injury models. Remote damage is a multifactorial phenomenon in which many components become active in specific time frames. Days, weeks, or months after injury onset, the interplay between key effectors differentially affects neuronal survival and functional outcomes. In particular, we discuss apoptosis, inflammation, oxidative damage, and autophagy-all of which mediate remote degeneration at specific times. We also review current findings on the pharmacological manipulation of remote degeneration mechanisms in reducing damage and sustaining outcomes. These novel treatments differ from those that have been proposed to limit primary lesion site damage, representing new perspectives on neuroprotection.
Collapse
Affiliation(s)
- Maria Teresa Viscomi
- Experimental Neurorehabilitation Laboratory, Santa Lucia Foundation I.R.C.C.S., Via del Fosso di Fiorano 65, 00143, Rome, Italy,
| | | |
Collapse
|
47
|
Beardsley PM, Hauser KF. Glial modulators as potential treatments of psychostimulant abuse. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2014; 69:1-69. [PMID: 24484974 DOI: 10.1016/b978-0-12-420118-7.00001-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Glia (including astrocytes, microglia, and oligodendrocytes), which constitute the majority of cells in the brain, have many of the same receptors as neurons, secrete neurotransmitters and neurotrophic and neuroinflammatory factors, control clearance of neurotransmitters from synaptic clefts, and are intimately involved in synaptic plasticity. Despite their prevalence and spectrum of functions, appreciation of their potential general importance has been elusive since their identification in the mid-1800s, and only relatively recently have they been gaining their due respect. This development of appreciation has been nurtured by the growing awareness that drugs of abuse, including the psychostimulants, affect glial activity, and glial activity, in turn, has been found to modulate the effects of the psychostimulants. This developing awareness has begun to illuminate novel pharmacotherapeutic targets for treating psychostimulant abuse, for which targeting more conventional neuronal targets has not yet resulted in a single, approved medication. In this chapter, we discuss the molecular pharmacology, physiology, and functional relationships that the glia have especially in the light in which they present themselves as targets for pharmacotherapeutics intended to treat psychostimulant abuse disorders. We then review a cross section of preclinical studies that have manipulated glial processes whose behavioral effects have been supportive of considering the glia as drug targets for psychostimulant-abuse medications. We then close with comments regarding the current clinical evaluation of relevant compounds for treating psychostimulant abuse, as well as the likelihood of future prospects.
Collapse
Affiliation(s)
| | - Kurt F Hauser
- Virginia Commonwealth University, Richmond, Virginia, USA
| |
Collapse
|
48
|
Tronel C, Page G, Bodard S, Chalon S, Antier D. The specific PKR inhibitor C16 prevents apoptosis and IL-1β production in an acute excitotoxic rat model with a neuroinflammatory component. Neurochem Int 2013; 64:73-83. [PMID: 24211709 DOI: 10.1016/j.neuint.2013.10.012] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 10/10/2013] [Accepted: 10/22/2013] [Indexed: 12/20/2022]
Abstract
The double-stranded RNA-dependent protein kinase (PKR), an apoptotic inducer, regulates much pro-inflammatory cytokine production. The purpose of this study was to evaluate in vivo the effects of the specific PKR inhibitor C16 in the striatum in an acute excitotoxic rat model with an important neuroinflammatory component. Inflammation was induced by unilateral striatal injection of quinolinic acid (QA) in 10-week-old normotensive rats. Animals were separated into groups receiving either vehicle or C16 for both sham and QA rats. The effects were assessed in ipsi- and contralateral striata by immunoblotting for PKR activation, by Luminex assay for cytokine levels and by immunofluorescent staining for cleaved caspase-3 to detect neuronal apoptosis. The highest dose of C16 (600μg/kg; C16-2) in QA rats reduced expression of the active catalytic domain of the PKR vs. that in vehicle-injected QA rats. A robust increase of IL-1β levels on the contralateral side of QA rats was prevented by C16-2 (97% inhibition). Macroscopic and microscopic observation of cerebral tissue (Hematoxylin & Eosin staining) revealed that tissue integrity was more preserved with C16-2 treatment than its vehicle in QA rats. Furthermore, C16-2 treatment decreased by 47% the neuronal loss and by 37% the number of positive cleaved caspase-3 neurons induced by QA injection. In conclusion, C16 prevented not only the PKR-induced neuronal loss but also the inflammatory response in this acute excitotoxic in vivo model, highlighting its promising neuroprotective properties to rescue acute brain lesions.
Collapse
Affiliation(s)
- C Tronel
- INSERM U930, Tours, France; Université François Rabelais de Tours, UMR U930, Tours, France.
| | - G Page
- EA 3808, University of Poitiers, "Molecular Targets and Therapeutics of Alzheimer's Disease (CiMoTheMA)", 6 rue de la Milétrie, BP 199, 86034 Poitiers, France
| | - S Bodard
- INSERM U930, Tours, France; Université François Rabelais de Tours, UMR U930, Tours, France
| | - S Chalon
- INSERM U930, Tours, France; Université François Rabelais de Tours, UMR U930, Tours, France
| | - D Antier
- INSERM U930, Tours, France; Université François Rabelais de Tours, UMR U930, Tours, France
| |
Collapse
|
49
|
Abstract
Although there has been unequivocal progress in the development of treatments for multiple sclerosis over the last 20 years, currently licensed treatments have demonstrated convincing effects on disease course only with reference to relapse frequency. This review summarises the progress made, highlights the indications for, and limitations of, current disease-modifying therapies and discusses some interventions currently in development.
Collapse
|
50
|
Abstract
Multiple sclerosis (MS) is a demyelinating disease of the central nervous system that is pathologically characterized by inflammatory demyelination and neurodegeneration. Axonal damage, along with neuronal loss, occurs from disease onset and may lead to progressive and permanent disability. In contrast with the inflammatory pathways, the molecular mechanisms leading to MS neurodegeneration remain largely elusive. With improved understanding of these mechanisms, new potential therapeutic targets for neuroprotection have emerged. We review the current understanding of neurodegenerative processes at play in MS and discuss potential outcome measures and targets for neuroprotection trials.
Collapse
Affiliation(s)
- Amir-Hadi Maghzi
- Multiple Sclerosis Center, Department of Neurology, University of California San Francisco (UCSF), 675 Nelson Rising Lane, 2nd floor, Room 221F, Box 3206, 94158, San Francisco, CA, USA,
| | | | | |
Collapse
|